1
|
Wang Y, Li J, Chen R, Xu Q, Wang D, Mao C, Xiang Z, Wu G, Yu Y, Li J, Zheng Y, Chen K. Emerging concepts in mucosal immunity and oral microecological control of respiratory virus infection-related inflammatory diseases. Microbiol Res 2024; 289:127930. [PMID: 39427450 DOI: 10.1016/j.micres.2024.127930] [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: 01/08/2024] [Revised: 09/22/2024] [Accepted: 10/06/2024] [Indexed: 10/22/2024]
Abstract
Oral microecological imbalance is closely linked to oral mucosal inflammation and is implicated in the development of both local and systemic diseases, including those caused by viral infections. This review examines the critical role of the interleukin (IL)-17/helper T cell 17 (Th17) axis in regulating immune responses within the oral mucosa, focusing on both its protective and pathogenic roles during inflammation. We specifically highlight how the IL-17/Th17 pathway contributes to dysregulated inflammation in the context of respiratory viral infections. Furthermore, this review explores the potential interactions between respiratory viruses and the oral microbiota, emphasizing how alterations in the oral microbiome and increased production of proinflammatory factors may serve as early, non-invasive biomarkers for predicting the severity of respiratory viral infections. These findings provide insights into novel diagnostic approaches and therapeutic strategies aimed at mitigating respiratory disease severity through monitoring and modulating the oral microbiome.
Collapse
Affiliation(s)
- Ying Wang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, China
| | - Jiaxuan Li
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, P.R. China
| | - Ruyi Chen
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, P.R. China
| | - Qiuyi Xu
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, P.R. China
| | - Di Wang
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, P.R. China
| | - Chenxi Mao
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, P.R. China
| | - Ziyi Xiang
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, P.R. China
| | - Guangshang Wu
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, P.R. China
| | - Ying Yu
- School of Public Health, Hangzhou Medical College, Hangzhou, Zhejiang 310063, China
| | - Jianhua Li
- Zhejiang Key Laboratory of Public Health Detection and Pathogenesis Research, Department of Microbiology, Zhejiang Provincial Center for Disease Control and Prevention, Hangzhou, China.
| | - Yuejuan Zheng
- The Research Center for Traditional Chinese Medicine, Shanghai Institute of Infectious Diseases and Biosecurity, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Keda Chen
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, P.R. China.
| |
Collapse
|
2
|
Li B, Bai J, Xiong Y, Guo D, Fu B, Deng G, Wu H. Understanding the mechanisms and treatments of long COVID to address future public health risks. Life Sci 2024; 353:122938. [PMID: 39084516 DOI: 10.1016/j.lfs.2024.122938] [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: 02/20/2024] [Revised: 07/20/2024] [Accepted: 07/27/2024] [Indexed: 08/02/2024]
Abstract
The 2019 coronavirus disease (COVID-19), triggered by the severe acute respiratory syndrome coronavirus (SARS-CoV-2), has seen numerous individuals undergo and recover from it, drawing extensive attention to their health conditions. Extensive studies indicate that even after surpassing the acute phase of infection, patients continue to experience persistent symptoms such as fatigue, pain, depression, weakening, and anosmia. COVID-19 appears not to have concluded but rather to persist long-term in certain individuals, termed as "long COVID." This represents a heterogeneous ailment involving multiple organ systems, with a perceived complex and still elusive pathogenesis. Among patients with long COVID, observations reveal immune dysregulation, coagulation impairments, and microbial dysbiosis, considered potential mechanisms explaining sustained adverse outcomes post COVID-19. Based on the multifactorial nature, varied symptoms, and heterogeneity of long COVID, we have summarized several categories of current therapeutic approaches. Furthermore, the symptoms of long COVID resemble those of other viral illnesses, suggesting that existing knowledge may offer novel insights into long-term COVID implications. Here, we provide an overview of existing literature associated with long COVID and summarize potential mechanisms, treatment modalities, and other analogous conditions. Lastly, we underscore the inadequacies in long COVID treatment approaches and emphasize the significance of conducting further research and clinical trials.
Collapse
Affiliation(s)
- Bohao Li
- School of Life Sciences, Chongqing University, Chongqing 401331, China
| | - Junlu Bai
- School of Life Sciences, Chongqing University, Chongqing 401331, China
| | - Yan Xiong
- School of Life Sciences, Chongqing University, Chongqing 401331, China
| | - Dong Guo
- School of Life Sciences, Chongqing University, Chongqing 401331, China
| | - Beibei Fu
- School of Life Sciences, Chongqing University, Chongqing 401331, China
| | - Guohong Deng
- Department of Infectious Diseases, First Affiliated Hospital, Army Medical University, Chongqing, China.
| | - Haibo Wu
- School of Life Sciences, Chongqing University, Chongqing 401331, China.
