1
|
Kellingray L, Savva GM, Garcia-Gutierrez E, Snell J, Romano S, Yara DA, Altera A, de Oliveira Martins L, Hutchins C, Baker D, Hayhoe A, Hacon C, Elumogo N, Narbad A, Sayavedra L. Temporal dynamics of SARS-CoV-2 shedding in feces and saliva: a longitudinal study in Norfolk, United Kingdom during the 2021-2022 COVID-19 waves. Microbiol Spectr 2025:e0319524. [PMID: 40131871 DOI: 10.1128/spectrum.03195-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Accepted: 02/24/2025] [Indexed: 03/27/2025] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) was originally described as a respiratory illness; however, it is now known that the infection can spread to the gastrointestinal tract, leading to shedding in feces potentially being a source of infection through wastewater. We aimed to assess the prevalence and persistence of SARS-CoV-2 in fecal and saliva samples for up to 7 weeks post-detection in a cohort of 98 participants from Norfolk, United Kingdom using RT-qPCR. Secondary goals included sequencing the viral isolates present in fecal samples and comparing the genetic sequence with isolates in the saliva of the same participant. Furthermore, we sought to identify factors associated with the presence of detectable virus in feces or saliva after a positive SARS-CoV-2 test. Saliva remained SARS-CoV-2-positive for longer periods compared to fecal samples, with all positive fecal samples occurring within 4 weeks of the initial positive test. Detectable virus in fecal samples was positively associated with the number of symptoms experienced by the individuals. Based on the genome sequencing and taxonomic classification of the virus, one donor had a distinct strain in feces compared to saliva on the same collection date, which suggests that different isolates could dominate different tissues. Our results underscore the importance of considering multiple biological samples, such as feces, in the detection and characterization of SARS-CoV-2, particularly in clinical procedures involving patient fecal material transplant. Such insights could contribute to enhancing the safety protocols surrounding the handling of patient samples and aid in devising effective strategies for mitigating the spread of coronavirus disease. IMPORTANCE This study provides critical insights into the dynamics of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) shedding in fecal and saliva samples, demonstrating that while viral RNA is detectable shortly after diagnosis, its prevalence declines rapidly over the course of infection. Detection was more common among individuals with more concurrent symptoms, emphasizing the potential influence of symptom burden on viral persistence. By analyzing a United Kingdom-based cohort, this study fills a significant gap in the literature, which has largely focused on Asian and North American populations, offering a geographically unique perspective on viral shedding dynamics. Our findings contribute to a globally relevant understanding of SARS-CoV-2 shedding by revealing differences in shedding durations compared to studies from other regions. These differences highlight the need for geographically diverse research to account for variations in genetic background, immune response, and healthcare practices.
Collapse
Affiliation(s)
- Lee Kellingray
- Quadram Institute Bioscience, Norwich Research Park, Norwich, United Kingdom
| | - George M Savva
- Quadram Institute Bioscience, Norwich Research Park, Norwich, United Kingdom
| | - Enriqueta Garcia-Gutierrez
- Quadram Institute Bioscience, Norwich Research Park, Norwich, United Kingdom
- Department of Agronomic Engineering-ETSIA, Universidad Politécnica de Cartagena, Paseo Alfonso XIII, Cartagena, Region of Murcia, Spain
| | - Jemma Snell
- Quadram Institute Bioscience, Norwich Research Park, Norwich, United Kingdom
| | - Stefano Romano
- Quadram Institute Bioscience, Norwich Research Park, Norwich, United Kingdom
| | | | - Annalisa Altera
- Quadram Institute Bioscience, Norwich Research Park, Norwich, United Kingdom
| | | | - Chloe Hutchins
- Quadram Institute Bioscience, Norwich Research Park, Norwich, United Kingdom
| | - David Baker
- Quadram Institute Bioscience, Norwich Research Park, Norwich, United Kingdom
| | - Antonietta Hayhoe
- Quadram Institute Bioscience, Norwich Research Park, Norwich, United Kingdom
| | - Christian Hacon
- James Paget University Hospitals NHS Foundation Trust, Great Yarmouth, England, United Kingdom
| | - Ngozi Elumogo
- Norfolk and Norwich University Hospitals NHS Foundation Trust, Norwich, United Kingdom
| | - Arjan Narbad
- Quadram Institute Bioscience, Norwich Research Park, Norwich, United Kingdom
| | - Lizbeth Sayavedra
- Quadram Institute Bioscience, Norwich Research Park, Norwich, United Kingdom
| |
Collapse
|
2
|
Hdoufane I, Oubahmane M, Habibi Y, Delaite C, Alanazi MM, Cherqaoui D. Identification of potent TMPRSS4 inhibitors through structural modeling and molecular dynamics simulations. Sci Rep 2025; 15:2748. [PMID: 39838126 PMCID: PMC11750979 DOI: 10.1038/s41598-025-86961-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Accepted: 01/15/2025] [Indexed: 01/23/2025] Open
Abstract
TMPRSS4, a transmembrane serine protease type II, is associated with various pathological illnesses. It has been found to activate SARS-CoV-2, enhance viral infection of human small-intestinal enterocytes and is overexpressed in different types of cancers. Therefore, this study aims to disover potential TMPRSS4 inhibitors that have better binding affinity than the approved inhibitors: 2-hydroxydiarylamide and tyroserleutide. Since no 3D-structure is known for TMPRSS4, structural models for the TMPRSS4 serine protease domain were developed. The modeled structures were validated and subjected to molecular dynamics simulations. FDA-approved, clinical/preclinical drugs and natural products were docked to the pocket of TMPRSS4. Moreover, through a systematic analysis, MD simulations and MM-GBSA binding free energy calculations revealed that the best candidates Ergotamine, S55746, NPC478048, Lifirafenib, and NPC77101 are highly stable drug candidates in complex with TMPRSS4, displaying low RMSD and RMSF values with strong binding stability. Among these compounds, Ergotamine showed the most favorable binding energy (-33.73 kcal/mol). Overall, our in silico results revealed that these compounds could act as potent TMPRSS4 inhibitors and need to be validated by future experimental studies.
Collapse
Affiliation(s)
- Ismail Hdoufane
- Laboratory of Molecular Chemistry, Department of Chemistry, Faculty of Sciences Semlalia, Cadi Ayyad University, BP 2390, 40000, Marrakech, Morocco.
| | - Mehdi Oubahmane
- Laboratory of Molecular Chemistry, Department of Chemistry, Faculty of Sciences Semlalia, Cadi Ayyad University, BP 2390, 40000, Marrakech, Morocco
| | - Youssef Habibi
- Sustainable Materials Research Center (SUSMAT-RC), University Mohamed VI Polytechnic (UM6P), Hay Moulay Rachid, Benguerir, Morocco
| | - Christelle Delaite
- Laboratoire de Photochimie et d'Ingénierie Macromoléculaires (LPIM EA 4567), Université de Haute-Alsace, 68100, Mulhouse, France
| | - Mohammed M Alanazi
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh, 11451, Saudi Arabia
| | - Driss Cherqaoui
- Laboratory of Molecular Chemistry, Department of Chemistry, Faculty of Sciences Semlalia, Cadi Ayyad University, BP 2390, 40000, Marrakech, Morocco
- Sustainable Materials Research Center (SUSMAT-RC), University Mohamed VI Polytechnic (UM6P), Hay Moulay Rachid, Benguerir, Morocco
| |
Collapse
|
3
|
Pan J, Li J. Gastroesophageal reflux disease increases predisposition to severe COVID-19: Insights from integrated Mendelian randomization and genetic analysis. Ann Hum Genet 2025; 89:54-65. [PMID: 39530352 DOI: 10.1111/ahg.12584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 10/24/2024] [Accepted: 11/01/2024] [Indexed: 11/16/2024]
Abstract
OBJECTIVE This study aims to investigate the potential causal relationship, shared genomic loci, as well as potential molecular pathways and tissue-specific expression patterns between gastroesophageal reflux disease (GERD) and the risk of hospitalized/severe 2019 coronavirus disease (COVID-19). METHODS We employed linkage disequilibrium score regression and bidirectional Mendelian randomization (MR) analysis to explore potential genetic associations between GERD (N = 602,604) and hospitalized COVID-19 (N = 2095,324) as well as severe COVID-19 (N = 1086,211). Additionally, shared genomic loci were extracted from common pivotal regions, further confirmed through corresponding colocalization analyses. GERD-driven molecular pathway network was constructed using extensive literature data mining to understand the molecular-level impacts of GERD on COVID-19. RESULTS Our results revealed a significant positive genetic correlation between GERD and both hospitalized (rg = 0.418) and severe COVID-19 (rg = 0.314). Furthermore, the MR analysis demonstrated a unidirectional causal effect of genetic predisposition to GERD on COVID-19 outcomes, including hospitalized COVID-19 (odds ratio [OR]: 1.33, 95% confidence interval [CI]: 1.27-1.44, p = 9.17e - 12) and severe COVID-19 (OR: 1.27, 95% CI: 1.18-1.37, p = 1.20e - 05). Additionally, GERD and both COVID-19 conditions shared one genomic locus with lead-SNPs rs1011407 and rs1123573, corresponding to the transcription factor BCL11A. Colocalization analysis further demonstrated a significant positive correlation between genome-wide association study and expression quantitative trait locus (eQTL) abnormalities, including rs1011407 (eQTL_p = 2.35e - 07) and rs1123573 (eQTL_p = 2.74e - 05). Molecular pathway analysis indicated that GERD might promote the progression of COVID-19 by inducting immune-activated and inflammation-related pathways. CONCLUSION These findings confirm that genetically determined GERD may increase the susceptibility to hospitalized/severe COVID-19. The shared genetic loci and the potential molecular pathways offer valuable insights into causal connections between GERD and COVID-19.
Collapse
Affiliation(s)
- Jingjing Pan
- Department of Microbiology, Zhejiang Provincial Centers for Disease Control and Prevention, Hangzhou, China
- Zhejiang Key Laboratory of Public Health Detection and Pathogenesis Research, Hangzhou, China
| | - Jianhua Li
- Department of Microbiology, Zhejiang Provincial Centers for Disease Control and Prevention, Hangzhou, China
- Zhejiang Key Laboratory of Public Health Detection and Pathogenesis Research, Hangzhou, China
| |
Collapse
|
4
|
Umakanthan S, Katwaroo AR, Bukelo M, Bg S, Boralingaiah P, Ranade AV, Rangan P, Shashidhar S, Kini JR, Kini G. Post-Acute Sequelae of Covid-19: A System-wise Approach on the Effects of Long-Covid-19. AMERICAN JOURNAL OF MEDICINE OPEN 2024; 12:100071. [PMID: 39268246 PMCID: PMC11387218 DOI: 10.1016/j.ajmo.2024.100071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 06/01/2024] [Indexed: 09/15/2024]
Abstract
The SARS-CoV-2 virus responsible for the COVID-19 pandemic has profoundly impacted global health, economics, and society. This review seeks to encompass an overview of current knowledge on COVID-19, including its transmission, pathogenesis, and clinical presentation related to various systems within the human body. COVID-19 is a highly contagious illness that has rapidly spread worldwide. As of August 4, 2023, the WHO reported over 570 million confirmed cases of COVID-19 and over 6.3 million deaths. Although the virus is most common in adults, children can also be infected. Respiratory droplets that are produced when an infected person coughs or sneezes are the primary transmission mode for COVID-19. Additionally, the virus can be disseminated via contact with contaminated surfaces or objects, as it can remain viable for several hours or days. SARS-CoV-2 is a respiratory virus that enters cells by bonding with the angiotensin-converting enzyme 2 (ACE2) receptor. Once inside the cell, the virus replicates and produces new particles that can infect other cells. Interestingly, the effects of post-acute sequelae of SARS-CoV-2 infection (PASC) encompass more than just respiratory system. The findings presented in the data suggest that PASC significantly impacts multiple organs and their respective physiological processes. In light of these observations, we aim to provide a detailed discussion of the relevant findings in this paper. Through our review, we hope to provide healthcare professionals with a deeper understanding of the effects of PASC on the human body, which could ultimately lead to improved patient outcomes and treatment strategies.
Collapse
Affiliation(s)
- Srikanth Umakanthan
- Department of Paraclinical Sciences, Faculty of Medical Sciences, The University of the West Indies, St. Augustine, Trinidad and Tobago
| | - Arun Rabindra Katwaroo
- Trinidad Institute of Medical Technology, Department of Medicine, St. Augustine, Trinidad and Tobago
| | - Maryann Bukelo
- Department of Anatomical Pathology, Laboratory Services, North Central Regional Health Authority, Champ Fleurs, Trinidad and Tobago
| | - Shashidhar Bg
- Department of Critical care Medicine, Manipal Hospital, Bengaluru, India
| | - Prashanth Boralingaiah
- Early Psychosis Prevention and Intervention Center (EPPIC), Orygen Youth Health, Sunshine, Australia
| | - Anu V Ranade
- Department of Basic Medical Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
| | | | | | - Jyoti Ramanath Kini
- Department of Pathology, Kasturba Medical College, Mangalore, Manipal Academy of Higher Education, Karnataka, India
| | - Gayathri Kini
- Department of Pathology, Kasturba Medical College, Mangalore, Manipal Academy of Higher Education, Karnataka, India
| |
Collapse
|
5
|
Tsai TY, Wu JF, Weng MT, Chuang CH, Huang TY, Tai WC, Tai CM, Chung CS, Chen CC, Lin CP, Tsai YY, Wei SC. Exacerbated gastrointestinal symptoms and long COVID in IBD patients with SARS-CoV-2 infection: A multi-center study from taiwan. J Formos Med Assoc 2024; 123:866-874. [PMID: 38553294 DOI: 10.1016/j.jfma.2024.03.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 03/09/2024] [Accepted: 03/18/2024] [Indexed: 08/10/2024] Open
Abstract
BACKGROUND/PURPOSE Limited studies have addressed the exacerbation of symptoms and long COVID in inflammatory bowel disease (IBD) patients following non-severe COVID-19 infection, particularly with post-COVID-19 vaccination. We aim to investigate factors associated with exacerbated gastrointestinal symptoms (EGS) and long COVID in IBD patients with non-severe COVID-19, which is most common situation in daily practice. METHODS This is an observational study by multiple centers in Taiwan from May 2020 to March 2023. We collected clinical manifestation, data, and medication information from IBD patients with non-severe COVID-19. EGS was defined as increased frequency of diarrhea, bloody stool, and abdomen pain within 14 days after SARS-COV-2 infection. Long COVID was defined following the guidelines of the World Health Organization. RESULTS Out of 90 patients, most of them (88.9%) received at least standard two doses of COVID-19 vaccination and the majority (87.8%) were mild diseases of COVID-19.30% of patients experienced EGS during COVID-19 with higher ESR levels serving as a predictive factor (Odds ratio: 3.6, 95% confidence interval: 1.2-10.5, P = 0.02). 38.1% of those patients developed long COVID. The patients who experienced EGS during COVID-19 and with a history of longer IBD duration showed a significant association with long COVID (p = 0.03 and p = 0.02). CONCLUSION Our study revealed that EGS and long COVID occurred in one third of IBD patients with non-severe COVID-19, even though most of them had received the standard plus booster vaccination. We identified associated factors for EGS and long COVID, emphasizing the importance of post-COVID-19 follow-up in IBD patients.