| |
Collapse
|
3
|
Xiang B, Zhang Q, Wu H, Lin J, Xu Z, Zhang M, Zhu L, Hu J, Zhi M. Impact of Mild COVID-19 History on Oral-Gut Microbiota and Serum Metabolomics in Adult Patients with Crohn's Disease: Potential Beneficial Effects. Biomedicines 2024; 12:2103. [PMID: 39335616 PMCID: PMC11429124 DOI: 10.3390/biomedicines12092103] [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: 08/09/2024] [Revised: 09/01/2024] [Accepted: 09/08/2024] [Indexed: 09/30/2024] Open
Abstract
The impact of coronavirus disease 2019 (COVID-19) history on Crohn's disease (CD) is unknown. This investigation aimed to examine the effect of COVID-19 history on the disease course, oral-gut microbiota, and serum metabolomics in patients with CD. In this study, oral-gut microbiota and serum metabolomic profiles in 30 patients with CD and a history of mild COVID-19 (positive group, PG), 30 patients with CD without COVID-19 history (negative group, NG), and 60 healthy controls (HC) were assessed using 16S rDNA sequencing and targeted metabolomics. During follow-up, the CD activity index showed a stronger decrease in the PG than in the NG (p = 0.0496). PG patients demonstrated higher α-diversity and distinct β-diversity clustering in both salivary and fecal microbiota compared to NG and HC individuals. Notably, the gut microbiota composition in the PG patients showed a significantly greater similarity to that of HC than NG individuals. The interaction between oral and intestinal microbiota in the PG was reduced. Moreover, serum metabolome analysis revealed significantly increased anti-inflammatory metabolites, including short-chain fatty acids and N-Acetylserotonin, among PG patients; meanwhile, inflammation-related metabolites such as arachidonic acid were significantly reduced in this group. Our data suggest that the gut microbiota mediates a potential beneficial effect of a mild COVID-19 history in CD patients.
Collapse
Affiliation(s)
- Bingjie Xiang
- Department of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China; (B.X.); (Q.Z.); (H.W.); (J.L.); (Z.X.); (M.Z.)
- Guangdong Institute of Gastroenterology, Guangzhou 510655, China;
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Disease, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China
- Key Laboratory of Human Microbiome and Chronic Diseases, Sun Yat-sen University, Ministry of Education, Guangzhou 510655, China
| | - Qi Zhang
- Department of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China; (B.X.); (Q.Z.); (H.W.); (J.L.); (Z.X.); (M.Z.)
- Guangdong Institute of Gastroenterology, Guangzhou 510655, China;
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Disease, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China
- Key Laboratory of Human Microbiome and Chronic Diseases, Sun Yat-sen University, Ministry of Education, Guangzhou 510655, China
| | - Huibo Wu
- Department of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China; (B.X.); (Q.Z.); (H.W.); (J.L.); (Z.X.); (M.Z.)
- Guangdong Institute of Gastroenterology, Guangzhou 510655, China;
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Disease, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China
- Key Laboratory of Human Microbiome and Chronic Diseases, Sun Yat-sen University, Ministry of Education, Guangzhou 510655, China
| | - Jue Lin
- Department of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China; (B.X.); (Q.Z.); (H.W.); (J.L.); (Z.X.); (M.Z.)
- Guangdong Institute of Gastroenterology, Guangzhou 510655, China;
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Disease, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China
- Key Laboratory of Human Microbiome and Chronic Diseases, Sun Yat-sen University, Ministry of Education, Guangzhou 510655, China
| | - Zhaoyuan Xu
- Department of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China; (B.X.); (Q.Z.); (H.W.); (J.L.); (Z.X.); (M.Z.)
- Guangdong Institute of Gastroenterology, Guangzhou 510655, China;
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Disease, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China
- Key Laboratory of Human Microbiome and Chronic Diseases, Sun Yat-sen University, Ministry of Education, Guangzhou 510655, China
| | - Min Zhang
- Department of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China; (B.X.); (Q.Z.); (H.W.); (J.L.); (Z.X.); (M.Z.)
- Guangdong Institute of Gastroenterology, Guangzhou 510655, China;
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Disease, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China
- Key Laboratory of Human Microbiome and Chronic Diseases, Sun Yat-sen University, Ministry of Education, Guangzhou 510655, China
| | - Lixin Zhu
- Guangdong Institute of Gastroenterology, Guangzhou 510655, China;
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Disease, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China
- Key Laboratory of Human Microbiome and Chronic Diseases, Sun Yat-sen University, Ministry of Education, Guangzhou 510655, China
- Department of Colorectal Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China
| | - Jun Hu
- Guangdong Institute of Gastroenterology, Guangzhou 510655, China;
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Disease, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China
- Key Laboratory of Human Microbiome and Chronic Diseases, Sun Yat-sen University, Ministry of Education, Guangzhou 510655, China
- Department of Colorectal Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China
| | - Min Zhi
- Department of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China; (B.X.); (Q.Z.); (H.W.); (J.L.); (Z.X.); (M.Z.)
- Guangdong Institute of Gastroenterology, Guangzhou 510655, China;
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Disease, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China
- Key Laboratory of Human Microbiome and Chronic Diseases, Sun Yat-sen University, Ministry of Education, Guangzhou 510655, China
- Department of Colorectal Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China
| |
Collapse
|
4
|
Trimarco V, Izzo R, Jankauskas SS, Fordellone M, Signoriello G, Manzi MV, Lembo M, Gallo P, Esposito G, Piccinocchi R, Rozza F, Morisco C, Mone P, Piccinocchi G, Varzideh F, Trimarco B, Santulli G. A six-year study in a real-world population reveals an increased incidence of dyslipidemia during COVID-19. J Clin Invest 2024; 134:e183777. [PMID: 39264723 PMCID: PMC11527440 DOI: 10.1172/jci183777] [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: 06/11/2024] [Accepted: 09/06/2024] [Indexed: 09/14/2024] Open
Abstract
BACKGROUNDRecent studies conducted in individuals who survived COVID-19 suggest that SARS-CoV-2 infection is associated with an increased risk of dyslipidemia. However, it remains unclear whether this augmented risk is confirmed in the general population and how this phenomenon is affecting the overall burden of cardiometabolic diseases.METHODSTo address these aspects, we conducted a 6-year longitudinal study to examine the broader effects of COVID-19 on dyslipidemia incidence in a real-world population (228,266 individuals) residing in Naples in southern Italy. The pre-COVID-19 and COVID-19 groups were balanced for demographic and clinical factors using propensity score matching.RESULTSOur analysis spans a period of 3 years during the COVID-19 pandemic (2020-2022), comparing dyslipidemia incidence with pre-pandemic data (2017-2019), with a follow-up of at least 1,095 days corresponding to 21,349,215 person-years. During the COVID-19 period, we detected an increased risk of developing any dyslipidemia when compared with the pre-COVID-19 triennium (OR = 1.29; 95% CI, 1.19-1.39). Importantly, these estimates were adjusted for comorbidities by a multivariate analysis.CONCLUSIONSTaken together, our data reveal a notable rise in dyslipidemia incidence during the COVID-19 pandemic, suggesting the utility of establishing specialized clinical monitoring protocols for patients who survive COVID-19 to mitigate the risk of developing dyslipidemia.
Collapse
Affiliation(s)
| | - Raffaele Izzo
- Department of Advanced Biomedical Sciences, “Federico II” University, Naples, Italy
| | - Stanislovas S. Jankauskas
- Department of Molecular Pharmacology, Fleischer Institute for Diabetes and Metabolism (FIDAM), Einstein Institute for Aging Research, Albert Einstein College of Medicine, New York, New York, USA
| | - Mario Fordellone
- Department of Mental, Physical Health and Preventive Medicine and
| | - Giuseppe Signoriello
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli,” Naples, Italy
| | - Maria Virginia Manzi
- Department of Advanced Biomedical Sciences, “Federico II” University, Naples, Italy
| | - Maria Lembo
- Department of Advanced Biomedical Sciences, “Federico II” University, Naples, Italy
| | - Paola Gallo
- Department of Neuroscience, Reproductive Sciences, and Dentistry and
| | - Giovanni Esposito
- Department of Advanced Biomedical Sciences, “Federico II” University, Naples, Italy
| | | | - Francesco Rozza
- Department of Advanced Biomedical Sciences, “Federico II” University, Naples, Italy
| | - Carmine Morisco
- Department of Advanced Biomedical Sciences, “Federico II” University, Naples, Italy
- Italian Society for Cardiovascular Prevention (SIPREC), Rome, Italy
- International Translational Research and Medical Education (ITME), Academic Research Unit, Naples, Italy
| | - Pasquale Mone
- Department of Molecular Pharmacology, Fleischer Institute for Diabetes and Metabolism (FIDAM), Einstein Institute for Aging Research, Albert Einstein College of Medicine, New York, New York, USA
- Department of Medicine and Health Sciences “Vincenzo Tiberio,” Molise University, Campobasso, Italy
- Casa di Cura Montevergine, Mercogliano, Avellino, Italy
| | - Gaetano Piccinocchi
- COMEGEN Primary Care Physicians Cooperative, Italian Society of General Medicine (SIMG), Naples, Italy
| | - Fahimeh Varzideh
- Department of Molecular Pharmacology, Fleischer Institute for Diabetes and Metabolism (FIDAM), Einstein Institute for Aging Research, Albert Einstein College of Medicine, New York, New York, USA
| | - Bruno Trimarco
- Department of Advanced Biomedical Sciences, “Federico II” University, Naples, Italy
- International Translational Research and Medical Education (ITME), Academic Research Unit, Naples, Italy
| | - Gaetano Santulli
- Department of Advanced Biomedical Sciences, “Federico II” University, Naples, Italy
- Department of Molecular Pharmacology, Fleischer Institute for Diabetes and Metabolism (FIDAM), Einstein Institute for Aging Research, Albert Einstein College of Medicine, New York, New York, USA
- International Translational Research and Medical Education (ITME), Academic Research Unit, Naples, Italy
- Department of Medicine, Division of Cardiology, Einstein–Mount Sinai Diabetes Research Center (ES-DRC), Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, New York, New York, USA
| |
Collapse
|
5
|
Zaatry R, Herren R, Gefen T, Geva-Zatorsky N. Microbiome and infectious disease: diagnostics to therapeutics. Microbes Infect 2024; 26:105345. [PMID: 38670215 DOI: 10.1016/j.micinf.2024.105345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 04/22/2024] [Accepted: 04/22/2024] [Indexed: 04/28/2024]
Abstract
Over 300 years of research on the microbial world has revealed their importance in human health and disease. This review explores the impact and potential of microbial-based detection methods and therapeutic interventions, integrating research of early microbiologists, current findings, and future perspectives.