Collapse
Affiliation(s)
- Tsung-Yu Tsai
- Center for Digestive Medicine, Department of Internal Medicine, China Medical University Hospital, Taichung, Taiwan; School of Medicine, China Medical University, Taichung, Taiwan; Center for Translational Genomics & Regenerative Medicine Research, China Medical University Hospital, Taichung, Taiwan
| | - Jia-Feng Wu
- Department of Pediatrics, National Taiwan University Hospital, Taipei, Taiwan
| | - Meng-Tzu Weng
- Department of Medical Research, National Taiwan University Hospital, Hsin-Chu Branch, Hsinchu, Taiwan; Division of Gastroenterology, National Taiwan University Hospital, Taipei, Taiwan
| | - Chiao-Hsiung Chuang
- Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Tien-Yu Huang
- Division of Gastroenterology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Wei-Chen Tai
- Division of Gastroenterology, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| | - Chi-Ming Tai
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, E-Da Hospital, I-Shou University, Kaohsiung, Taiwan; School of Medicine for International Students, College of Medicine, I-Shou University, Kaohsiung, Taiwan
| | - Chen-Shuan Chung
- Division for Gastroenterology and Hepatology, Far Eastern Memorial Hospital, New Taipei City, Taiwan
| | - Chih-Cheng Chen
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, E-Da Hospital, I-Shou University, Kaohsiung, Taiwan; Division of Gastroenterology and Hepatology, Internal Medicine, E-Da Cancer Hospital, I-Shou University, Kaohsiung, Taiwan; The School of Chinese Medicine for Post Baccalaureate, I-Shou University, Kaohsiung, Taiwan
| | - Ching-Pin Lin
- Division of Gastroenterology, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Yuan-Yao Tsai
- Department of Colorectal Surgery, China Medical University Hospital, Taichung, Taiwan
| | - Shu-Chen Wei
- Division of Gastroenterology, National Taiwan University Hospital, Taipei, Taiwan.
| |
Collapse
|
6
|
Resendez S, Brown SH, Ruiz Ayala HS, Rangan P, Nebeker J, Montella D, Elkin PL. Defining the Subtypes of Long COVID and Risk Factors for Prolonged Disease: Population-Based Case-Crossover Study. JMIR Public Health Surveill 2024; 10:e49841. [PMID: 38687984 PMCID: PMC11094603 DOI: 10.2196/49841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Revised: 01/19/2024] [Accepted: 02/15/2024] [Indexed: 05/02/2024] Open
Abstract
BACKGROUND There have been over 772 million confirmed cases of COVID-19 worldwide. A significant portion of these infections will lead to long COVID (post-COVID-19 condition) and its attendant morbidities and costs. Numerous life-altering complications have already been associated with the development of long COVID, including chronic fatigue, brain fog, and dangerous heart rhythms. OBJECTIVE We aim to derive an actionable long COVID case definition consisting of significantly increased signs, symptoms, and diagnoses to support pandemic-related clinical, public health, research, and policy initiatives. METHODS This research employs a case-crossover population-based study using International Classification of Diseases, 10th Revision, Clinical Modification (ICD-10-CM) data generated at Veterans Affairs medical centers nationwide between January 1, 2020, and August 18, 2022. In total, 367,148 individuals with ICD-10-CM data both before and after a positive COVID-19 test were selected for analysis. We compared ICD-10-CM codes assigned 1 to 7 months following each patient's positive test with those assigned up to 6 months prior. Further, 350,315 patients had novel codes assigned during this window of time. We defined signs, symptoms, and diagnoses as being associated with long COVID if they had a novel case frequency of ≥1:1000, and they significantly increased in our entire cohort after a positive test. We present odds ratios with CIs for long COVID signs, symptoms, and diagnoses, organized by ICD-10-CM functional groups and medical specialty. We used our definition to assess long COVID risk based on a patient's demographics, Elixhauser score, vaccination status, and COVID-19 disease severity. RESULTS We developed a long COVID definition consisting of 323 ICD-10-CM diagnosis codes grouped into 143 ICD-10-CM functional groups that were significantly increased in our 367,148 patient post-COVID-19 population. We defined 17 medical-specialty long COVID subtypes such as cardiology long COVID. Patients who were COVID-19-positive developed signs, symptoms, or diagnoses included in our long COVID definition at a proportion of at least 59.7% (268,320/449,450, based on a denominator of all patients who were COVID-19-positive). The long COVID cohort was 8 years older with more comorbidities (2-year Elixhauser score 7.97 in the patients with long COVID vs 4.21 in the patients with non-long COVID). Patients who had a more severe bout of COVID-19, as judged by their minimum oxygen saturation level, were also more likely to develop long COVID. CONCLUSIONS An actionable, data-driven definition of long COVID can help clinicians screen for and diagnose long COVID, allowing identified patients to be admitted into appropriate monitoring and treatment programs. This long COVID definition can also support public health, research, and policy initiatives. Patients with COVID-19 who are older or have low oxygen saturation levels during their bout of COVID-19, or those who have multiple comorbidities should be preferentially watched for the development of long COVID.
Collapse
Affiliation(s)
- Skyler Resendez
- Department of Biomedical Informatics, University at Buffalo, State University of New York, Buffalo, NY, United States
| | - Steven H Brown
- Office of Health Informatics, Department of Veterans Affairs, Washington, DC, United States
| | - Hugo Sebastian Ruiz Ayala
- Department of Biomedical Informatics, University at Buffalo, State University of New York, Buffalo, NY, United States
| | - Prahalad Rangan
- Department of Biomedical Informatics, University at Buffalo, State University of New York, Buffalo, NY, United States
| | - Jonathan Nebeker
- Office of Health Informatics, Department of Veterans Affairs, Washington, DC, United States
| | - Diane Montella
- Office of Health Informatics, Department of Veterans Affairs, Washington, DC, United States
| | - Peter L Elkin
- Department of Biomedical Informatics, University at Buffalo, State University of New York, Buffalo, NY, United States
- Office of Health Informatics, Department of Veterans Affairs, Washington, DC, United States
| |
Collapse
|
7
|
Zeng Z, Tang W. Gut microbiota: A potential player in psychiatric symptoms during COVID-19. World J Biol Psychiatry 2024; 25:267-280. [PMID: 38607962 DOI: 10.1080/15622975.2024.2342846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 04/04/2024] [Indexed: 04/14/2024]
Abstract
OBJECTIVES This study aims to explore the potential interconnections among gut microbiota, COVID-19 infection, depression and anxiety disorder. Additionally, it tries to assess potential therapeutic interventions that may improve the dysbiosis of gut microbiota. METHODS To achieve these objectives, we reviewed existing literature, encompassing studies and critical reviews that intersect the domains of gut microbiota, COVID-19, depression and anxiety disorders. RESULTS The findings highlight a notable correlation between the dysbiosis of gut microbiota and psychiatric symptoms in the context of COVID-19. Specifically, there is a marked reduction in the populations of bacteria that generate anti-inflammatory short-chain fatty acids (SCFAs), alongside a rise in the prevalence of gut bacterial clusters linked to inflammatory processes. Furthermore, several potential treatment strategies were summarised for improving the dysbiosis. CONCLUSIONS Gut microbiota plays a significant role in psychiatric symptoms during COVID-19, which has significant implications for the study and prevention of psychiatric symptoms in major epidemic diseases.
Collapse
Affiliation(s)
- Zijie Zeng
- Department of Psychology, School of Public Health, Southern Medical University, Guangzhou, China
| | | |
Collapse
|
8
|
Yang R, Guan X, Niu Z, Zhang R, Lv S, Xu X, Zhao Y, Wu J. Establishment of sex-specific predictive models for critical illness in Chinese people with the Omicron variant. Front Microbiol 2024; 14:1224132. [PMID: 38322760 PMCID: PMC10844546 DOI: 10.3389/fmicb.2023.1224132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 12/27/2023] [Indexed: 02/08/2024] Open
Abstract
Introduction The Omicron variant has rapidly spread throughout the world compared to the Delta variant and poses a great threat to global healthcare systems due to its immune evasion and rapid spread. Sex has been identified as a factor significantly associated with COVID-19 mortality, but it remains unclear which clinical indicators could be identified as risk factors in each sex group and which sex-specific risk factors might shape the worse clinical outcome, especially for Omicrons. This study aimed to confirm the relationship between sex and the progression of the Omicron variant and to explore its sex-biased risk factors. Methods We conducted a retrospective study including 1,132 hospitalized patients with the COVID-19 Omicron variant from 5 December 2022 to 25 January 2023 at Shanghai General Hospital, and the medical history data and clinical index data of the inpatients for possible sex differences were compared and analyzed. Then, a sex-specific Lasso regression was performed to select the variables significantly associated with critical illness, including intensive care unit admission, invasive mechanical ventilation, or death. A logistic regression was used to construct a sex-specific predictive model distinctively for the critical illness outcome using selected covariates. Results Among the collected 115 clinical indicators, up to 72 showed significant sex differences, including the difference in merit and the proportion of people with abnormalities. More importantly, males had greater critical illness (28.4% vs. 19.9%) and a significantly higher intensive care unit occupancy (20.96% vs. 14.49%) and mortality (13.2% vs. 4.9%), and males over 80 showed worse outcomes than females. Predictive models (AUC: 0.861 for males and 0.898 for females) showed 12 risk factors for males and 10 for females. Through a comprehensive sex-stratified analysis of a large cohort of hospitalized Omicron-infected patients, we identified the specific risk factors for critical illness by developing prediction models. Discussion Sex disparities and the identified risk factors should be considered, especially in the personalized prevention and treatment of the COVID-19 Omicron variant.
Collapse
Affiliation(s)
- Rui Yang
- Department of Laboratory Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Laboratory Medicine, Jiading Branch of Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xin Guan
- Department of Laboratory Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ziguang Niu
- Department of Laboratory Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Rulin Zhang
- Department of Laboratory Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Laboratory Medicine, Jiading Branch of Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Siang Lv
- Department of Laboratory Medicine, Jiading Branch of Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Pathology, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
| | - Xiang Xu
- Department of Laboratory Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Laboratory Medicine, Jiading Branch of Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yingying Zhao
- Department of Medical Affairs, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jun Wu
- Department of Laboratory Medicine, Jiading Branch of Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Pathology, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
| |
Collapse
|
9
|
Mafra D, Kemp JA, Cardozo LFMF, Borges NA, Nerbass FB, Alvarenga L, Kalantar-Zadeh K. COVID-19 and Nutrition: Focus on Chronic Kidney Disease. J Ren Nutr 2023; 33:S118-S127. [PMID: 37632513 DOI: 10.1053/j.jrn.2023.01.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 12/14/2022] [Accepted: 01/09/2023] [Indexed: 08/28/2023] Open
Abstract
Some chronic diseases, including chronic kidney disease (CKD), may be associated with poor outcomes, including a high rate of hospitalization and death after COVID-19 infection. In addition to the vaccination program, diet intervention is essential for boosting immunity and preventing complications. A healthy diet containing bioactive compounds may help mitigate inflammatory responses and oxidative stress caused by COVID-19. In this review, we discuss dietary interventions for mitigating COVID-19 complications, including in persons with CKD, which can worsen COVID-19 symptoms and its clinical outcomes, while diet may help patients with CKD to resist the ravages of COVID-19 by improving the immune system, modulating gut dysbiosis, mitigating COVID-19 complications, and reducing hospitalization and mortality. The concept of food as medicine, also known as culinary medicine, for patients with CKD can be extrapolated to COVID-19 infection because healthy foods and nutraceuticals have the potential to exert an important antiviral, anti-inflammatory, and antioxidant role.
Collapse
Affiliation(s)
- Denise Mafra
- Graduate Program in Nutrition Sciences, Fluminense Federal University (UFF), Niterói, Rio de Janeiro, Brazil; Graduate Program in Biological Sciences - Physiology, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro Rio de Janeiro, Brazil; Graduate Program in Medical Sciences, Fluminense Federal University (UFF), Niterói, Rio de Janeiro, Brazil.
| | - Julie A Kemp
- Graduate Program in Nutrition Sciences, Fluminense Federal University (UFF), Niterói, Rio de Janeiro, Brazil
| | - Ludmila F M F Cardozo
- Graduate Program in Cardiovascular Sciences, Fluminense Federal University (UFF), Niterói, Rio de Janeiro, Brazil
| | - Natália A Borges
- Institute of Nutrition, Rio de Janeiro State University (UERJ), Rio de Janeiro, Brazil
| | - Fabiana B Nerbass
- Research Department, Fundação Pró-Rim, Joinville, Santa Catarina, Brazil
| | - Lívia Alvarenga
- Graduate Program in Medical Sciences, Fluminense Federal University (UFF), Niterói, Rio de Janeiro, Brazil
| | - Kamyar Kalantar-Zadeh
- Divsion of Nephrology, Hypertension and Kidney Transplantation, University of California Irvine, Orange, California
| |
Collapse
|
10
|
Khemiri H, Gdoura M, Ben Halima S, Krichen H, Cammà C, Lorusso A, Ancora M, Di Pasquale A, Cherni A, Touzi H, Sadraoui A, Meddeb Z, Hogga N, Ammi R, Triki H, Haddad-Boubaker S. SARS-CoV-2 excretion kinetics in nasopharyngeal and stool samples from the pediatric population. Front Med (Lausanne) 2023; 10:1226207. [PMID: 38020093 PMCID: PMC10643538 DOI: 10.3389/fmed.2023.1226207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Accepted: 10/02/2023] [Indexed: 12/01/2023] Open
Abstract
Background The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is responsible for serious respiratory infections in humans. Even in the absence of respiratory symptoms, gastrointestinal (GI) signs were commonly reported in adults and children. Thus, oral-fecal transmission was suspected as a possible route of infection. The objective of this study was to describe RNA shedding in nasopharyngeal and stool samples obtained from asymptomatic and symptomatic children and to investigate virus viability. Methods This study included 179 stool and 191 nasopharyngeal samples obtained from 71 children, which included symptomatic (n = 64) and asymptomatic (n = 7) ones. They were collected every 7 days from the onset of the infection until negativation. Viral RNA was detected by real-time RT-PCR, targeting the N and ORF1 genes. Whole-genome sequencing was performed for positive cases. Viral isolation was assessed on Vero cells, followed by molecular detection confirmation. Results All cases included in this study (n = 71) were positive in their nasopharyngeal samples. SARS-CoV-2 RNA was detected in 36 stool samples obtained from 15 out of 71 (21.1%) children; 13 were symptomatic and two were asymptomatic. Excretion periods varied from 7 to 21 days and 7 to 14 days in nasopharyngeal and fecal samples, respectively. Four variants were detected: Alpha (n = 3), B.1.160 (n = 3), Delta (n = 7), and Omicron (n = 1). Inoculation of stool samples on cell culture showed no specific cytopathic effect. All cell culture supernatants were negative for RT-qPCR. Conclusion Our study demonstrated nasopharyngeal and fecal shedding of SARS-CoV-2 RNA by children up to 21 and 14 days, respectively. Fecal shedding was recorded in symptomatic and asymptomatic children. Nevertheless, SARS-CoV-2 was not isolated from positive stool samples.