Collapse
Affiliation(s)
- Rawan Zaatry
- Rappaport Faculty of Medicine, Rappaport Technion Integrated Cancer Center, Technion, Haifa, Israel
| | - Rachel Herren
- Rappaport Faculty of Medicine, Rappaport Technion Integrated Cancer Center, Technion, Haifa, Israel
| | - Tal Gefen
- Rappaport Faculty of Medicine, Rappaport Technion Integrated Cancer Center, Technion, Haifa, Israel
| | - Naama Geva-Zatorsky
- Rappaport Faculty of Medicine, Rappaport Technion Integrated Cancer Center, Technion, Haifa, Israel; CIFAR, Humans & the Microbiome, Toronto, Canada.
| |
Collapse
|
6
|
B Gowda SG, Shekhar C, Gowda D, Chen Y, Chiba H, Hui SP. Mass spectrometric approaches in discovering lipid biomarkers for COVID-19 by lipidomics: Future challenges and perspectives. MASS SPECTROMETRY REVIEWS 2024; 43:1041-1065. [PMID: 37102760 DOI: 10.1002/mas.21848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Revised: 03/14/2023] [Accepted: 04/17/2023] [Indexed: 05/09/2023]
Abstract
Coronavirus disease 2019 (COVID-19) has emerged as a global health threat and has rapidly spread worldwide. Significant changes in the lipid profile before and after COVID-19 confirmed the significance of lipid metabolism in regulating the response to viral infection. Therefore, understanding the role of lipid metabolism may facilitate the development of new therapeutics for COVID-19. Owing to their high sensitivity and accuracy, mass spectrometry (MS)-based methods are widely used for rapidly identifying and quantifying of thousands of lipid species present in a small amount of sample. To enhance the capabilities of MS for the qualitative and quantitative analysis of lipids, different platforms have been combined to cover a wide range of lipidomes with high sensitivity, specificity, and accuracy. Currently, MS-based technologies are being established as efficient methods for discovering potential diagnostic biomarkers for COVID-19 and related diseases. As the lipidome of the host cell is drastically affected by the viral replication process, investigating lipid profile alterations in patients with COVID-19 and targeting lipid metabolism pathways are considered to be crucial steps in host-directed drug targeting to develop better therapeutic strategies. This review summarizes various MS-based strategies that have been developed for lipidomic analyzes and biomarker discoveries to combat COVID-19 by integrating various other potential approaches using different human samples. Furthermore, this review discusses the challenges in using MS technologies and future perspectives in terms of drug discovery and diagnosis of COVID-19.
Collapse
Affiliation(s)
- Siddabasave Gowda B Gowda
- Faculty of Health Sciences, Hokkaido University, Sapporo, Japan
- Graduate School of Global Food Resources, Hokkaido University, Sapporo, Japan
| | - Chandra Shekhar
- Faculty of Health Sciences, Hokkaido University, Sapporo, Japan
| | - Divyavani Gowda
- Faculty of Health Sciences, Hokkaido University, Sapporo, Japan
| | - Yifan Chen
- Faculty of Health Sciences, Hokkaido University, Sapporo, Japan
| | - Hitoshi Chiba
- Department of Nutrition, Sapporo University of Health Sciences, Sapporo, Japan
| | - Shu-Ping Hui
- Faculty of Health Sciences, Hokkaido University, Sapporo, Japan
| |
Collapse
|
7
|
Xu J, Wu D, Yang J, Zhao Y, Liu X, Chang Y, Tang Y, Sun F, Zhao Y. Adult Outpatients with Long COVID Infected with SARS-CoV-2 Omicron Variant. Part 1: Oral Microbiota Alterations. Am J Med 2024:S0002-9343(24)00489-3. [PMID: 39151680 DOI: 10.1016/j.amjmed.2024.07.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 07/27/2024] [Accepted: 07/30/2024] [Indexed: 08/19/2024]
Abstract
BACKGROUND Many individuals experience long COVID after SARS-CoV-2 infection. As microbiota can influence health, it may change with COVID-19. This study investigated differences in oral microbiota between COVID-19 patients with and without long COVID. METHODS Based on a prospective follow-up investigation, this nested case-control study evaluated the differences in oral microbiota in individuals with and without long COVID (Symptomatic and Asymptomatic groups), which were assessed by 16S rRNA sequencing on tongue coating samples. A predictive model was established using machine learning based on specific differential microbial communities. RESULTS One-hundred-and-eight patients were included (n=54 Symptomatic group). The Symptomatic group had higher Alpha diversity indices (observed_otus, Chao1, Shannon, and Simpson indices), differences in microbial composition (Beta diversity), and microbial dysbiosis with increased diversity and relative abundance of pathogenic bacteria. Marker bacteria (c__Campylobacterota, o__Coriobacteriales, o__Pseudomonadales, and o__Campylobacterales) were associated with long COVID by linear discriminant analysis effect size and receiver operating characteristic curves (AUC 0.821). CONCLUSION There were distinct variations in oral microbiota between COVID-19 patients with and without long COVID. Changes in oral microbiota may indicate long COVID.
Collapse
Affiliation(s)
- Jianchao Xu
- Hebei University of Chinese Medicine, Shijiazhuang, China; Shijiazhuang People's Hospital, Shijiazhuang, China
| | - Di Wu
- Hebei University of Chinese Medicine, Shijiazhuang, China; The Traditional Chinese Medicine Hospital of Shijiazhuang, Shijiazhuang, China
| | - Jie Yang
- Hebei General Hospital, Shijiazhuang, China
| | - Yinuo Zhao
- Faculty of Biology, Medicine and Health, School of Biological Sciences, The University of Manchester, Manchester, UK
| | - Xuzhao Liu
- Handan Hospital of Integrated Chinese and Western Medicine, Handan, China
| | - Yingying Chang
- The Traditional Chinese Medicine Hospital of Shijiazhuang, Shijiazhuang, China
| | - Yao Tang
- Wuhan Metware Biotechnology Co, Ltd, Wuhan, China
| | - Feng Sun
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, China; Key Laboratory of Epidemiology of Major Diseases (Peking University), Beijing, China
| | - Yubin Zhao
- Hebei University of Chinese Medicine, Shijiazhuang, China; Shijiazhuang People's Hospital, Shijiazhuang, China; Shijiazhuang College of Applied Technology, China.
| |
Collapse
|
8
|
Li H, Wen J, Zhang X, Dai Z, Liu M, Zhang H, Zhang N, Lei R, Luo P, Zhang J. Large-scale genetic correlation studies explore the causal relationship and potential mechanism between gut microbiota and COVID-19-associated risks. BMC Microbiol 2024; 24:292. [PMID: 39103761 DOI: 10.1186/s12866-024-03423-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 07/10/2024] [Indexed: 08/07/2024] Open
Abstract
Recent observational studies suggest that gut microorganisms are involved in the onset and development of coronavirus disease 2019 (COVID-19), but the potential causal relationship behind them remains unclear. Exposure data were derived from the MiBioGen consortium, encompassing 211 gut microbiota (n = 18,340). The outcome data were sourced from the COVID-19 host genetics initiative (round 7), including COVID-19 severity (n = 1,086,211), hospitalization (n = 2,095,324), and susceptibility (n = 2,597,856). First, a two-sample Mendelian randomization (TSMR) was performed to investigate the causal effect between gut microbiota and COVID-19 outcomes. Second, a two-step MR was used to explore the potential mediators and underlying mechanisms. Third, several sensitivity analyses were performed to verify the robustness of the results. Five gut microbes were found to have a potential causality with COVID-19 severity, namely Betaproteobacteria (beta = 0.096, p = 0.034), Christensenellaceae (beta = -0.092, p = 0.023), Adlercreutzia (beta = 0.072, p = 0.048), Coprococcus 1 (beta = 0.089, p = 0.032), Eisenbergiella (beta = 0.064, p = 0.024). Seven gut microbes were found to have a potential causality with COVID-19 hospitalization, namely Victivallaceae (beta = 0.037, p = 0.028), Actinomyces (beta = 0.047, p = 0.046), Coprococcus 2 (beta = -0.061, p = 0.031), Dorea (beta = 0.067, p = 0.016), Peptococcus (beta = -0.035, p = 0.049), Rikenellaceae RC9 gut group (beta = 0.034, p = 0.018), and Proteobacteria (beta = -0.069, p = 0.035). Two gut microbes were found to have a potential causality with COVID-19 susceptibility, namely Holdemanella (beta = -0.024, p = 0.023) and Lachnospiraceae FCS020 group (beta = 0.026, p = 0.027). Multi-omics mediation analyses indicate that numerous plasma proteins, metabolites, and immune factors are critical mediators linking gut microbiota with COVID-19 outcomes. Sensitivity analysis suggested no significant heterogeneity or pleiotropy. These findings revealed the causal correlation and potential mechanism between gut microbiota and COVID-19 outcomes, which may improve our understanding of the gut-lung axis in the etiology and pathology of COVID-19 in the future.
Collapse
Affiliation(s)
- He Li
- The Animal Laboratory Center, Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Jie Wen
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- Hypothalamic Pituitary Research Centre, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Xiangbin Zhang
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Ziyu Dai
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- Hypothalamic Pituitary Research Centre, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Mingren Liu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- Hypothalamic Pituitary Research Centre, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Hao Zhang
- Department of Neurosurgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Nan Zhang
- College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Ruoyan Lei
- Xiangya School of Public Health, Central South University, Changsha, China
| | - Peng Luo
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China.
| | - Jingwei Zhang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China.
- Hypothalamic Pituitary Research Centre, Xiangya Hospital, Central South University, Changsha, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.