Collapse
Affiliation(s)
- Haifa Khemiri
- Laboratory of Clinical Virology, WHO Regional Reference Laboratory for Poliomyelitis and Measles for the EMR, Institut Pasteur de Tunis, University of Tunis El Manar, Tunis, Tunisia
- LR 20 IPT 02 Laboratory of Virus, Host and Vectors, Institut Pasteur de Tunis, University of Tunis El Manar, Tunis, Tunisia
| | - Mariem Gdoura
- Laboratory of Clinical Virology, WHO Regional Reference Laboratory for Poliomyelitis and Measles for the EMR, Institut Pasteur de Tunis, University of Tunis El Manar, Tunis, Tunisia
- LR 20 IPT 02 Laboratory of Virus, Host and Vectors, Institut Pasteur de Tunis, University of Tunis El Manar, Tunis, Tunisia
| | - Samar Ben Halima
- Laboratory of Clinical Virology, WHO Regional Reference Laboratory for Poliomyelitis and Measles for the EMR, Institut Pasteur de Tunis, University of Tunis El Manar, Tunis, Tunisia
- LR 20 IPT 02 Laboratory of Virus, Host and Vectors, Institut Pasteur de Tunis, University of Tunis El Manar, Tunis, Tunisia
| | - Henda Krichen
- Laboratory of Clinical Virology, WHO Regional Reference Laboratory for Poliomyelitis and Measles for the EMR, Institut Pasteur de Tunis, University of Tunis El Manar, Tunis, Tunisia
- LR 20 IPT 02 Laboratory of Virus, Host and Vectors, Institut Pasteur de Tunis, University of Tunis El Manar, Tunis, Tunisia
| | - Cesare Cammà
- Istituto Zooprofilattico Sperimentale dell'Abruzzo e del Molise, Teramo, Italy
| | - Alessio Lorusso
- Istituto Zooprofilattico Sperimentale dell'Abruzzo e del Molise, Teramo, Italy
| | - Massimo Ancora
- Istituto Zooprofilattico Sperimentale dell'Abruzzo e del Molise, Teramo, Italy
| | - Adriano Di Pasquale
- Istituto Zooprofilattico Sperimentale dell'Abruzzo e del Molise, Teramo, Italy
| | - Asma Cherni
- Laboratory of Clinical Virology, WHO Regional Reference Laboratory for Poliomyelitis and Measles for the EMR, Institut Pasteur de Tunis, University of Tunis El Manar, Tunis, Tunisia
- LR 20 IPT 02 Laboratory of Virus, Host and Vectors, Institut Pasteur de Tunis, University of Tunis El Manar, Tunis, Tunisia
| | - Henda Touzi
- Laboratory of Clinical Virology, WHO Regional Reference Laboratory for Poliomyelitis and Measles for the EMR, Institut Pasteur de Tunis, University of Tunis El Manar, Tunis, Tunisia
- LR 20 IPT 02 Laboratory of Virus, Host and Vectors, Institut Pasteur de Tunis, University of Tunis El Manar, Tunis, Tunisia
| | - Amel Sadraoui
- Laboratory of Clinical Virology, WHO Regional Reference Laboratory for Poliomyelitis and Measles for the EMR, Institut Pasteur de Tunis, University of Tunis El Manar, Tunis, Tunisia
- LR 20 IPT 02 Laboratory of Virus, Host and Vectors, Institut Pasteur de Tunis, University of Tunis El Manar, Tunis, Tunisia
| | - Zina Meddeb
- Laboratory of Clinical Virology, WHO Regional Reference Laboratory for Poliomyelitis and Measles for the EMR, Institut Pasteur de Tunis, University of Tunis El Manar, Tunis, Tunisia
- LR 20 IPT 02 Laboratory of Virus, Host and Vectors, Institut Pasteur de Tunis, University of Tunis El Manar, Tunis, Tunisia
| | - Nahed Hogga
- Laboratory of Clinical Virology, WHO Regional Reference Laboratory for Poliomyelitis and Measles for the EMR, Institut Pasteur de Tunis, University of Tunis El Manar, Tunis, Tunisia
- LR 20 IPT 02 Laboratory of Virus, Host and Vectors, Institut Pasteur de Tunis, University of Tunis El Manar, Tunis, Tunisia
| | - Radhia Ammi
- Service of External Consultants, Institut Pasteur de Tunis, Tunis, Tunisia
| | - Henda Triki
- Laboratory of Clinical Virology, WHO Regional Reference Laboratory for Poliomyelitis and Measles for the EMR, Institut Pasteur de Tunis, University of Tunis El Manar, Tunis, Tunisia
- LR 20 IPT 02 Laboratory of Virus, Host and Vectors, Institut Pasteur de Tunis, University of Tunis El Manar, Tunis, Tunisia
| | - Sondes Haddad-Boubaker
- Laboratory of Clinical Virology, WHO Regional Reference Laboratory for Poliomyelitis and Measles for the EMR, Institut Pasteur de Tunis, University of Tunis El Manar, Tunis, Tunisia
- LR 20 IPT 02 Laboratory of Virus, Host and Vectors, Institut Pasteur de Tunis, University of Tunis El Manar, Tunis, Tunisia
| |
Collapse
|
11
|
Ho QY, Sultana R, Lee TL, Thangaraju S, Kee T, Htay H. Coronavirus disease 2019 in kidney transplant recipients: a systematic review and meta-analysis. Singapore Med J 2023; 64:593-602. [PMID: 34688231 PMCID: PMC10645004 DOI: 10.11622/smedj.2021171] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Accepted: 05/27/2021] [Indexed: 11/18/2022]
Abstract
Introduction The clinical presentation and outcomes of coronavirus disease 2019 (COVID-19) in kidney transplant recipients (KTRs) have not been well studied. Methods We performed a meta-analysis to examine the presenting features, outcomes and the effect of treatment on outcomes of KTRs with COVID-19. Database search was performed up to 5 September 2020 through PubMed, Embase, Web of Science, Scopus and CENTRAL. Results Overall, 23 studies (1,373 patients) were included in the review and meta-analysis. The most common presenting symptoms included fever (74.0%, 95% confidence interval [CI] 65.3-81.1), cough (63.3%, 95% CI 56.5-69.6) and dyspnoea (47.5%, 95% CI 39.6-55.6). Pooled rates of mortality and critical illness were 21.1% (95% CI 15.3-28.4) and 27.7% (95% CI 21.5-34.8), respectively. Acute kidney injury occurred in 38.9% (95% CI 30.6-48.1) and dialysis was required in 12.4% (95% CI 8.3-18.0) of the cases. Conclusion Kidney transplant recipients with COVID-19 have a similar clinical presentation as the general population, but they have higher morbidity and mortality. It is uncertain whether high-dose corticosteroid or hydroxychloroquine reduces the risks of mortality in KTRs with COVID-19.
Collapse
Affiliation(s)
- Quan Yao Ho
- Department of Renal Medicine, Singapore General Hospital, Singapore
- SingHealth Duke-NUS Transplant Centre, Singapore
| | | | - Tung Lin Lee
- Department of Renal Medicine, Singapore General Hospital, Singapore
| | - Sobhana Thangaraju
- Department of Renal Medicine, Singapore General Hospital, Singapore
- SingHealth Duke-NUS Transplant Centre, Singapore
| | - Terence Kee
- Department of Renal Medicine, Singapore General Hospital, Singapore
- SingHealth Duke-NUS Transplant Centre, Singapore
| | - Htay Htay
- Department of Renal Medicine, Singapore General Hospital, Singapore
- Duke-NUS Medical School, Singapore
| |
Collapse
|
12
|
Azarbakhsh H, Moftakhar L, Valipour A, Mirahmadizadeh A, Moradi HA, Piraee E. Epidemiological features and consequences of COVID-19 in patients with and without gastrointestinal symptoms in southwestern Iran. A retrospective observational study. Health Sci Rep 2023; 6:e1499. [PMID: 37732104 PMCID: PMC10507146 DOI: 10.1002/hsr2.1499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Revised: 07/31/2023] [Accepted: 08/02/2023] [Indexed: 09/22/2023] Open
Abstract
Background and Aims Some studies have shown that in addition to respiratory symptoms, gastrointestinal (GI) manifestations reported in patients with coronavirus disease 2019 (COVID-19). The aim of this study was to compare the epidemiological features and consequences of COVID-19 in patients with and without GI symptoms. Methods This retrospective observational study concluded on 15,323 COVID-19 patients with GI symptoms and 95,724 patients without symptoms. All symptoms and comorbidities of the patients collected. To investigate the differences between qualitative variables in the two groups, χ 2 test was used. Logistic regression analysis also used to identify determinants of mortality in patients with COVID-19. Results During the course of the study, 111,047 cases of COVID-19 occurred. Of these, 13.8% of patients had GI symptoms, and 9.9% of deaths due to COVID-19 occurred in these patients. The most common reported GI symptoms among COVID-19 patients were nausea, vomiting, and diarrhea. In addition, comorbidities, such as diabetes, cardiovascular disease, and thyroid disease were significantly higher in patients with GI symptoms. The result of multiple logistic regression showed that the chance of mortality is higher in a patient with COVID-19 who have dyspnea, fever, cough, hypertension, cardiovascular disease, diabetes, immunodeficiency, chronic kidney disease, thyroid disease, chronic pulmonary disease, and male gender. The chance of death was lower in people with GI symptoms. Conclusion According to the findings of this study, nausea, vomiting, and diarrhea were the most common GI symptoms. Also, the chance of death is higher in people with co-morbidities such as cardiovascular diseases, diabetes, and high blood pressure. Therefore, it is necessary to follow these people closely.
Collapse
Affiliation(s)
| | - Leila Moftakhar
- Department of Public Health, Student Research Committee Shiraz University of Medical Sciences Shiraz Iran
| | - Aliasghar Valipour
- Department of Public Health Abadan Faculty of Medical Sciences Abadan Iran
| | - Alireza Mirahmadizadeh
- Department of Epidemiology, Non-communicable Diseases Research Center Shiraz University of Medical Sciences Shiraz Iran
| | - Hekmat Allah Moradi
- Department of Disaster and Emergency Health, Health Human Resources Research Center, School of Health Management and Information Sciences Shiraz University of Medical Sciences Shiraz Iran
| | - Elahe Piraee
- Department of Public Health Social Determinants of Health Research Center, Yasuj University of Medical Sciences Yasuj Iran
| |
Collapse
|
13
|
Durairajan SSK, Singh AK, Saravanan UB, Namachivayam M, Radhakrishnan M, Huang JD, Dhodapkar R, Zhang H. Gastrointestinal Manifestations of SARS-CoV-2: Transmission, Pathogenesis, Immunomodulation, Microflora Dysbiosis, and Clinical Implications. Viruses 2023; 15:1231. [PMID: 37376531 DOI: 10.3390/v15061231] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 05/12/2023] [Accepted: 05/15/2023] [Indexed: 06/29/2023] Open
Abstract
The clinical manifestation of COVID-19, caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), in the respiratory system of humans is widely recognized. There is increasing evidence suggesting that SARS-CoV-2 possesses the capability to invade the gastrointestinal (GI) system, leading to the manifestation of symptoms such as vomiting, diarrhea, abdominal pain, and GI lesions. These symptoms subsequently contribute to the development of gastroenteritis and inflammatory bowel disease (IBD). Nevertheless, the pathophysiological mechanisms linking these GI symptoms to SARS-CoV-2 infection remain unelucidated. During infection, SARS-CoV-2 binds to angiotensin-converting enzyme 2 and other host proteases in the GI tract during the infection, possibly causing GI symptoms by damaging the intestinal barrier and stimulating inflammatory factor production, respectively. The symptoms of COVID-19-induced GI infection and IBD include intestinal inflammation, mucosal hyperpermeability, bacterial overgrowth, dysbiosis, and changes in blood and fecal metabolomics. Deciphering the pathogenesis of COVID-19 and understanding its exacerbation may provide insights into disease prognosis and pave the way for the discovery of potential novel targets for disease prevention or treatment. Besides the usual transmission routes, SARS-CoV-2 can also be transmitted via the feces of an infected person. Hence, it is crucial to implement preventive and control measures in order to mitigate the fecal-to-oral transmission of SARS-CoV-2. Within this context, the identification and diagnosis of GI tract symptoms during these infections assume significance as they facilitate early detection of the disease and the development of targeted therapeutics. The present review discusses the receptors, pathogenesis, and transmission of SARS-CoV-2, with a particular focus on the induction of gut immune responses, the influence of gut microbes, and potential therapeutic targets against COVID-19-induced GI infection and IBD.
Collapse
Affiliation(s)
| | - Abhay Kumar Singh
- Department of Microbiology, School of Life Sciences, Central University of Tamil Nadu, Tiruvarur 610005, India
| | - Udhaya Bharathy Saravanan
- Department of Microbiology, School of Life Sciences, Central University of Tamil Nadu, Tiruvarur 610005, India
| | - Mayurikaa Namachivayam
- Department of Microbiology, School of Life Sciences, Central University of Tamil Nadu, Tiruvarur 610005, India
| | - Moorthi Radhakrishnan
- Department of Microbiology, School of Life Sciences, Central University of Tamil Nadu, Tiruvarur 610005, India
| | - Jian-Dong Huang
- Department of Biochemistry, School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 21 Sassoon Road, Pokfulam, Hong Kong 999077, China
- CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Rahul Dhodapkar
- Department of Microbiology, Jawaharlal Institute of Postgraduate Medical Education & Research (JIPMER), Government of India, Puducherry 605006, India
| | - Hongjie Zhang
- School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong 999077, China
| |
Collapse
|
14
|
Wang S, Wu J, Ran D, Ou G, Chen Y, Xu H, Deng L, Chen X. Study of the Relationship between Mucosal Immunity and Commensal Microbiota: A Bibliometric Analysis. Nutrients 2023; 15:nu15102398. [PMID: 37242281 DOI: 10.3390/nu15102398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 05/17/2023] [Accepted: 05/18/2023] [Indexed: 05/28/2023] Open
Abstract
This study presents the first bibliometric evaluation and systematic analysis of publications related to mucosal immunity and commensal microbiota over the last two decades and summarizes the contribution of countries, institutions, and scholars in the study of this field. A total of 1423 articles related to mucosal immunity and commensal microbiota in vivo published in 532 journals by 7774 authors from 1771 institutions in 74 countries/regions were analyzed. The interaction between commensal microbiota in vivo and mucosal immunity is essential in regulating the immune response of the body, maintaining communication between different kinds of commensal microbiota and the host, and so on. Several hot spots in this field have been found to have received extensive attention in recent years, especially the effects of metabolites of key strains on mucosal immunity, the physiopathological phenomena of commensal microbiota in various sites including the intestine, and the relationship between COVID-19, mucosal immunity and microbiota. We hope that the full picture of the last 20 years in this research area provided in this study will serve to deliver necessary cutting-edge information to relevant researchers.
Collapse
Affiliation(s)
- Shiqi Wang
- School of Traditional Chinese Medicine, Jinan University, Guangzhou 510632, China
| | - Jialin Wu
- School of Traditional Chinese Medicine, Jinan University, Guangzhou 510632, China
| | - Duo Ran
- School of Traditional Chinese Medicine, Jinan University, Guangzhou 510632, China
| | - Guosen Ou
- School of Traditional Chinese Medicine, Jinan University, Guangzhou 510632, China
| | - Yaokang Chen
- School of Traditional Chinese Medicine, Jinan University, Guangzhou 510632, China
| | - Huachong Xu
- School of Traditional Chinese Medicine, Jinan University, Guangzhou 510632, China
| | - Li Deng
- School of Traditional Chinese Medicine, Jinan University, Guangzhou 510632, China
| | - Xiaoyin Chen
- School of Traditional Chinese Medicine, Jinan University, Guangzhou 510632, China
| |
Collapse
|
15
|
Moon Y. Gut distress and intervention via communications of SARS-CoV-2 with mucosal exposome. Front Public Health 2023; 11:1098774. [PMID: 37139365 PMCID: PMC10150023 DOI: 10.3389/fpubh.2023.1098774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 03/27/2023] [Indexed: 05/05/2023] Open
Abstract
Acute coronavirus disease 2019 (COVID-19) has been associated with prevalent gastrointestinal distress, characterized by fecal shedding of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) RNA or persistent antigen presence in the gut. Using a meta-analysis, the present review addressed gastrointestinal symptoms, such as nausea, vomiting, abdominal pain, and diarrhea. Despite limited data on the gut-lung axis, viral transmission to the gut and its influence on gut mucosa and microbial community were found to be associated by means of various biochemical mechanisms. Notably, the prolonged presence of viral antigens and disrupted mucosal immunity may increase gut microbial and inflammatory risks, leading to acute pathological outcomes or post-acute COVID-19 symptoms. Patients with COVID-19 exhibit lower bacterial diversity and a higher relative abundance of opportunistic pathogens in their gut microbiota than healthy controls. Considering the dysbiotic changes during infection, remodeling or supplementation with beneficial microbial communities may counteract adverse outcomes in the gut and other organs in patients with COVID-19. Moreover, nutritional status, such as vitamin D deficiency, has been associated with disease severity in patients with COVID-19 via the regulation of the gut microbial community and host immunity. The nutritional and microbiological interventions improve the gut exposome including the host immunity, gut microbiota, and nutritional status, contributing to defense against acute or post-acute COVID-19 in the gut-lung axis.
Collapse
Affiliation(s)
- Yuseok Moon
- Laboratory of Mucosal Exposome and Biomodulation, Department of Integrative Biomedical Sciences, Pusan National University, Yangsan-si, Republic of Korea
- Biomedical Research Institute, Pusan National University, Busan, Republic of Korea
- Graduate Program of Genomic Data Sciences, Pusan National University, Yangsan-si, Republic of Korea
| |
Collapse
|
16
|
Leyfman Y, Emmanuel N, Menon GP, Joshi M, Wilkerson WB, Cappelli J, Erick TK, Park CH, Sharma P. Cancer and COVID-19: unravelling the immunological interplay with a review of promising therapies against severe SARS-CoV-2 for cancer patients. J Hematol Oncol 2023; 16:39. [PMID: 37055774 PMCID: PMC10100631 DOI: 10.1186/s13045-023-01432-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Accepted: 03/25/2023] [Indexed: 04/15/2023] Open
Abstract
Cancer patients, due to their immunocompromised status, are at an increased risk for severe SARS-CoV-2 infection. Since severe SARS-CoV-2 infection causes multiple organ damage through IL-6-mediated inflammation while stimulating hypoxia, and malignancy promotes hypoxia-induced cellular metabolic alterations leading to cell death, we propose a mechanistic interplay between both conditions that results in an upregulation of IL-6 secretion resulting in enhanced cytokine production and systemic injury. Hypoxia mediated by both conditions results in cell necrosis, dysregulation of oxidative phosphorylation, and mitochondrial dysfunction. This produces free radicals and cytokines that result in systemic inflammatory injury. Hypoxia also catalyzes the breakdown of COX-1 and 2 resulting in bronchoconstriction and pulmonary edema, which further exacerbates tissue hypoxia. Given this disease model, therapeutic options are currently being studied against severe SARS-COV-2. In this study, we review several promising therapies against severe disease supported by clinical trial evidence-including Allocetra, monoclonal antibodies (Tixagevimab-Cilgavimab), peginterferon lambda, Baricitinib, Remdesivir, Sarilumab, Tocilizumab, Anakinra, Bevacizumab, exosomes, and mesenchymal stem cells. Due to the virus's rapid adaptive evolution and diverse symptomatic manifestation, the use of combination therapies offers a promising approach to decrease systemic injury. By investing in such targeted interventions, cases of severe SARS-CoV-2 should decrease along with its associated long-term sequelae and thereby allow cancer patients to resume their treatments.