| |
Collapse
|
9
|
Narayanan A, Kieri O, Vesterbacka J, Manoharan L, Chen P, Ghorbani M, Ljunggren HG, Sällberg Chen M, Aleman S, Sönnerborg A, Ray S, Nowak P. Exploring the interplay between antiretroviral therapy and the gut-oral microbiome axis in people living with HIV. Sci Rep 2024; 14:17820. [PMID: 39090139 PMCID: PMC11294597 DOI: 10.1038/s41598-024-68479-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 07/24/2024] [Indexed: 08/04/2024] Open
Abstract
The gut and oral microbiome is altered in people living with HIV (PLWH). While antiretroviral treatment (ART) is pivotal in restoring immune function in PLWH, several studies have identified an association between specific antiretrovirals, particularly integrase inhibitors (INSTI), and weight gain. In our study, we explored the differences in the oral and gut microbiota of PLWH under different ART regimens, and its correlation to Body Mass Index (BMI). Fecal and salivary samples were collected from PLWH (n = 69) and healthy controls (HC, n = 80). We performed taxonomy analysis to determine the microbial composition and relationship between microbial abundance and ART regimens, BMI, CD4+T-cell count, CD4/CD8 ratio, and ART duration. PLWH showed significantly lower richness compared to HC in both the oral and gut environment. The gut microbiome composition of INSTI-treated individuals was enriched with Faecalibacterium and Bifidobacterium, whereas non-nucleotide reverse transcriptase inhibitor (NNRTI)-treated individuals were enriched with Gordonibacter, Megasphaera, and Staphylococcus. In the oral microenvironment, Veillonella was significantly more abundant in INSTI-treated individuals and Fusobacterium and Alloprevotella in the NNRTI-treated individuals. Furthermore, Bifidobacterium and Dorea were enriched in gut milieu of PLWH with high BMI. Collectively, our findings identify distinct microbial profiles, which are associated with different ART regimens and BMI in PLWH on successful ART, thereby highlighting significant effects of specific antiretrovirals on the microbiome.
Collapse
Affiliation(s)
- Aswathy Narayanan
- Department of Medicine Huddinge, Division of Infectious Diseases, Karolinska Institutet, Stockholm, Sweden.
| | - Oscar Kieri
- Department of Medicine Huddinge, Division of Infectious Diseases, Karolinska Institutet, Stockholm, Sweden
- Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden
| | - Jan Vesterbacka
- Department of Medicine Huddinge, Division of Infectious Diseases, Karolinska Institutet, Stockholm, Sweden
- Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden
| | - Lokeshwaran Manoharan
- Department of Laboratory Medicine, National Bioinformatics Infrastructure Sweden (NBIS), SciLife, Lund University, Lund, Sweden
| | - Puran Chen
- Department of Medicine Huddinge, Center for Infectious Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Mahin Ghorbani
- Department of Dental Medicine, Karolinska Institutet, Stockholm, Sweden
- Department of Laboratory Medicine, Division of Pathology, ANA Futura, Karolinska Institutet, 141 52, Stockholm, Sweden
| | - Hans-Gustaf Ljunggren
- Department of Medicine Huddinge, Center for Infectious Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Margaret Sällberg Chen
- Department of Dental Medicine, Karolinska Institutet, Stockholm, Sweden
- Department of Laboratory Medicine, Division of Pathology, ANA Futura, Karolinska Institutet, 141 52, Stockholm, Sweden
| | - Soo Aleman
- Department of Medicine Huddinge, Division of Infectious Diseases, Karolinska Institutet, Stockholm, Sweden
- Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden
| | - Anders Sönnerborg
- Department of Medicine Huddinge, Division of Infectious Diseases, Karolinska Institutet, Stockholm, Sweden
- Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden
- Department of Laboratory Medicine, Division of Clinical Microbiology, ANA Futura, Karolinska Institutet, 141 52, Stockholm, Sweden
| | - Shilpa Ray
- Department of Medicine Huddinge, Division of Infectious Diseases, Karolinska Institutet, Stockholm, Sweden.
| | - Piotr Nowak
- Department of Medicine Huddinge, Division of Infectious Diseases, Karolinska Institutet, Stockholm, Sweden
- Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
10
|
Essex M, Millet Pascual-Leone B, Löber U, Kuhring M, Zhang B, Brüning U, Fritsche-Guenther R, Krzanowski M, Fiocca Vernengo F, Brumhard S, Röwekamp I, Anna Bielecka A, Lesker TR, Wyler E, Landthaler M, Mantei A, Meisel C, Caesar S, Thibeault C, Corman VM, Marko L, Suttorp N, Strowig T, Kurth F, Sander LE, Li Y, Kirwan JA, Forslund SK, Opitz B. Gut microbiota dysbiosis is associated with altered tryptophan metabolism and dysregulated inflammatory response in COVID-19. NPJ Biofilms Microbiomes 2024; 10:66. [PMID: 39085233 PMCID: PMC11291933 DOI: 10.1038/s41522-024-00538-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 07/22/2024] [Indexed: 08/02/2024] Open
Abstract
The clinical course of COVID-19 is variable and often unpredictable. To test the hypothesis that disease progression and inflammatory responses associate with alterations in the microbiome and metabolome, we analyzed metagenome, metabolome, cytokine, and transcriptome profiles of repeated samples from hospitalized COVID-19 patients and uninfected controls, and leveraged clinical information and post-hoc confounder analysis. Severe COVID-19 was associated with a depletion of beneficial intestinal microbes, whereas oropharyngeal microbiota disturbance was mainly linked to antibiotic use. COVID-19 severity was also associated with enhanced plasma concentrations of kynurenine and reduced levels of several other tryptophan metabolites, lysophosphatidylcholines, and secondary bile acids. Moreover, reduced concentrations of various tryptophan metabolites were associated with depletion of Faecalibacterium, and tryptophan decrease and kynurenine increase were linked to enhanced production of inflammatory cytokines. Collectively, our study identifies correlated microbiome and metabolome alterations as a potential contributor to inflammatory dysregulation in severe COVID-19.
Collapse
Affiliation(s)
- Morgan Essex
- Experimental and Clinical Research Center (ECRC), a cooperation of the Max Delbrück Center and Charité-Universitätsmedizin, Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Charité-Universitätsmedizin Berlin, a corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Belén Millet Pascual-Leone
- Department of Infectious Diseases, Respiratory Medicine and Critical Care, Charité-Universitätsmedizin Berlin, a corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Ulrike Löber
- Experimental and Clinical Research Center (ECRC), a cooperation of the Max Delbrück Center and Charité-Universitätsmedizin, Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Charité-Universitätsmedizin Berlin, a corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Mathias Kuhring
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Berlin Institute of Health (BIH) at Charité, BIH Metabolomics Platform, Berlin, Germany
- Berlin Institute of Health (BIH) at Charité, Core Unit Bioinformatics, Berlin, Germany
| | - Bowen Zhang
- Department of Computational Biology for Individualized Infection Medicine, Center for Individualized Infection Medicine (CiiM), a joint venture between the Helmholtz-Center for Infection Research (HZI) and the Hannover Medical School (MHH), Hannover, Germany
- TWINCORE, joint ventures between the Helmholtz Center for Infection Research (HZI) and the Hannover Medical School (MHH), Hannover, Germany
- College of Life Sciences, Beijing Normal University, Beijing, China
| | - Ulrike Brüning
- Berlin Institute of Health (BIH) at Charité, BIH Metabolomics Platform, Berlin, Germany
| | | | - Marta Krzanowski
- Department of Infectious Diseases, Respiratory Medicine and Critical Care, Charité-Universitätsmedizin Berlin, a corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Facundo Fiocca Vernengo
- Department of Infectious Diseases, Respiratory Medicine and Critical Care, Charité-Universitätsmedizin Berlin, a corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Sophia Brumhard
- Department of Infectious Diseases, Respiratory Medicine and Critical Care, Charité-Universitätsmedizin Berlin, a corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Ivo Röwekamp
- Department of Infectious Diseases, Respiratory Medicine and Critical Care, Charité-Universitätsmedizin Berlin, a corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Agata Anna Bielecka
- Department of Microbial Immune Regulation, Helmholtz Center for Infection Research (HZI), Braunschweig, Germany
- German Center for Infection Research (DZIF), partner site Hannover-Braunschweig, Braunschweig, Germany
| | - Till Robin Lesker
- Department of Microbial Immune Regulation, Helmholtz Center for Infection Research (HZI), Braunschweig, Germany
| | - Emanuel Wyler
- Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Markus Landthaler
- Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Institute of Biology, Humboldt-Universität zu Berlin, Berlin, Germany
| | | | - Christian Meisel
- Labor Berlin-Charité Vivantes GmbH, Berlin, Germany
- Institute of Medical Immunology, Charité-Universitätsmedizin Berlin, a corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Sandra Caesar
- Department of Infectious Diseases, Respiratory Medicine and Critical Care, Charité-Universitätsmedizin Berlin, a corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Charlotte Thibeault
- Department of Infectious Diseases, Respiratory Medicine and Critical Care, Charité-Universitätsmedizin Berlin, a corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Victor M Corman
- Labor Berlin-Charité Vivantes GmbH, Berlin, Germany
- Institute of Virology, Charité-Universitätsmedizin Berlin, a corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- German Center for Infection Research (DZIF), Berlin, Germany
| | - Lajos Marko
- Experimental and Clinical Research Center (ECRC), a cooperation of the Max Delbrück Center and Charité-Universitätsmedizin, Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Charité-Universitätsmedizin Berlin, a corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- German Center for Cardiovascular Research (DZHK), partner site Berlin, Berlin, Germany
| | - Norbert Suttorp
- Department of Infectious Diseases, Respiratory Medicine and Critical Care, Charité-Universitätsmedizin Berlin, a corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- German Center for Lung Research (DZL), Berlin, Germany
| | - Till Strowig
- Department of Computational Biology for Individualized Infection Medicine, Center for Individualized Infection Medicine (CiiM), a joint venture between the Helmholtz-Center for Infection Research (HZI) and the Hannover Medical School (MHH), Hannover, Germany
- Department of Microbial Immune Regulation, Helmholtz Center for Infection Research (HZI), Braunschweig, Germany
- German Center for Infection Research (DZIF), partner site Hannover-Braunschweig, Braunschweig, Germany
| | - Florian Kurth
- Department of Infectious Diseases, Respiratory Medicine and Critical Care, Charité-Universitätsmedizin Berlin, a corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Leif E Sander
- Department of Infectious Diseases, Respiratory Medicine and Critical Care, Charité-Universitätsmedizin Berlin, a corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- German Center for Lung Research (DZL), Berlin, Germany
| | - Yang Li
- Department of Computational Biology for Individualized Infection Medicine, Center for Individualized Infection Medicine (CiiM), a joint venture between the Helmholtz-Center for Infection Research (HZI) and the Hannover Medical School (MHH), Hannover, Germany
- TWINCORE, joint ventures between the Helmholtz Center for Infection Research (HZI) and the Hannover Medical School (MHH), Hannover, Germany
| | - Jennifer A Kirwan
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Berlin Institute of Health (BIH) at Charité, BIH Metabolomics Platform, Berlin, Germany
- University of Nottingham School of Veterinary Medicine and Science, Loughborough, UK
| | - Sofia K Forslund
- Experimental and Clinical Research Center (ECRC), a cooperation of the Max Delbrück Center and Charité-Universitätsmedizin, Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Charité-Universitätsmedizin Berlin, a corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- German Center for Cardiovascular Research (DZHK), partner site Berlin, Berlin, Germany
- Structural and Computational Biology Unit, European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
| | - Bastian Opitz
- Department of Infectious Diseases, Respiratory Medicine and Critical Care, Charité-Universitätsmedizin Berlin, a corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.