Collapse
Affiliation(s)
- Yan Leyfman
- Icahn School of Medicine at Mount Sinai South Nassau, Rockville Centre, NY, USA
| | - Nancy Emmanuel
- Hospital das Clínicas of the Faculty of Medicine of the University of São Paulo, São Paulo, Brazil
| | | | - Muskan Joshi
- Tbilisi State Medical University, Tbilisi, Georgia
| | | | | | | | | | - Pushpa Sharma
- Department of Anesthesiology, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA.
| |
Collapse
|
17
|
Esseili MA. Great escape: how infectious SARS-CoV-2 avoids inactivation by gastric acidity and intestinal bile. Gut 2023; 72:808-810. [PMID: 35672040 DOI: 10.1136/gutjnl-2021-326624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 06/01/2022] [Indexed: 11/04/2022]
Affiliation(s)
- Malak A Esseili
- Food Science and Technology, Center for Food Safety, University of Georgia, Griffin, Georgia, USA
| |
Collapse
|
18
|
El Hennawy HM, Safar O, Elatreisy A, Al Faifi AS, Shalkamy O, Hadi SA, Alqahtani M, Alkahtani SA, Alqahtani FS, El Nazer W, Al Atta E, Ibrahim AT, Abdelaziz AA, Mirza N, Mahedy A, Tom NM, Assiri Y, Al Fageeh A, Elgamal G, Al Shehri AA, Zaitoun MF. The Outcome of COVID-19 Infection on Kidney Transplantation Recipients in Southern Saudi Arabia: Single-Center Experience. Transplant Proc 2023; 55:521-529. [PMID: 36681582 PMCID: PMC9826984 DOI: 10.1016/j.transproceed.2022.12.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 11/27/2022] [Accepted: 12/19/2022] [Indexed: 01/11/2023]
Abstract
BACKGROUND To report the incidence, risk factors, and outcome of severe COVID-19 disease in kidney transplant recipients attending a Saudi hospital at a single center in the Kingdom of Saudi Arabia. METHODS A retrospective chart-based cohort study involving all kidney transplant recipients tested for COVID-19 in the Armed Forces Hospital Southern Region, KSA. RESULTS Of 532 kidney transplant recipients who reported to the center, from March 2020 to June 2022, 180 were tested for COVID-19. Of these recipients, 31 (17%) tested positive. Among the 31 positive recipients, 11 were treated at home, 15 were admitted to the noncritical isolation ward, and 5 were admitted to the intensive care unit (ICU). Older age (P = .0001), higher body mass index (P = .0001), and history of hypertension (P = .0023) were more frequent in the COVID-19-positive recipients. Admission to the ICU was more frequent in older recipients (P = .0322) with a history of ischemic heart disease (P = .06) and higher creatinine baseline (P = .08) presenting with dyspnea (P = .0174), and acute allograft dysfunction (P = .002). In the ICU group, 4 (80%) patients required hemodialysis, and 4 (80%) died. CONCLUSIONS Kidney transplant recipients with COVID-19 could have a higher risk for developing acute kidney injury, dialysis, and mortality than the general population. ICU admission and renal replacement therapy were more evident in older recipients with a history of ischemic heart disease, presenting with shortness of breath (P = .017) and a higher serum creatinine baseline. Acute allograft dysfunction was the independent predictor of mortality among patients admitted to the ICU.
Collapse
Affiliation(s)
- Hany M El Hennawy
- Surgery Department, Section of Transplantation, Armed Forces Hospitals Southern Region, Khamis Mushayte, Saudi Arabia,Address correspondence to Hany M El Hennawy, MD, Department of Surgery, Section of Transplantation, Armed Forces Hospitals Southern Region, Khamis Mushayte, 101, KSA
| | - Omar Safar
- Urology Department, Armed Forces Hospitals Southern Region, Khamis Mushayte, Saudi Arabia
| | - Adel Elatreisy
- Urology Department, Faculty of Medicine, Al-Azhar University, Cairo, Egypt
| | - Abdullah S Al Faifi
- Surgery Department, Section of Transplantation, Armed Forces Hospitals Southern Region, Khamis Mushayte, Saudi Arabia
| | - Osama Shalkamy
- Urology Department, Faculty of Medicine, Al-Azhar University, Cairo, Egypt
| | - Sara Abdullah Hadi
- Surgery Department, Section of Transplantation, Armed Forces Hospitals Southern Region, Khamis Mushayte, Saudi Arabia
| | - Mofareh Alqahtani
- Urology Department, Armed Forces Hospitals Southern Region, Khamis Mushayte, Saudi Arabia
| | - Sultan Ahmad Alkahtani
- Pathology and Laboratory Department, Armed Forces Hospitals Southern Region, Khamis Mushayte, Saudi Arabia
| | - Faisal Saeed Alqahtani
- Adult Infectious Diseases Section, Internal Medicine Department, Armed Forces Hospitals Southern Region, Khamis Mushayte, Saudi Arabia
| | - Weam El Nazer
- Nephrology Department, Armed Forces Hospitals Southern Region, Khamis Mushayte, Saudi Arabia
| | - Eisa Al Atta
- Surgery Department, Section of Transplantation, Armed Forces Hospitals Southern Region, Khamis Mushayte, Saudi Arabia
| | - Asad Taha Ibrahim
- Anesthesia Department, Armed Forces Hospitals Southern Region, Khamis Mushayte, Saudi Arabia
| | - Abdelaziz a Abdelaziz
- Nephrology Department, Armed Forces Hospitals Southern Region, Khamis Mushayte, Saudi Arabia
| | - Naveed Mirza
- Nephrology Department, Armed Forces Hospitals Southern Region, Khamis Mushayte, Saudi Arabia
| | - Ahmed Mahedy
- Nephrology Department, Armed Forces Hospitals Southern Region, Khamis Mushayte, Saudi Arabia
| | - Nayana Mary Tom
- Anesthesia Department, Armed Forces Hospitals Southern Region, Khamis Mushayte, Saudi Arabia
| | - Yahya Assiri
- Nephrology Department, Armed Forces Hospitals Southern Region, Khamis Mushayte, Saudi Arabia
| | - Ali Al Fageeh
- Nephrology Department, Armed Forces Hospitals Southern Region, Khamis Mushayte, Saudi Arabia
| | - Galal Elgamal
- Anesthesia Department, Armed Forces Hospitals Southern Region, Khamis Mushayte, Saudi Arabia
| | - Ali Amer Al Shehri
- Adult Infectious Diseases Section, Internal Medicine Department, Armed Forces Hospitals Southern Region, Khamis Mushayte, Saudi Arabia
| | - Mohammad F Zaitoun
- Pharmacy Department, Armed Forces Hospitals Southern Region, Khamis Mushayte, Saudi Arabia
| |
Collapse
|
19
|
Brogna C, Viduto V, Fabrowski M, Cristoni S, Marino G, Montano L, Piscopo M. The importance of the gut microbiome in the pathogenesis and transmission of SARS-CoV-2. Gut Microbes 2023; 15:2244718. [PMID: 37559387 PMCID: PMC10416738 DOI: 10.1080/19490976.2023.2244718] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Revised: 07/22/2023] [Accepted: 08/01/2023] [Indexed: 08/11/2023] Open
Abstract
Zhou et al. study nicely traces a significant topic in COVID-19 infection: the persistence of the virus within the intestinal tract. Many pathological mechanisms have been noted in the current literature about the mode of infection and propagation of SARS-CoV-2 in the human body. Nevertheless, there are still many concerns about this: only some things seem well understood. We present a different point of view by illustrating the importance of the gut microbiome in the pathogenesis of COVID-19 disorders.
Collapse
Affiliation(s)
- Carlo Brogna
- Department of Research, Craniomed Group Facility Srl, Bresso, Italy
| | | | - Mark Fabrowski
- Emergency Department, University Hospitals Sussex, Brighton, UK
| | - Simone Cristoni
- Department of Chemistry, ISB – Ion Source & Biotechnologies Srl, Bresso, Italy
| | - Giuliano Marino
- Marsan Consulting Srl., Public Health Company; via Dei Fiorentini, Napoli, Italy
| | - Luigi Montano
- Andrology Unit and Service of LifeStyle Medicine in Uro-Andrology, Local Health Authority (ASL) Salerno, Salerno, Italy
| | - Marina Piscopo
- Department of Biology, University of Naples Federico II, Naples, Italy
| |
Collapse
|
20
|
In Silico Analysis of Bacteriocins from Lactic Acid Bacteria Against SARS-CoV-2. Probiotics Antimicrob Proteins 2023; 15:17-29. [PMID: 34837166 PMCID: PMC8626284 DOI: 10.1007/s12602-021-09879-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/11/2021] [Indexed: 01/18/2023]
Abstract
The COVID-19 pandemic caused by a novel coronavirus (SARS-CoV-2) is a serious health concern in the twenty-first century for scientists, health workers, and all humans. The absence of specific biotherapeutics requires new strategies to prevent the spread and prophylaxis of the novel virus and its variants. The SARS-CoV-2 virus shows pathogenesis by entering the host cells via spike protein and Angiotensin-Converting Enzyme 2 receptor protein. Thus, the present study aims to compute the binding energies between a wide range of bacteriocins with receptor-binding domain (RBD) on spike proteins of wild type (WT) and beta variant (lineage B.1.351). Molecular docking analyses were performed to evaluate binding energies. Upon achieving the best bio-peptides with the highest docking scores, further molecular dynamics (MD) simulations were performed to validate the structure and interaction stability. Protein-protein docking of the chosen 22 biopeptides with WT-RBD showed docking scores lower than -7.9 kcal/mol. Pediocin PA-1 and salivaricin P showed the lowest (best) docking scores of - 12 kcal/mol. Pediocin PA-1, salivaricin B, and salivaricin P showed a remarkable increase in the double mutant's predicted binding affinity with -13.8 kcal/mol, -13.0 kcal/mol, and -12.5 kcal/mol, respectively. Also, a better predicted binding affinity of pediocin PA-1 and salivaricin B against triple mutant was observed compared to the WT. Thus, pediocin PA-1 binds stronger to mutants of the RBD, particularly to double and triple mutants. Salivaricin B showed a better predicted binding affinity towards triple mutant compared to WT, showing that it might be another bacteriocin with potential activity against the SARS-CoV-2 beta variant. Overall, pediocin PA-1, salivaricin P, and salivaricin B are the most promising candidates for inhibiting SARS-CoV-2 (including lineage B.1.351) entrance into the human cells. These bacteriocins derived from lactic acid bacteria hold promising potential for paving an alternative way for treatment and prophylaxis of WT and beta variants.
Collapse
|
21
|
Pooled evidence from preclinical and clinical studies for stem cell-based therapy in ARDS and COVID-19. Mol Cell Biochem 2022; 478:1487-1518. [PMID: 36394787 DOI: 10.1007/s11010-022-04601-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 10/24/2022] [Indexed: 11/18/2022]
|
22
|
Liu Q, Su Q, Zhang F, Tun HM, Mak JWY, Lui GCY, Ng SSS, Ching JYL, Li A, Lu W, Liu C, Cheung CP, Hui DSC, Chan PKS, Chan FKL, Ng SC. Multi-kingdom gut microbiota analyses define COVID-19 severity and post-acute COVID-19 syndrome. Nat Commun 2022; 13:6806. [PMID: 36357381 PMCID: PMC9648868 DOI: 10.1038/s41467-022-34535-8] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 10/26/2022] [Indexed: 11/12/2022] Open
Abstract
Our knowledge of the role of the gut microbiome in acute coronavirus disease 2019 (COVID-19) and post-acute COVID-19 is rapidly increasing, whereas little is known regarding the contribution of multi-kingdom microbiota and host-microbial interactions to COVID-19 severity and consequences. Herein, we perform an integrated analysis using 296 fecal metagenomes, 79 fecal metabolomics, viral load in 1378 respiratory tract samples, and clinical features of 133 COVID-19 patients prospectively followed for up to 6 months. Metagenomic-based clustering identifies two robust ecological clusters (hereafter referred to as Clusters 1 and 2), of which Cluster 1 is significantly associated with severe COVID-19 and the development of post-acute COVID-19 syndrome. Significant differences between clusters could be explained by both multi-kingdom ecological drivers (bacteria, fungi, and viruses) and host factors with a good predictive value and an area under the curve (AUC) of 0.98. A model combining host and microbial factors could predict the duration of respiratory viral shedding with 82.1% accuracy (error ± 3 days). These results highlight the potential utility of host phenotype and multi-kingdom microbiota profiling as a prognostic tool for patients with COVID-19.
Collapse
Affiliation(s)
- Qin Liu
- Microbiota I-Center (MagIC), Hong Kong SAR, China
- Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Li Ka Shing Institute of Health Sciences, State Key Laboratory of Digestive Disease, Institute of Digestive Disease, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
- Center for Gut Microbiota Research, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Qi Su
- Microbiota I-Center (MagIC), Hong Kong SAR, China
- Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Li Ka Shing Institute of Health Sciences, State Key Laboratory of Digestive Disease, Institute of Digestive Disease, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
- Center for Gut Microbiota Research, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Fen Zhang
- Microbiota I-Center (MagIC), Hong Kong SAR, China
- Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Li Ka Shing Institute of Health Sciences, State Key Laboratory of Digestive Disease, Institute of Digestive Disease, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
- Center for Gut Microbiota Research, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Hein M Tun
- Microbiota I-Center (MagIC), Hong Kong SAR, China
- Li Ka Shing Institute of Health Sciences, State Key Laboratory of Digestive Disease, Institute of Digestive Disease, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
- The Jockey Club School of Public Health and Primary Care, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Joyce Wing Yan Mak
- Microbiota I-Center (MagIC), Hong Kong SAR, China
- Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Li Ka Shing Institute of Health Sciences, State Key Laboratory of Digestive Disease, Institute of Digestive Disease, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
- Center for Gut Microbiota Research, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Grace Chung-Yan Lui
- Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Stanley Ho Centre for Emerging Infectious Diseases, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Susanna So Shan Ng
- Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Jessica Y L Ching
- Microbiota I-Center (MagIC), Hong Kong SAR, China
- Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Li Ka Shing Institute of Health Sciences, State Key Laboratory of Digestive Disease, Institute of Digestive Disease, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
- Center for Gut Microbiota Research, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Amy Li
- Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Li Ka Shing Institute of Health Sciences, State Key Laboratory of Digestive Disease, Institute of Digestive Disease, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
- Center for Gut Microbiota Research, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Wenqi Lu
- Microbiota I-Center (MagIC), Hong Kong SAR, China
- Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Li Ka Shing Institute of Health Sciences, State Key Laboratory of Digestive Disease, Institute of Digestive Disease, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
- Center for Gut Microbiota Research, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Chenyu Liu
- Microbiota I-Center (MagIC), Hong Kong SAR, China
- Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Li Ka Shing Institute of Health Sciences, State Key Laboratory of Digestive Disease, Institute of Digestive Disease, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
- Center for Gut Microbiota Research, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Chun Pan Cheung
- Microbiota I-Center (MagIC), Hong Kong SAR, China
- Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Li Ka Shing Institute of Health Sciences, State Key Laboratory of Digestive Disease, Institute of Digestive Disease, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
- Center for Gut Microbiota Research, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - David S C Hui
- Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Stanley Ho Centre for Emerging Infectious Diseases, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Paul K S Chan
- Center for Gut Microbiota Research, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
- Department of Microbiology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Francis Ka Leung Chan
- Microbiota I-Center (MagIC), Hong Kong SAR, China
- Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Li Ka Shing Institute of Health Sciences, State Key Laboratory of Digestive Disease, Institute of Digestive Disease, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
- Center for Gut Microbiota Research, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Siew C Ng
- Microbiota I-Center (MagIC), Hong Kong SAR, China.
- Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China.
- Li Ka Shing Institute of Health Sciences, State Key Laboratory of Digestive Disease, Institute of Digestive Disease, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China.
- Center for Gut Microbiota Research, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China.
| |
Collapse
|
23
|
Abstract
Infectious esophagitis is the third most common cause of esophagitis after gastroesophageal reflux disease and eosinophilic esophagitis (EoE) and should always be considered in the differential of patients with dysphagia and odynophagia. The most common organisms causing disease are candida, Herpes simplex virus (HSV) and cytomegalovirus (CMV). It is well recognized that an impaired local or systemic immune system is a risk factor for disease; however, esophageal dysmotility and disruptions in esophageal homeostasis and the esophageal milieu are likely to represent additional risk factors in disease pathogenesis.
Collapse
|
24
|
Hernández-Flores TDJ, Pedraza-Brindis EJ, Cárdenas-Bedoya J, Ruíz-Carrillo JD, Méndez-Clemente AS, Martínez-Guzmán MA, Iñiguez-Gutiérrez L. Role of Micronutrients and Gut Microbiota-Derived Metabolites in COVID-19 Recovery. Int J Mol Sci 2022; 23:12324. [PMID: 36293182 PMCID: PMC9604189 DOI: 10.3390/ijms232012324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 10/11/2022] [Accepted: 10/11/2022] [Indexed: 01/08/2023] Open
Abstract
A balanced and varied diet provides diverse beneficial effects on health, such as adequate micronutrient availability and a gut microbiome in homeostasis. Besides their participation in biochemical processes as cofactors and coenzymes, vitamins and minerals have an immunoregulatory function; meanwhile, gut microbiota and its metabolites coordinate directly and indirectly the cell response through the interaction with the host receptors. Malnourishment is a crucial risk factor for several pathologies, and its involvement during the Coronavirus Disease 2019 pandemic has been reported. This pandemic has caused a significant decline in the worldwide population, especially those with chronic diseases, reduced physical activity, and elder age. Diet and gut microbiota composition are probable causes for this susceptibility, and its supplementation can play a role in reestablishing microbial homeostasis and improving immunity response against Coronavirus Disease 2019 infection and recovery. This study reviews the role of micronutrients and microbiomes in the risk of infection, the severity of disease, and the Coronavirus Disease 2019 sequelae.
Collapse
Affiliation(s)
- Teresita de Jesús Hernández-Flores
- Departamento de Disciplinas Filosófico, Metodológicas e Instrumentales, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Jalisco, Mexico
- Instituto de Investigación de Inmunodeficiencias y VIH, Hospital Civil de Guadalajara “Fray Antonio Alcalde”, Guadalajara 44280, Jalisco, Mexico
| | - Eliza Julia Pedraza-Brindis
- Departamento de Aparatos y Sistemas I, Facultad de Medicina, Universidad Autónoma de Guadalajara, Guadalajara 44670, Jalisco, Mexico
| | - Jhonathan Cárdenas-Bedoya
- Departamento de Disciplinas Filosófico, Metodológicas e Instrumentales, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Jalisco, Mexico
- Laboratorio de Inmunodeficiencias y Retrovirus Humanos, Centro de Investigación Biomédica de Occidente, Centro Médico Nacional de Occidente, Instituto Mexicano del Seguro Social, Guadalajara 44340, Jalisco, Mexico
| | - José Daniel Ruíz-Carrillo
- Clínica Medicina Familiar 1 del ISSSTE “Dr. Arturo González Guzmán”, Guadalajara 44340, Jalisco, Mexico
| | - Anibal Samael Méndez-Clemente
- Instituto de Investigación de Inmunodeficiencias y VIH, Hospital Civil de Guadalajara “Fray Antonio Alcalde”, Guadalajara 44280, Jalisco, Mexico
| | - Marco Alonso Martínez-Guzmán
- Departamento de Aparatos y Sistemas I, Facultad de Medicina, Universidad Autónoma de Guadalajara, Guadalajara 44670, Jalisco, Mexico
| | - Liliana Iñiguez-Gutiérrez
- Instituto de Investigación de Inmunodeficiencias y VIH, Hospital Civil de Guadalajara “Fray Antonio Alcalde”, Guadalajara 44280, Jalisco, Mexico
- Departamento de Aparatos y Sistemas I, Facultad de Medicina, Universidad Autónoma de Guadalajara, Guadalajara 44670, Jalisco, Mexico
| |
Collapse
|
25
|
Gut Microbiota Dynamics in Relation to Long-COVID-19 Syndrome: Role of Probiotics to Combat Psychiatric Complications. Metabolites 2022; 12:metabo12100912. [PMID: 36295814 PMCID: PMC9611210 DOI: 10.3390/metabo12100912] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Revised: 09/11/2022] [Accepted: 09/19/2022] [Indexed: 11/17/2022] Open
Abstract
Increasing numbers of patients who recover from COVID-19 report lasting symptoms, such as fatigue, muscle weakness, dementia, and insomnia, known collectively as post-acute COVID syndrome or long COVID. These lasting symptoms have been examined in different studies and found to influence multiple organs, sometimes resulting in life-threating conditions. In this review, these symptoms are discussed in connection to the COVID-19 and long-COVID-19 immune changes, highlighting oral and psychiatric health, as this work focuses on the gut microbiota’s link to long-COVID-19 manifestations in the liver, heart, kidney, brain, and spleen. A model of this is presented to show the biological and clinical implications of gut microbiota in SARS-CoV-2 infection and how they could possibly affect the therapeutic aspects of the disease. Probiotics can support the body’s systems in fighting viral infections. This review focuses on current knowledge about the use of probiotics as adjuvant therapies for COVID-19 patients that might help to prevent long-COVID-19 complications.
Collapse
|
26
|
Zhao R, Shi J, Song Y, Zhang R. Comparison of laboratory characteristics of gastrointestinal symptoms and nongastrointestinal symptoms in patients infected with COVID-19: a systematic review and meta-analysis. Therap Adv Gastroenterol 2022; 15:17562848221116264. [PMID: 36035309 PMCID: PMC9403443 DOI: 10.1177/17562848221116264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Accepted: 07/11/2022] [Indexed: 02/04/2023] Open
Abstract
Background The Coronavirus Disease 2019 (COVID-19) pandemic poses a massive crisis to global public health. Gastrointestinal (GI) symptoms are increasingly reported in COVID-19. The characteristics of laboratory findings of COVID-19 are critical for clinical diagnosis and treatment. Objectives The study aimed to summarize laboratory features in COVID-19 with GI symptoms and non-GI symptoms. Design This study was a systematic review and meta-analysis. Electronic literature searches were conducted for studies that included patients infected COVID-19 with GI symptoms and non-GI symptoms. GI symptoms included diarrhea, abdominal pain, nausea and vomiting, and anorexia. This study used a random-effects model to assess pooled data. Data sources and methods We systematically searched PubMed, Embase, Cochrane, Web of Science for studies through 31 October 2021, with no language restrictions. We used the following search terms: 'COVID-19' OR '2019-nCoV' OR 'SARS-CoV-2' OR 'coronavirus 2019' OR 'severe acute respiratory syndrome coronavirus 2' OR 'coronavirus' OR 'novel coronavirus' OR 'nCoV' AND 'gastrointestinal symptoms' OR 'digestive symptoms' AND 'clinical feature' OR 'clinical characteristics.' Data mostly originated from Chinese and American studies. Results Of 796 identified studies, 14 were eligible and were included in our analysis (N = 8396 participants). Meta-analysis showed that GI symptoms group had an elevated alanine aminotransferase (ALT) [pooled mean difference (MD), 4.5 U/L; 95% confidence interval, [0.45, 8.55]; p = 0.03; I 2 = 87%]. No publication bias was detected by Begg's and Egger's regression test (p = 0.130). COVID-19 with the GI symptoms also showed a trend toward decreased white blood cell count, lymphopenia, neutrophilia, thrombocytopenia and elevated total bilirubin. Conclusion GI symptoms are common in COVID-19. No significant differences were found in most laboratory indicators except elevated ALT. Registration CRD42020209039 (PROSPERO).
Collapse
Affiliation(s)
- Rui Zhao
- Department of Clinical Laboratory, Beijing
Chaoyang Hospital, Capital Medical University, Beijing, PR China
| | - Jie Shi
- Department of Clinical Laboratory, Beijing
Chaoyang Hospital, Capital Medical University, Beijing, PR China
| | - Yichuan Song
- Department of Clinical Laboratory, Beijing
Chaoyang Hospital, Capital Medical University, Beijing, PR China
| | - Rui Zhang
- Department of Clinical Laboratory, Beijing
Chaoyang Hospital, Capital Medical University, No. 8, Gongtinan Road,
Chaoyang District, Beijing 100020, PR China
| |
Collapse
|
27
|
Chakraborty C, Sharma AR, Bhattacharya M, Dhama K, Lee SS. Altered gut microbiota patterns in COVID-19: Markers for inflammation and disease severity. World J Gastroenterol 2022; 28:2802-2822. [PMID: 35978881 PMCID: PMC9280735 DOI: 10.3748/wjg.v28.i25.2802] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 03/19/2022] [Accepted: 05/14/2022] [Indexed: 02/06/2023] Open
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection leads to a severe respiratory illness and alters the gut microbiota, which dynamically interacts with the human immune system. Microbiota alterations include decreased levels of beneficial bacteria and augmentation of opportunistic pathogens. Here, we describe critical factors affecting the microbiota in coronavirus disease 2019 (COVID-19) patients. These include, such as gut microbiota imbalance and gastrointestinal symptoms, the pattern of altered gut microbiota composition in COVID-19 patients, and crosstalk between the microbiome and the gut-lung axis/gut-brain-lung axis. Moreover, we have illustrated the hypoxia state in COVID-19 associated gut microbiota alteration. The role of ACE2 in the digestive system, and control of its expression using the gut microbiota is discussed, highlighting the interactions between the lungs, the gut, and the brain during COVID-19 infection. Similarly, we address the gut microbiota in elderly or co-morbid patients as well as gut microbiota dysbiosis of in severe COVID-19. Several clinical trials to understand the role of probiotics in COVID-19 patients are listed in this review. Augmented inflammation is one of the major driving forces for COVID-19 symptoms and gut microbiome disruption and is associated with disease severity. However, understanding the role of the gut microbiota in immune modulation during SARS-CoV-2 infection may help improve therapeutic strategies for COVID-19 treatment.
Collapse
Affiliation(s)
| | - Ashish Ranjan Sharma
- Institute for Skeletal Aging & Orthopaedic Surgery, Hallym University, Chuncheon-si 24252, South Korea
| | | | - Kuldeep Dhama
- Division of Pathology, Indian Council of Agricultural Research (ICAR)-Indian Veterinary Research Institute (IVRI), Bareilly 243122, Uttar Pradesh, India
| | - Sang-Soo Lee
- Institute for Skeletal Aging & Orthopedic Surgery, Hallym University, Chuncheon-si 24252, South Korea
| |
Collapse
|
28
|
Nayebi A, Navashenaq JG, Soleimani D, Nachvak SM. Probiotic supplementation: A prospective approach in the treatment of COVID-19. Nutr Health 2022; 28:163-175. [PMID: 34747257 PMCID: PMC9160438 DOI: 10.1177/02601060211049631] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Background: Despite strategies based on social distancing, the coronavirus disease 2019 (COVID-19) expands globally, and so far, many attempts have been made to achieve effective treatment for patients with COVID-19. This disease infects the lower respiratory tract and may lead to severe acute respiratory syndrome coronavirus (SARS-CoV). COVID-19 also can cause gastrointestinal infections. Therefore, COVID-19 patients with gastrointestinal symptoms are more likely to be complicated by SARS-CoV. In this disease, acquired immune responses are impaired, and uncontrolled inflammatory responses result in cytokine storms, leading to acute lung injury and thrombus formation. Probiotics are living microorganisms that contribute to the health of the host if administered in appropriate doses. Aim: This study aimed to provide evidence to show the importance of gut dysbiosis in viral disease, especially COVID-19. Therefore, we have focused on the impact of probiotics consumption on preventing severe symptoms of the disease. Methods: We have entirely searched SCOPUS, PubMed, and Google Scholar databases to collect evidence regarding the relationship between probiotics and viral infections to expand this relationship to the COVID-19. Results: It has been shown that probiotics directly counteract SARS-CoV in the gastrointestinal and respiratory tracts. Moreover, probiotics suppress severe immune responses and prevent cytokine storms to inhibit pathologic inflammatory conditions in the body via modulation of immune responses. Conclusion: According to available evidence based on their antiviral and respiratory activities, using probiotics might be an adjuvant therapy to reduce the burden and severity of this disease.
Collapse
Affiliation(s)
- Atiyeh Nayebi
- Student Research Committee, Nutritional Sciences Department, School of Nutrition Sciences and Food Technology, Kermanshah University of Medical Sciences, Kermanshah, Iran
- Nutritional Sciences Department, School of Nutrition Sciences and Food Technology, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | | | - Davood Soleimani
- Student Research Committee, Nutritional Sciences Department, School of Nutrition Sciences and Food Technology, Kermanshah University of Medical Sciences, Kermanshah, Iran
- Research Center of Oils and Fats, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Seyyed Mostafa Nachvak
- Student Research Committee, Nutritional Sciences Department, School of Nutrition Sciences and Food Technology, Kermanshah University of Medical Sciences, Kermanshah, Iran
- Nutritional Sciences Department, School of Nutrition Sciences and Food Technology, Kermanshah University of Medical Sciences, Kermanshah, Iran
| |
Collapse
|
29
|
Shupp B, Mehta SV, Chirayath S, Patel N, Aiad M, Sapin J, Stoltzfus J, Schneider Y. Proton pump inhibitor therapy usage and associated hospitalization rates and critical care outcomes of COVID-19 patients. Sci Rep 2022; 12:7596. [PMID: 35534666 PMCID: PMC9084256 DOI: 10.1038/s41598-022-11680-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 04/12/2022] [Indexed: 11/17/2022] Open
Abstract
Proton Pump Inhibitors (PPI) are one of the most prescribed medications in the United States. However, PPIs have been shown to increase the risk of enteric infections. Our study aims to evaluate the correlation between PPI and COVID-19 severity. We performed a retrospective cohort study on patients who tested positive for SARS-CoV-2 from March to August 2020. Patients were categorized based on PPI user status. Primary outcomes included need for hospital or ICU admission and 30-day mortality. Secondary outcomes looked to determine the severity of COVID-19 infection and effect of comorbid conditions. 2,594 patients were reviewed. The primary outcomes of our study found that neither active nor past PPI use was associated with increased hospital admission or 30-day mortality following completion of multivariate analysis. Additionally, there was no association between COVID-19 infection and the strength of PPI dosing (low, standard, high). However, the following covariates were independently and significantly associated with increased admission: age, male gender, diabetes, COPD, composite cardiovascular disease, kidney disease, and obesity. The following covariates were associated with increased mortality: age, male gender, COPD, and kidney disease. In conclusion, the high risk features and comorbidities of PPI users were found to have a stronger correlation to severe COVID-19 infection and poor outcomes as opposed to the use of PPI therapy.
Collapse
Affiliation(s)
- Brittney Shupp
- Internal Medicine Resident, St. Luke's University Health Network, Bethlehem, PA, USA. .,Division of Internal Medicine, St. Luke's University Health Network, 801 Ostrum St., Suite 201, Bethlehem, 18015, PA, USA.
| | - Sagar V Mehta
- Department of Gastroenterology, St. Luke's University Health Network, Bethlehem, PA, USA
| | - Subin Chirayath
- Internal Medicine Resident, St. Luke's University Health Network, Bethlehem, PA, USA
| | - Nishit Patel
- Internal Medicine Resident, St. Luke's University Health Network, Easton, PA, USA
| | - Mina Aiad
- Internal Medicine Resident, St. Luke's University Health Network, Bethlehem, PA, USA
| | - Jared Sapin
- Lewis Katz School of Medicine at Temple University - St. Luke's Campus, Bethlehem, PA, USA
| | - Jill Stoltzfus
- St. Luke's University Health Network, Bethlehem, PA, USA
| | - Yecheskel Schneider
- Department of Gastroenterology, St. Luke's University Health Network, Bethlehem, PA, USA
| |
Collapse
|
30
|
Sansotta N, Norsa L, D'Antiga L. Gastrointestinal coronavirus disease 2019 manifestations in childhood. Curr Opin Clin Nutr Metab Care 2022; 25:195-202. [PMID: 35199658 DOI: 10.1097/mco.0000000000000825] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
PURPOSE OF THE REVIEW The pandemic of coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has emerged and caused a massive global health crisis. The aim of this review is first, to provide the latest evidence on what is known about the pathophysiology and the transmission of SARS-CoV-2 and then to focus on the manifestations of the gastrointestinal (GI) tract in children with COVID-19. Lastly, we summarise the impact of COVID-19 on patients with preexisting GI diseases. RECENT FINDINGS Even though the virus is mostly transmitted from human to human via respiratory droplets, ACE2 is known to be expressed throughout the GI tract, and SARS-CoV-2 ribonucleic acid has been isolated from patients' stools. GI symptoms including abdominal pain, diarrhoea and vomiting are frequently reported in paediatric patients. Interestingly, a small number of patients seem to exhibit solely GI symptoms. In addition, a multisystem inflammatory syndrome in children (MIS-C) related to SARS-COV-2 described in children, has a high rate of GI involvement. Several etiopathogenetic mechanisms have been postulated to explain the GI involvement of COVID-19. SUMMARY Clinicians should not underestimate or disregard these early or mild GI symptoms, because the patients may be infected and transmit the virus, or develop a more severe condition such as MIS-C.
Collapse
Affiliation(s)
- Naire Sansotta
- Paediatric Hepatology Gastroenterology and Transplantation, Papa Giovanni XXIII Hospital, Bergamo, Italy
| | | | | |
Collapse
|
31
|
Ajmera K, Bansal R, Wilkinson H, Goyal L. Gastrointestinal Complications of COVID-19 Vaccines. Cureus 2022; 14:e24070. [PMID: 35573556 PMCID: PMC9097558 DOI: 10.7759/cureus.24070] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/12/2022] [Indexed: 11/07/2022] Open
Abstract
Much of the control over the coronavirus disease 2019 (COVID-19) pandemic has been achieved by mass vaccination against the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the etiologic agent that causes COVID-19. The COVID-19 mRNA (messenger RNA) vaccines are relatively newly approved and have been widely used in the US since they first became available. However, with passing time, data regarding adverse events associated with the mRNA vaccines have become clearer. Vaccines are safe in general, and the benefits outweigh the risks of adverse events. In this case report, we present the first documented case report of post-vaccination acute diverticulitis and colon micro-perforation following Moderna booster dose (Moderna Inc, Cambridge, USA) in a young adult. Vaccine recipients should be educated on vaccine-associated gastrointestinal (GI) adverse events in order to reduce morbidity and mortality. We also recommend that vaccine recipients with pre-existing GI disorders should be carefully monitored for the worsening of pre-existing conditions post-COVID-19 vaccination.
Collapse
Affiliation(s)
- Kunal Ajmera
- Hospital Medicine, Calvert Health Medical Center, Prince Frederick, USA
| | - Rashika Bansal
- Endocrinology, Diabetes and Metabolism, Cleveland Clinic, Cleveland, USA
| | | | - Lokesh Goyal
- Hospital Medicine, Christus Spohn, Corpus Christi, USA
| |
Collapse
|
32
|
Ambrose PA, Goodman WA. Impact of COVID-19 on Patients with Inflammatory Bowel Disease. JOURNAL OF EXPLORATORY RESEARCH IN PHARMACOLOGY 2022; 7:37-44. [PMID: 35966234 PMCID: PMC9373928 DOI: 10.14218/jerp.2021.00014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) was first identified in Wuhan, China, in late 2019. Responsible for the ongoing coronavirus disease 2019 (COVID-19) pandemic, SARS-CoV-2 is one of three structurally similar beta-coronaviruses that can cause a strong upregulation of cytokines referred to as cytokine release syndrome (CRS). Unresolved CRS leads to respiratory symptoms, including pneumonia, and in more severe cases, acute respiratory distress syndrome (ARDS). Although COVID-19 is widely known for these hallmark respiratory symptoms, it also impacts the gut, causing gastrointestinal (GI) tract inflammation and diarrhea. COVID-19's GI symptoms may be due to the high intestinal expression of angiotensin converting enzyme-2 receptors, which are for the binding of SARS-CoV-2 viral particles. Reports have shown that SARS-CoV-2 can be passed through fecal matter, with one study finding that 48.1% of COVID-19 patients expressed viral SARS-CoV-2 mRNA in their stool. Given that the GI tract is a target tissue affected by COVID-19, this causes concern for those with underlying GI pathologies, such as inflammatory bowel disease (IBD). Regrettably, there have been only limited studies on the impact of COVID-19 on gut health, and the impact of COVID-19 on intestinal inflammation among IBD patients remains unclear. In particular, questions regarding susceptibility to SARS-CoV-2 infection, clinical impact of COVID-19 on IBD, and the potential influence of age, sex, and immunosuppressant medications are still poorly understood. An improved understanding of these issues is needed to address the unique risks of COVID-19 among IBD patients, as well as the potential impact of SARS-CoV-2 on the host intestinal microbiota.
Collapse
Affiliation(s)
- Paula A. Ambrose
- Department of Pathology, Case Western Reserve University School of Medicine, OH, USA
| | - Wendy A. Goodman
- Department of Pathology, Case Western Reserve University School of Medicine, OH, USA
| |
Collapse
|
33
|
Liu Q, Mak JWY, Su Q, Yeoh YK, Lui GCY, Ng SSS, Zhang F, Li AYL, Lu W, Hui DSC, Chan PK, Chan FKL, Ng SC. Gut microbiota dynamics in a prospective cohort of patients with post-acute COVID-19 syndrome. Gut 2022; 71:544-552. [PMID: 35082169 PMCID: PMC8814432 DOI: 10.1136/gutjnl-2021-325989] [Citation(s) in RCA: 334] [Impact Index Per Article: 111.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Accepted: 11/06/2021] [Indexed: 12/12/2022]
Abstract
BACKGROUND Long-term complications after COVID-19 are common, but the potential cause for persistent symptoms after viral clearance remains unclear. OBJECTIVE To investigate whether gut microbiome composition is linked to post-acute COVID-19 syndrome (PACS), defined as at least one persistent symptom 4 weeks after clearance of the SARS-CoV-2 virus. METHODS We conducted a prospective study of 106 patients with a spectrum of COVID-19 severity followed up from admission to 6 months and 68 non-COVID-19 controls. We analysed serial faecal microbiome of 258 samples using shotgun metagenomic sequencing, and correlated the results with persistent symptoms at 6 months. RESULTS At 6 months, 76% of patients had PACS and the most common symptoms were fatigue, poor memory and hair loss. Gut microbiota composition at admission was associated with occurrence of PACS. Patients without PACS showed recovered gut microbiome profile at 6 months comparable to that of non-COVID-19 controls. Gut microbiome of patients with PACS were characterised by higher levels of Ruminococcus gnavus, Bacteroides vulgatus and lower levels of Faecalibacterium prausnitzii. Persistent respiratory symptoms were correlated with opportunistic gut pathogens, and neuropsychiatric symptoms and fatigue were correlated with nosocomial gut pathogens, including Clostridium innocuum and Actinomyces naeslundii (all p<0.05). Butyrate-producing bacteria, including Bifidobacterium pseudocatenulatum and Faecalibacterium prausnitzii showed the largest inverse correlations with PACS at 6 months. CONCLUSION These findings provided observational evidence of compositional alterations of gut microbiome in patients with long-term complications of COVID-19. Further studies should investigate whether microbiota modulation can facilitate timely recovery from post-acute COVID-19 syndrome.
Collapse
Affiliation(s)
- Qin Liu
- Center for Gut Microbiota Research, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
- State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Science, Institute of Digestive Disease, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Microbiota I-Center (MagIC), Hong Kong, Hong Kong SAR, China
| | - Joyce Wing Yan Mak
- Center for Gut Microbiota Research, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
- State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Science, Institute of Digestive Disease, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Qi Su
- Center for Gut Microbiota Research, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
- State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Science, Institute of Digestive Disease, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Microbiota I-Center (MagIC), Hong Kong, Hong Kong SAR, China
| | - Yun Kit Yeoh
- Center for Gut Microbiota Research, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Microbiota I-Center (MagIC), Hong Kong, Hong Kong SAR, China
- Department of Microbiology, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Grace Chung-Yan Lui
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Stanley Ho Centre for Emerging Infectious Diseases, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Susanna So Shan Ng
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Fen Zhang
- Center for Gut Microbiota Research, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
- State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Science, Institute of Digestive Disease, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Microbiota I-Center (MagIC), Hong Kong, Hong Kong SAR, China
| | - Amy Y L Li
- Center for Gut Microbiota Research, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
- State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Science, Institute of Digestive Disease, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Wenqi Lu
- Center for Gut Microbiota Research, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
- State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Science, Institute of Digestive Disease, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Microbiota I-Center (MagIC), Hong Kong, Hong Kong SAR, China
| | - David Shu-Cheong Hui
- Stanley Ho Centre for Emerging Infectious Diseases, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Paul Ks Chan
- Center for Gut Microbiota Research, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Department of Microbiology, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Francis K L Chan
- Center for Gut Microbiota Research, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
- State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Science, Institute of Digestive Disease, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Microbiota I-Center (MagIC), Hong Kong, Hong Kong SAR, China
| | - Siew C Ng
- Center for Gut Microbiota Research, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
- State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Science, Institute of Digestive Disease, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Microbiota I-Center (MagIC), Hong Kong, Hong Kong SAR, China
| |
Collapse
|
34
|
Cruz VB, Júnior LFFF, Kobal CR, da Silva NA. Does sensitization by SARS-CoV-2 immune complexes trigger DRESS syndrome? Braz J Infect Dis 2022; 26:102337. [PMID: 35276095 PMCID: PMC8882399 DOI: 10.1016/j.bjid.2022.102337] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Revised: 01/18/2022] [Accepted: 02/16/2022] [Indexed: 12/17/2022] Open
Abstract
The diagnosis of coronavirus disease (COVID-19) has been a great challenge since the infection affects not only the respiratory system, but also different organs, given the intense inflammatory and autoimmune reaction triggered by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Herein we present a case of a 36-year-old male patient, with some comorbidities and previous use of carbamazepine, who developed a severe condition triggered by COVID-19, including extensive exfoliative erythroderma and severe impairment of liver function, which lasted approximately 80 days.
Collapse
Affiliation(s)
- Virgínia Barbeitos Cruz
- Health Sciences Program, School of Medicine, Universidade Federal de Goiás, Goiânia, GO, Brazil.
| | | | - Christiane Reis Kobal
- Department of Infectious Diseases, Hospital of Tropical Diseases of Goiás, Goiânia, GO, Brazil
| | - Nilzio Antonio da Silva
- Health Sciences Program, School of Medicine, Universidade Federal de Goiás, Goiânia, GO, Brazil
| |
Collapse
|
35
|
Kerslake R, Randeva HS, Jonigk D, Werlein C, Robertus JL, Katopodis P, Jasker P, Spandidos DA, Kyrou I, Karteris E. Protein expression of transmembrane protease serine 4 in the gastrointestinal tract and in healthy, cancer, and SARS‑CoV‑2 infected lungs. Mol Med Rep 2022; 25:138. [PMID: 35211765 PMCID: PMC8908323 DOI: 10.3892/mmr.2022.12654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2021] [Accepted: 02/01/2022] [Indexed: 01/08/2023] Open
Abstract
In addition to the angiotensin-converting enzyme 2 (ACE2), a number of host cell entry mediators have been identified for severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), including transmembrane protease serine 4 (TMPRSS4). The authors have recently demonstrated the upregulation of TMPRSS4 in 11 different cancers, as well as its specific expression within the central nervous system using in silico tools. The present study aimed to expand the initial observations and, using immunohistochemistry, TMPRSS4 protein expression in the gastrointestinal (GI) tract and lungs was further mapped. Immunohistochemistry was performed on tissue arrays and lung tissues of patients with non-small cell lung cancer with concurrent coronavirus disease 2019 (COVID-19) infection using TMPRSS4 antibody. The results revealed that TMPRSS4 was abundantly expressed in the oesophagus, stomach, small intestine, jejunum, ileum, colon, liver and pancreas. Moreover, the extensive TMPRSS4 protein expression in the lungs of a deceased patient with COVID-19 with chronic obstructive pulmonary disease and bronchial carcinoma, as well in the adjacent normal tissue, was demonstrated for the first time, at least to the best of our knowledge. On the whole, the immunohistochemistry data of the present study suggest that TMPRSS4 may be implicated in the broader (pulmonary and extra-pulmonary) COVID-19 symptomatology; thus, it may be responsible for the tropism of this coronavirus both in the GI tract and lungs.
Collapse
Affiliation(s)
- Rachel Kerslake
- Division of Biosciences, College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge UB8 3PH
| | - Harpal S Randeva
- Warwickshire Institute for The Study of Diabetes, Endocrinology and Metabolism (WISDEM), University Hospitals Coventry and Warwickshire (UHCW) NHS Trust, Coventry CV2 2DX, UK
| | - Danny Jonigk
- Institute for Pathology, Hannover Medical School, D‑30625 Hannover, Germany
| | | | - Jan L Robertus
- National Heart and Lung Institute, Imperial College London, London SW3 6LY, UK
| | - Periklis Katopodis
- Division of Biosciences, College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge UB8 3PH, UK
| | - Petra Jasker
- Pathologie Grünstrasse, D‑40212 Düsseldorf, Germany
| | - Demetrios A Spandidos
- Laboratory of Clinical Virology, Medical School, University of Crete, 71409 Heraklion, Greece
| | - Ioannis Kyrou
- Warwickshire Institute for The Study of Diabetes, Endocrinology and Metabolism (WISDEM), University Hospitals Coventry and Warwickshire (UHCW) NHS Trust, Coventry CV2 2DX, UK
| | - Emmanouil Karteris
- Division of Biosciences, College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge UB8 3PH, UK
| |
Collapse
|
36
|
Gray-Rodriguez S, Jensen MP, Otero-Jimenez M, Hanley B, Swann OC, Ward PA, Salguero FJ, Querido N, Farkas I, Velentza-Almpani E, Weir J, Barclay WS, Carroll MW, Jaunmuktane Z, Brandner S, Pohl U, Allinson K, Thom M, Troakes C, Al-Sarraj S, Sastre M, Gveric D, Gentleman S, Roufosse C, Osborn M, Alegre-Abarrategui J. Multisystem screening reveals SARS-CoV-2 in neurons of the myenteric plexus and in megakaryocytes. J Pathol 2022; 257:198-217. [PMID: 35107828 PMCID: PMC9325073 DOI: 10.1002/path.5878] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 12/12/2021] [Accepted: 01/29/2022] [Indexed: 11/30/2022]
Abstract
SARS‐CoV‐2, the causative agent of COVID‐19, typically manifests as a respiratory illness, although extrapulmonary involvement, such as in the gastrointestinal tract and nervous system, as well as frequent thrombotic events, are increasingly recognised. How this maps onto SARS‐CoV‐2 organ tropism at the histological level, however, remains unclear. Here, we perform a comprehensive validation of a monoclonal antibody against the SARS‐CoV‐2 nucleocapsid protein (NP) followed by systematic multisystem organ immunohistochemistry analysis of the viral cellular tropism in tissue from 36 patients, 16 postmortem cases and 16 biopsies with polymerase chain reaction (PCR)‐confirmed SARS‐CoV‐2 status from the peaks of the pandemic in 2020 and four pre‐COVID postmortem controls. SARS‐CoV‐2 anti‐NP staining in the postmortem cases revealed broad multiorgan involvement of the respiratory, digestive, haematopoietic, genitourinary and nervous systems, with a typical pattern of staining characterised by punctate paranuclear and apical cytoplasmic labelling. The average time from symptom onset to time of death was shorter in positively versus negatively stained postmortem cases (mean = 10.3 days versus mean = 20.3 days, p = 0.0416, with no cases showing definitive staining if the interval exceeded 15 days). One striking finding was the widespread presence of SARS‐CoV‐2 NP in neurons of the myenteric plexus, a site of high ACE2 expression, the entry receptor for SARS‐CoV‐2, and one of the earliest affected cells in Parkinson's disease. In the bone marrow, we observed viral SARS‐CoV‐2 NP within megakaryocytes, key cells in platelet production and thrombus formation. In 15 tracheal biopsies performed in patients requiring ventilation, there was a near complete concordance between immunohistochemistry and PCR swab results. Going forward, our findings have relevance to correlating clinical symptoms with the organ tropism of SARS‐CoV‐2 in contemporary cases as well as providing insights into potential long‐term complications of COVID‐19. © 2022 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Sandra Gray-Rodriguez
- Department of Brain Sciences, Imperial College London, Hammersmith Hospital, London, W12 0NN, UK
| | - Melanie P Jensen
- Department of Cellular Pathology, Northwest London Pathology, Charing Cross Hospital Campus, London, UK
| | - Maria Otero-Jimenez
- Department of Brain Sciences, Imperial College London, Hammersmith Hospital, London, W12 0NN, UK
| | - Brian Hanley
- Department of Cellular Pathology, Northwest London Pathology, Charing Cross Hospital Campus, London, UK.,Department of Immunology and Inflammation, Imperial College London, London, W12 0NN, UK
| | - Olivia C Swann
- Department of Infectious Disease, Imperial College London, London, UK
| | - Patrick A Ward
- Chelsea and Westminster NHS Foundation Trust, London, UK
| | - Francisco J Salguero
- National Infection Service, United Kingdom Health Security Agency, Porton Down, Salisbury, UK
| | - Nadira Querido
- Department of Brain Sciences, Imperial College London, Hammersmith Hospital, London, W12 0NN, UK
| | - Ildiko Farkas
- Department of Brain Sciences, Imperial College London, Hammersmith Hospital, London, W12 0NN, UK
| | | | - Justin Weir
- Department of Cellular Pathology, Northwest London Pathology, Charing Cross Hospital Campus, London, UK
| | - Wendy S Barclay
- Department of Infectious Disease, Imperial College London, London, UK
| | - Miles W Carroll
- National Infection Service, United Kingdom Health Security Agency, Porton Down, Salisbury, UK.,Pandemic Sciences Centre, Nuffield Department of Medicine, Oxford University, OX3 7BN, UK
| | - Zane Jaunmuktane
- Department of Neuropathology, UCL Queen Square Institute of Neurology, London, UK
| | - Sebastian Brandner
- Department of Neuropathology, UCL Queen Square Institute of Neurology, London, UK
| | - Ute Pohl
- Department of Cellular Pathology, Queen Elizabeth Hospital Birmingham/University Hospitals Birmingham, Birmingham, UK
| | - Kieren Allinson
- Department of Neuropathology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Maria Thom
- Department of Neuropathology, UCL Queen Square Institute of Neurology, London, UK
| | - Claire Troakes
- Basic and Clinical Neuroscience Department, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - Safa Al-Sarraj
- Basic and Clinical Neuroscience Department, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - Magdalena Sastre
- Department of Brain Sciences, Imperial College London, Hammersmith Hospital, London, W12 0NN, UK
| | - Djordje Gveric
- Multiple Sclerosis and Parkinson's Tissue Bank, Imperial College London, Hammersmith Hospital, London, W12 0NN, UK
| | - Steve Gentleman
- Department of Brain Sciences, Imperial College London, Hammersmith Hospital, London, W12 0NN, UK.,Multiple Sclerosis and Parkinson's Tissue Bank, Imperial College London, Hammersmith Hospital, London, W12 0NN, UK
| | - Candice Roufosse
- Department of Cellular Pathology, Northwest London Pathology, Charing Cross Hospital Campus, London, UK.,Department of Immunology and Inflammation, Imperial College London, London, W12 0NN, UK
| | - Michael Osborn
- Department of Cellular Pathology, Northwest London Pathology, Charing Cross Hospital Campus, London, UK
| | - Javier Alegre-Abarrategui
- Department of Brain Sciences, Imperial College London, Hammersmith Hospital, London, W12 0NN, UK.,Department of Cellular Pathology, Northwest London Pathology, Charing Cross Hospital Campus, London, UK.,Multiple Sclerosis and Parkinson's Tissue Bank, Imperial College London, Hammersmith Hospital, London, W12 0NN, UK
| |
Collapse
|
37
|
Ghani S, Kalantari S, Mirmotalebisohi SA, Sameni M, Poursheykhi H, Dadashkhan S, Abbasi M, Zali H. Specific Regulatory Motifs Network in SARS-CoV-2-Infected Caco-2 Cell Line, as a Model of Gastrointestinal Infections. Cell Reprogram 2022; 24:26-37. [PMID: 35100036 DOI: 10.1089/cell.2021.0055] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) was primarily noted as a respiratory pathogen, but later clinical reports highlighted its extrapulmonary effects particularly on the gastrointestinal (GI) tract. The aim of the current study was the prediction of crucial genes associated with the regulatory network motifs, probably responsible for the SARS-CoV-2 effects on the GI tract. The data were obtained from a published study on the effect of SARS-CoV-2 on the Caco-2 (colon carcinoma) cell line. We used transcription factors-microRNA-gene interaction databases to find the key regulatory molecules, then analyzed the data using the FANMOD software for detection of the crucial regulatory motifs. Cytoscape software was then used to construct and analyze the regulatory network of these motifs and identify their crucial genes. Finally, GEPIA2 (Gene Expression Profiling Interactive Analysis 2) and UALCAN datasets were used to evaluate the possible relationship between crucial genes and colon cancer development. Using bioinformatics tools, we demonstrated one 3edge feed-forward loop motifs and recognized 10 crucial genes in relationship with Caco-2 cell infected by SARS-CoV-2, including SP1, TSC22D2, POU2F1, REST, NFIC, CHD7, E2F1, CEBPA, TCF7L2, and TSC22D1. The box plot analysis indicated the significant overexpression of CEBPA in colon cancer compared to normal colon tissues, while it was in contrast with the results of stage plot. However, the overall survival analysis indicated that high expression of CEBPA has positive effect on colon cancer patient survivability, verifying the results of CEBPA stage plot. We predict that the SARS-CoV-2 GI infections may cause a serious risk in colon cancer patients. However, further experimental studies are required.
Collapse
Affiliation(s)
- Sepideh Ghani
- Department of Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Cellular and Molecular Biology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sima Kalantari
- Department of Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Cellular and Molecular Biology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Regenerative Medicine Group (REMED), Universal Scientific Education & Research Network (USERN), Tehran, Iran
| | - Seyed Amir Mirmotalebisohi
- Department of Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Cellular and Molecular Biology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Marzieh Sameni
- Department of Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Cellular and Molecular Biology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hossein Poursheykhi
- Department of New Scientist, Faculty of Medical Sciences, Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Sadaf Dadashkhan
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | | | - Hakimeh Zali
- Proteomics Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
38
|
Spigaglia P. Clostridioides difficile infection (CDI) during the COVID-19 pandemic. Anaerobe 2022; 74:102518. [PMID: 35063599 PMCID: PMC8767936 DOI: 10.1016/j.anaerobe.2022.102518] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 01/08/2022] [Accepted: 01/14/2022] [Indexed: 02/07/2023]
Abstract
The ongoing coronavirus disease (COVID-19) pandemic has dramatically tested healthcare systems around the world, with serious repercussions on the measures of prevention and control of hospital-acquired infections (HAIs). Among HAIs, Clostridioides difficile infection (CDI) represents one of the most important global public health threats. Although the full impact of the COVID-19 pandemic on CDI remains undetermined, depending on the development of the pandemic in the coming months, in this review literature studies of the last three years have been considered in order to depict the current situation, and make some considerations about possible future developments. If on the one hand, a general reduction in CDI incidence has been reported in several settings, mainly due to the extraordinary reinforcement of infection prevention measures, on the other hand, the critical circumstances experienced in many hospitals have limited the effectiveness of these measures, particularly in the intensive care units (ICUs), increasing the possibility of the occurrence of hospital-acquired CDI (HA-CDI). New concerns have arisen from the decrease in C. difficile testing and the increased use of broad-spectrum antibiotics reported during the pandemic. In particular, overuse of antibiotics and disinfectants may lead to a selection of resistant C. difficile strains not only in hospitals but also in the community. Furthermore, patients infected with severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) and patients that have survived COVID-19 may represent a new group of frail patients potentially at a higher risk of CDI, a group that could potentially increase in size due to SARS-CoV-2 evolution. In the dramatic COVID-19 era, the multifactorial nature of CDI has emerged more clearly than before, highlighting the necessity of a strong refocus on efforts to improve prevention strategies and to integrate CDI surveillance in a One Health prospective in order to curtail the public health threat posed by this infection in the next future.
Collapse
|
39
|
Tyagi N, Gurian PL, Kumar A. Using QMRA to understand possible exposure risks of SARS-CoV-2 from the water environment. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2022; 29:7240-7253. [PMID: 34467495 PMCID: PMC8408015 DOI: 10.1007/s11356-021-16188-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Accepted: 08/23/2021] [Indexed: 06/13/2023]
Abstract
This study investigated the human risk of infection due to inadvertent ingestion of water during swimming in a river that receives SARS-CoV-2-containing effluent from a wastewater treatment plant (WWTP). A quantitative microbial risk assessment (QMRA) approach was applied for risk estimation using dose-response models (DRM) of different surrogate coronaviruses (SARS-CoV-1, MERS-CoV) and the virus responsible for most infectious respiratory illnesses (i.e., influenza A H5N1) due to the unavailability of DRM for SARS-CoV-2. The ratio of infectious concentration to genomic copies of SARS-CoV-2 is unknown and also unavailable for other coronaviruses. Therefore, literature-based information on enteric viruses was used for formulating the ratio used for QMRA, although it is acknowledged that identifying this information for SARS-CoV-2 is a priority, and in the absence of information specific to SARS-CoV-2, another coronavirus would be a preferable surrogate to the enteric viruses used here. The calculated concentration of ingested SARS-CoV-2 ranged between 4.6 × 10-7 and 80.5 genomic copies/dip (one swim = 32 mL). The risk of infection (> 9 × 10-12 to 5.8 × 10-1) was found to be > 1/10,000 annual risk of infection. Moreover, the study revealed that the risk estimation was largely dependent on the value of the molecular concentration of SARS-CoV-2 (gc/mL). Overall immediate attention is required for obtaining information on the (i) ratio of infectious virus to genomic copies, (ii) DRM for SARS-CoV-2, and (iii) virus reduction rate after treatment in the WWTPs. The QMRA structure used in present findings is helpful in analyzing and prioritizing upcoming health risks due to swimming performed in contaminated rivers during the COVID-19 outbreak.
Collapse
Affiliation(s)
- Neha Tyagi
- Department of Civil Engineering, Indian Institute of Technology Delhi, New Delhi, 110016 India
| | - Patrick L. Gurian
- Department of Civil, Architectural and Environmental Engineering, Drexel University, Philadelphia, PA USA
| | - Arun Kumar
- Department of Civil Engineering, Indian Institute of Technology Delhi, New Delhi, 110016 India
| |
Collapse
|
40
|
Megiorni F, Pontecorvi P, Gerini G, Anastasiadou E, Marchese C, Ceccarelli S. Sex-Related Factors in Cardiovascular Complications Associated to COVID-19. Biomolecules 2021; 12:biom12010021. [PMID: 35053169 PMCID: PMC8773922 DOI: 10.3390/biom12010021] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 12/17/2021] [Accepted: 12/22/2021] [Indexed: 12/15/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19), the pandemic infection caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), presents with an extremely heterogeneous spectrum of symptoms and signs. The clinical manifestations seem to be correlated with disease severity. COVID-19 susceptibility and mortality show a significant sex imbalance, with men being more prone to infection and showing a higher rate of hospitalization and mortality compared to women. Such variability can be ascribed to both sex-related biological factors and gender-related behavioral cues. This review will discuss the potential mechanisms accounting for sex/gender influence in vulnerability to COVID-19. Cardiovascular diseases play a central role in determining COVID-19 outcome, whether they are pre-existent or arose upon infection. We will pay particular attention to the impact of sex and gender on cardiovascular manifestations related to COVID-19. Finally, we will discuss the sex-dependent variability in some biomarkers for the evaluation of COVID-19 infection and prognosis. The aim of this work is to highlight the significance of gendered medicine in setting up personalized programs for COVID-19 prevention, clinical evaluation and treatment.
Collapse
|
41
|
Laboratory manifestations and pathophysiological aspects of coronavirus disease 2019 pandemic: focusing on the digestive system. Eur J Gastroenterol Hepatol 2021; 33:e59-e65. [PMID: 35048645 DOI: 10.1097/meg.0000000000002068] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Since December 2019, the severe acute respiratory syndrome coronavirus 2 has constituted a serious threat to global health. So far, there is little published evidence on the laboratory features of coronavirus disease 2019 (COVID-19). We have reviewed laboratory findings from multiple studies, mostly relating to the digestive system, since the virus outbreak. Laboratory data from older coronaviruses endemics, as well as other RNA viruses, were also reported. Although the main route of transmission is considered to be respiratory droplets, the distribution of ACE2 receptors in the gastrointestinal tract in combination with the detection of the virus in feces may imply a potential fecal-oral transmission route, and thus, emphasis should be given to patients with gastrointestinal symptoms. Interestingly, there is evidence that severe acute respiratory syndrome coronavirus 2 displays similar laboratory and clinical findings with older members of the coronavirus family, and so, comparable diagnostic and therapeutic approaches may be used. Regarding laboratory abnormalities, lymphopenia appears to be the most common finding, together with coagulation disorders and inflammatory markers elevation, reflecting a sustained systemic response. Abnormal liver and, occasionally, pancreatic tests are also common and even more severe in patients with gastrointestinal symptoms or diseases. Thus, the aim of this study is to focus on the laboratory and pathophysiologic side of this novel disease in order to strengthen current knowledge and urge further research. Detailed investigation of numerous studies may suggest a common laboratory pattern between COVID-19 patients. It is important for clinicians not to underestimate patients with gastrointestinal comorbidities, as they have been associated with severe COVID-19 disease.
Collapse
|
42
|
Chia DKA, Lim Z, Ang JJ, Tambyah PA, Lau KSH, Ong J, Chow VTK, Allen DM, Fung J, Lau KJX, Luhung I, Schuster SC, Lee CN, Kim G, So JBY, Lomanto D, Shabbir A. Coronavirus viability in surgical plume and methods for safe disposal: a preclinical model. Br J Surg 2021; 109:15-20. [PMID: 34792098 DOI: 10.1093/bjs/znab385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2021] [Accepted: 09/23/2021] [Indexed: 11/14/2022]
Abstract
Smoke generated by cautery devices used during surgery may contain infective particles and may cause transmission of airborne viruses. This study determines whether live viruses are present in surgical smoke and evaluates the effectiveness of several proposed methods of removal so as to improve safety of healthcare workers.
Collapse
Affiliation(s)
- Daryl K A Chia
- Department of Surgery, University Surgical Cluster, National University Health System, Singapore
| | - Zhixue Lim
- Department of Hand & Reconstructive Microsurgery, University Orthopaedic, Hand & Reconstructive Microsurgical Cluster, National University Health System, Singapore
| | - Jia Jun Ang
- Department of Surgery, University Surgical Cluster, National University Health System, Singapore
| | - Paul A Tambyah
- Division of Infectious Diseases, Department of Medicine, University Medical Cluster, National University Health System, Singapore
| | - Kelly S H Lau
- Infectious Diseases Translational Research Programme, Department of Microbiology & Immunology, Yong Loo Lin School of Medicine, National University of Singapore
| | - Joe Ong
- Infectious Diseases Translational Research Programme, Department of Microbiology & Immunology, Yong Loo Lin School of Medicine, National University of Singapore
| | - Vincent T K Chow
- Infectious Diseases Translational Research Programme, Department of Microbiology & Immunology, Yong Loo Lin School of Medicine, National University of Singapore
| | - David M Allen
- Division of Infectious Diseases, Department of Medicine, University Medical Cluster, National University Health System, Singapore
| | - Javis Fung
- Division of General Surgery (Upper Gastrointestinal Surgery), Department of Surgery, University Surgical Cluster, National University Health System, Singapore
| | - Kenny J X Lau
- Singapore Centre for Environmental Life Sciences Engineering, Nanyang Technological University, Singapore
| | - Irvan Luhung
- Singapore Centre for Environmental Life Sciences Engineering, Nanyang Technological University, Singapore
| | - Stephan C Schuster
- Singapore Centre for Environmental Life Sciences Engineering, Nanyang Technological University, Singapore
| | - Chuen Neng Lee
- Department of Surgery, University Surgical Cluster, National University Health System, Singapore
| | - Guowei Kim
- Department of Surgery, University Surgical Cluster, National University Health System, Singapore.,Division of General Surgery (Upper Gastrointestinal Surgery), Department of Surgery, University Surgical Cluster, National University Health System, Singapore
| | - Jimmy B Y So
- Department of Surgery, University Surgical Cluster, National University Health System, Singapore.,Division of General Surgery (Upper Gastrointestinal Surgery), Department of Surgery, University Surgical Cluster, National University Health System, Singapore.,Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Davide Lomanto
- Department of Surgery, University Surgical Cluster, National University Health System, Singapore.,Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Minimally Invasive Surgical Centre, Department of Surgery, National University Health System, Singapore
| | - Asim Shabbir
- Department of Surgery, University Surgical Cluster, National University Health System, Singapore.,Division of General Surgery (Upper Gastrointestinal Surgery), Department of Surgery, University Surgical Cluster, National University Health System, Singapore.,Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| |
Collapse
|
43
|
Zhou D, Wang Q, Liu H. Coronavirus disease-19 and the gut-lung axis. Int J Infect Dis 2021; 113:300-307. [PMID: 34517046 PMCID: PMC8431834 DOI: 10.1016/j.ijid.2021.09.013] [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: 06/05/2021] [Revised: 08/19/2021] [Accepted: 09/06/2021] [Indexed: 01/08/2023] Open
Abstract
Gastrointestinal and respiratory tract diseases often occur together. There are many overlapping pathologies, leading to the concept of the ‘gut–lung axis’ in which stimulation on one side triggers a response on the other side. This axis appears to be implicated in infections involving severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), which has triggered the global coronavirus disease 2019 (COVID-19) pandemic, in which respiratory symptoms of fever, cough and dyspnoea often occur together with gastrointestinal symptoms such as nausea, vomiting, abdominal pain and diarrhoea. Besides the gut–lung axis, it should be noted that the gut participates in numerous axes which may affect lung function, and consequently the severity of COVID-19, through several pathways. This article focuses on the latest evidence and the mechanisms that drive the operation of the gut–lung axis, and discusses the interaction between the gut–lung axis and its possible involvement in COVID-19 from the perspective of microbiota, microbiota metabolites, microbial dysbiosis, common mucosal immunity and angiotensin-converting enzyme II, raising hypotheses and providing methods to guide future research on this new disease and its treatments.
Collapse
Affiliation(s)
- Dan Zhou
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China; Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education
| | - Qiu Wang
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education; Department of Rehabilitation Medicine, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Hanmin Liu
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China; Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education.
| |
Collapse
|
44
|
Umar Y, Al-Batty S, Rahman H, Ashwaq O, Sarief A, Sadique Z, Sreekumar PA, Haque SKM. Polymeric Materials as Potential Inhibitors Against SARS-CoV-2. JOURNAL OF POLYMERS AND THE ENVIRONMENT 2021; 30:1244-1263. [PMID: 34518763 PMCID: PMC8426594 DOI: 10.1007/s10924-021-02272-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 08/22/2021] [Indexed: 05/02/2023]
Abstract
Recently discovered SARS-CoV-2 caused a pandemic that triggered researchers worldwide to focus their research on all aspects of this new peril to humanity. However, in the absence of specific therapeutic intervention, some preventive strategies and supportive treatment minimize the viral transmission as studied by some factors such as basic reproduction number, case fatality rate, and incubation period in the epidemiology of viral diseases. This review briefly discusses coronaviruses' life cycle of SARS-CoV-2 in a human host cell and preventive strategies at some selected source of infection. The antiviral activities of synthetic and natural polymers such as chitosan, hydrophobically modified chitosan, galactosylated chitosan, amine-based dendrimers, cyclodextrin, carrageenans, polyethyleneimine, nanoparticles are highlighted in this article. Mechanism of virus inhibition, detection and diagnosis are also presented. It also suggests that polymeric materials and nanoparticles can be effective as potential inhibitors and immunization against coronaviruses which would further develop new technologies in the field of polymer and nanoscience.
Collapse
Affiliation(s)
- Yunusa Umar
- Department of Chemical and Process Engineering Technology, Jubail Industrial College, Jubail Industrial City, 31961 Saudi Arabia
| | - Sirhan Al-Batty
- Department of Chemical and Process Engineering Technology, Jubail Industrial College, Jubail Industrial City, 31961 Saudi Arabia
| | - Habibur Rahman
- Department of General Studies, Jubail Industrial College, Jubail Industrial City, 31961 Saudi Arabia
| | - Omar Ashwaq
- Department of Chemical and Process Engineering Technology, Jubail Industrial College, Jubail Industrial City, 31961 Saudi Arabia
| | - Abdulla Sarief
- Department of Chemical and Process Engineering Technology, Jubail Industrial College, Jubail Industrial City, 31961 Saudi Arabia
| | - Zakariya Sadique
- Department of Chemical and Process Engineering Technology, Jubail Industrial College, Jubail Industrial City, 31961 Saudi Arabia
| | - P. A. Sreekumar
- Department of Chemical and Process Engineering Technology, Jubail Industrial College, Jubail Industrial City, 31961 Saudi Arabia
| | - S. K. Manirul Haque
- Department of Chemical and Process Engineering Technology, Jubail Industrial College, Jubail Industrial City, 31961 Saudi Arabia
| |
Collapse
|
45
|
Pandanaboyana S. Exploring Koch's postulate for SARS-CoV-2-induced acute pancreatitis: is it all about the ACE? Br J Surg 2021; 108:879-881. [PMID: 34227650 PMCID: PMC8344617 DOI: 10.1093/bjs/znab178] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Accepted: 04/23/2021] [Indexed: 01/08/2023]
Affiliation(s)
- S Pandanaboyana
- Hepatopancreatobiliary and Transplant Unit, Freeman Hospital, Newcastle upon Tyne, UK.,Population Health Sciences Institute, Newcastle University, Newcastle upon Tyne, UK
| |
Collapse
|
46
|
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
|
47
|
Ong JS, Gharahkhani P, Vaughan TL, Whiteman D, Kendall BJ, MacGregor S. Assessing the genetic relationship between gastro-esophageal reflux disease and risk of COVID-19 infection. Hum Mol Genet 2021; 31:471-480. [PMID: 34553760 PMCID: PMC8522419 DOI: 10.1093/hmg/ddab253] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Revised: 07/29/2021] [Accepted: 08/26/2021] [Indexed: 12/11/2022] Open
Abstract
Background Symptoms related with Gastro-esophageal reflux disease (GERD) were previously shown to be linked with increased risk for the 2019 coronavirus disease (COVID-19). We aim to interrogate the possibility of a shared genetic basis between GERD and COVID-19 outcomes. Methods Using published GWAS data for GERD (78 707 cases; 288 734 controls) and COVID-19 susceptibility (up to 32 494 cases; 1.5 million controls), we examined the genetic relationship between GERD and three COVID-19 outcomes: risk of developing severe COVID-19, COVID-19 hospitalization and overall COVID-19 risk. We estimated the genetic correlation between GERD and COVID-19 outcomes followed by Mendelian randomization (MR) analyses to assess genetic causality. Conditional analyses were conducted to examine whether known COVID-19 risk factors (obesity, smoking, type-II diabetes, coronary artery disease) can explain the relationship between GERD and COVID-19. Results We found small to moderate genetic correlations between GERD and COVID-19 outcomes (rg between 0.06–0.24). MR analyses revealed a OR of 1.15 (95% CI: 0.96–1.39) for severe COVID-19; 1.16 (1.01–1.34) for risk of COVID-19 hospitalization; 1.05 (0.97–1.13) for overall risk of COVID-19 per doubling of odds in developing GERD. The genetic correlation/associations between GERD and COVID-19 showed mild attenuation towards the null when obesity and smoking was adjusted for. Conclusions Susceptibility for GERD and risk of COVID-19 hospitalization were genetically correlated, with MR findings supporting a potential causal role between the two. The genetic association between GERD and COVID-19 was partially attenuated when obesity is accounted for, consistent with obesity being a major risk factor for both diseases.
Collapse
Affiliation(s)
- Jue-Sheng Ong
- Department of Genetics and Computational Biology, QIMR Berghofer Medical Research Institute, 300 Herston Road, Herston, QLD 4006, Brisbane, Australia
| | - Puya Gharahkhani
- Department of Genetics and Computational Biology, QIMR Berghofer Medical Research Institute, 300 Herston Road, Herston, QLD 4006, Brisbane, Australia
| | - Thomas L Vaughan
- Department of Epidemiology, School of Public Health, University of Washington, Seattle, WA, USA
| | - David Whiteman
- Department of Population Health, QIMR Berghofer Medical Research Institute, 300 Herston Road, Herston, QLD 4006, Brisbane, Australia
| | - Bradley J Kendall
- Department of Medicine, The University of Queensland, , Herston, QLD 4006, Brisbane, Australia.,Department of Gastroenterology and Hepatology, Princess Alexandra Hospital, Woolloongabba, QLD 4102, Brisbane, Australia
| | - Stuart MacGregor
- Department of Genetics and Computational Biology, QIMR Berghofer Medical Research Institute, 300 Herston Road, Herston, QLD 4006, Brisbane, Australia
| |
Collapse
|
48
|
Chen J, Hall S, Vitetta L. Altered gut microbial metabolites could mediate the effects of risk factors in Covid-19. Rev Med Virol 2021; 31:1-13. [PMID: 34546607 PMCID: PMC7995004 DOI: 10.1002/rmv.2211] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 12/08/2020] [Accepted: 12/09/2020] [Indexed: 01/08/2023]
Abstract
Coronavirus disease 2019 (Covid-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, is now pandemic. While most Covid-19 patients will experience mild symptoms, a small proportion will develop severe disease, which could be fatal. Clinically, Covid-19 patients manifest fever with dry cough, fatigue and dyspnoea, and in severe cases develop into acute respiratory distress syndrome (ARDS), sepsis and multi-organ failure. These severe patients are characterized by hyperinflammation with highly increased pro-inflammatory cytokines including IL-6, IL-17 and TNF-alpha as well as C-reactive protein, which are accompanied by decreased lymphocyte counts. Clinical evidence supports that gut microbiota dysregulation is common in Covid-19 and plays a key role in the pathogenesis of Covid-19. In this narrative review, we summarize the roles of intestinal dysbiosis in Covid-19 pathogenesis and posit that the associated mechanisms are being mediated by gut bacterial metabolites. Based on this premise, we propose possible clinical implications. Various risk factors could be causal for severe Covid-19, and these include advanced age, concomitant chronic disease, SARS-CoV-2 infection of enterocytes, use of antibiotics and psychological distress. Gut dysbiosis is associated with risk factors and severe Covid-19 due to decreased commensal microbial metabolites, which cause reduced anti-inflammatory mechanisms and chronic low-grade inflammation. The preconditioned immune dysregulation enables SARS-CoV-2 infection to progress to an uncontrolled hyperinflammatory response. Thus, a pre-existing gut microbiota that is diverse and abundant could be beneficial for the prevention of severe Covid-19, and supplementation with commensal microbial metabolites may facilitate and augment the treatment of severe Covid-19.
Collapse
Affiliation(s)
| | - Sean Hall
- Research DepartmentMedlab ClinicalSydneyAustralia
| | - Luis Vitetta
- Research DepartmentMedlab ClinicalSydneyAustralia
- Faculty of Medicine and HealthThe University of SydneySydneyAustralia
| |
Collapse
|
49
|
Koureas M, Amoutzias GD, Vontas A, Kyritsi M, Pinaka O, Papakonstantinou A, Dadouli K, Hatzinikou M, Koutsolioutsou A, Mouchtouri VA, Speletas M, Tsiodras S, Hadjichristodoulou C. Wastewater monitoring as a supplementary surveillance tool for capturing SARS-COV-2 community spread. A case study in two Greek municipalities. ENVIRONMENTAL RESEARCH 2021; 200:111749. [PMID: 34310965 PMCID: PMC8302483 DOI: 10.1016/j.envres.2021.111749] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Revised: 07/16/2021] [Accepted: 07/16/2021] [Indexed: 05/06/2023]
Abstract
A pilot study was conducted from late October 2020 until mid-April 2021, aiming to examine the association between SARS-CoV-2 RNA concentrations in untreated wastewater and recorded COVID-19 cases in two Greek municipalities. A population of Random Forest and Linear Regression Machine Learning models was trained and evaluated incorporating the concentrations of SARS-CoV-2 RNA in 111 wastewater samples collected from the inlets of two Wastewater Treatment Plants, along with physicochemical parameters of the wastewater influent. The model's predictions were adequately associated with the 7-day cumulative cases with the correlation coefficients (after 5-fold cross validation) ranging from 0.754 to 0.960 while the mean relative errors ranged from 30.42% to 59.46%. Our results provide indications that wastewater-based predictions can be applied in diverse settings and in prolonged time periods, although the accuracy of these predictions may be mitigated. Wastewater-based epidemiology can support and strengthen epidemiological surveillance.
Collapse
Affiliation(s)
- Michalis Koureas
- Laboratory of Hygiene and Epidemiology, Faculty of Medicine, University of Thessaly, 22 Papakyriazi str, Larissa, Greece
| | - Grigoris D Amoutzias
- Bioinformatics Laboratory, Department of Biochemistry and Biotechnology, School of Health Sciences, University of Thessaly, Biopolis, Larissa, 41500, Greece
| | - Alexandros Vontas
- Laboratory of Hygiene and Epidemiology, Faculty of Medicine, University of Thessaly, 22 Papakyriazi str, Larissa, Greece
| | - Maria Kyritsi
- Laboratory of Hygiene and Epidemiology, Faculty of Medicine, University of Thessaly, 22 Papakyriazi str, Larissa, Greece
| | - Ourania Pinaka
- Laboratory of Hygiene and Epidemiology, Faculty of Medicine, University of Thessaly, 22 Papakyriazi str, Larissa, Greece
| | | | - Katerina Dadouli
- Laboratory of Hygiene and Epidemiology, Faculty of Medicine, University of Thessaly, 22 Papakyriazi str, Larissa, Greece
| | - Marina Hatzinikou
- Laboratory of Hygiene and Epidemiology, Faculty of Medicine, University of Thessaly, 22 Papakyriazi str, Larissa, Greece
| | | | - Varvara A Mouchtouri
- Laboratory of Hygiene and Epidemiology, Faculty of Medicine, University of Thessaly, 22 Papakyriazi str, Larissa, Greece
| | - Matthaios Speletas
- Department of Immunology and Histocompatibility, Faculty of Medicine, University of Thessaly, Larissa, Greece
| | - Sotirios Tsiodras
- Hellenic National Public Health Organisation, Chimarras 6, 15125, Marousi Attica, Greece; Fourth Department of Internal Medicine, National and Kapodistrian University of Athens, School of Medicine, Attikon University Hospital, Athens, Greece
| | - Christos Hadjichristodoulou
- Laboratory of Hygiene and Epidemiology, Faculty of Medicine, University of Thessaly, 22 Papakyriazi str, Larissa, Greece.
| |
Collapse
|
50
|
Veterini AS, Andriyanto L, Hamzah H. A CASE REPORT: RESPIRATORY MANIFESTATIONS OF COVID-19 STARTING WITH A GASTROINTESTINAL COMPLAINT: A COINCIDENCE OR A CORRELATION? Afr J Infect Dis 2021; 15:31-37. [PMID: 34595384 PMCID: PMC8457346 DOI: 10.21010/ajidv15i2.4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Revised: 07/28/2021] [Accepted: 07/28/2021] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND SARS COV-2 is the cause of the current outbreak of COVID-19. The infection of SARS COV-2 causes changes in the gut-lung axis and the intestinal microbiota pro-inflammatory cytokines interaction which leads to the injury of the gastrointestinal tract. One of the symptoms of COVID-19 outside the respiratory system is a complaint in the GIT. MATERIALS AND METHODS We present a COVID-19 case report that begins with a complaint of abdominal pain. RESULTS There was no previous suspicion of COVID-19, but after a radiological examination and SARS-COV2 positive PCR result, the patient was proven to be suffering from COVID-19. CONCLUSION After hospitalization in the ICU for about 14 days, a recovery occurred and the patient was able to go home in a very good clinical condition.
Collapse
Affiliation(s)
- Anna Surgean Veterini
- Anesthesiology and Intensive Care, Special Hospital for Infection-Universitas Airlangga, Indonasia
| | - Lucky Andriyanto
- Anesthesiology and Intensive Care, Special Hospital for Infection-Universitas Airlangga, Indonasia
| | - Hamzah Hamzah
- Anesthesiology and Intensive Care, Special Hospital for Infection-Universitas Airlangga, Indonasia
| |
Collapse
|