- Labor Berlin-Charité Vivantes GmbH, Berlin, Germany.
- German Center for Lung Research (DZL), Berlin, Germany.
| |
Collapse
|
11
|
Hawryłkowicz V, Stasiewicz B, Maciejewska D, Sołek-Pastuszka J, Komorniak N, Skonieczna-Żydecka K, Martynova-Van Kley A, Stachowska E. The Link between Inflammation, Lipid Derivatives, and Microbiota Metabolites in COVID-19 Patients: Implications on Eating Behaviors and Nutritional Status. Int J Mol Sci 2024; 25:7899. [PMID: 39063142 PMCID: PMC11276903 DOI: 10.3390/ijms25147899] [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: 06/07/2024] [Revised: 07/13/2024] [Accepted: 07/17/2024] [Indexed: 07/28/2024] Open
Abstract
Extreme inflammation that continues even after infections can lead to a cytokine storm. In recent times, one of the most common causes of cytokine storm activation has been SARS-CoV-2 infection. A cytokine storm leads to dysregulation and excessive stimulation of the immune system, producing symptoms typical of post-COVID syndrome, including chronic fatigue, shortness of breath, joint pain, trouble concentrating (known as "brain fog"), and even direct organ damage in the heart, lungs, kidneys, and brain. This work summarizes the current knowledge regarding inflammation and the cytokine storm related to SARS-CoV-2 infection. Additionally, changes in lipid metabolism and microbiota composition under the influence of inflammation in COVID-19, along with the possible underlying mechanisms, are described. Finally, this text explores potential health implications related to changes in eating behaviors and nutritional status in COVID-19 patients. Although research on the cytokine storm is still ongoing, there is convincing evidence suggesting that severe immune and inflammatory responses during the acute phase of COVID-19 may lead to long-term health consequences. Understanding these links is key to developing treatment strategies and supporting patients after infection.
Collapse
Affiliation(s)
- Viktoria Hawryłkowicz
- Department of Human Nutrition and Metabolomics, Pomeranian Medical University, 71-460 Szczecin, Poland; (V.H.); (D.M.); (N.K.)
| | - Beata Stasiewicz
- Department of Human Nutrition, The Faculty of Food Science, University of Warmia and Mazury in Olsztyn, Sloneczna 45f, 10-718 Olsztyn, Poland
| | - Dominika Maciejewska
- Department of Human Nutrition and Metabolomics, Pomeranian Medical University, 71-460 Szczecin, Poland; (V.H.); (D.M.); (N.K.)
| | - Joanna Sołek-Pastuszka
- Department of Anesthesiology and Intensive Care, Pomeranian Medical University, 71-242 Szczecin, Poland;
| | - Natalia Komorniak
- Department of Human Nutrition and Metabolomics, Pomeranian Medical University, 71-460 Szczecin, Poland; (V.H.); (D.M.); (N.K.)
| | | | | | - Ewa Stachowska
- Department of Human Nutrition and Metabolomics, Pomeranian Medical University, 71-460 Szczecin, Poland; (V.H.); (D.M.); (N.K.)
| |
Collapse
|
12
|
Giancola ML, Fontana A, Panebianco C, Mazzarelli A, Beccacece A, De Marco P, Cocomazzi G, De Giuli C, Grassi G, Fontana C, Baldini GM, Contu V, Copetti M, Perri F, Nicastri E, Pazienza V. Efficacy of a Multistrain Synbiotic Treatment in Acute and Post-Acute COVID-19 Patients: A Double-Blind, Placebo-Controlled Randomized Trial. Microorganisms 2024; 12:1443. [PMID: 39065211 PMCID: PMC11279369 DOI: 10.3390/microorganisms12071443] [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: 06/17/2024] [Revised: 07/03/2024] [Accepted: 07/15/2024] [Indexed: 07/28/2024] Open
Abstract
BACKGROUND AND AIMS Several studies reported the effect of COVID-19 on inducing gut dysbiosis, which is also correlated with disease severity. This study aims to investigate the effect of a nutraceutical formula on the shift of microbiota profiles and, secondly, on the clinical-pathological parameters of acute and post-acute COVID-19 patients. METHODS In this randomised, double-blind, placebo-controlled trial conducted at National Institute for Infectious diseases (INMI) Lazzaro Spallanzani (Italy), 52 patients were randomly assigned (1:1) to receive a multistrain synbiotic formula (Kebirah®) or placebo orally for 35 days at COVID-19 diagnosis. Health professionals, investigators, and patients were masked to group assignments. The V3-V4 hypervariable region of 16S rRNA gene sequencing was employed to study the gut microbiota composition in the two groups. RESULTS Supplementation with Kebirah® prevented the decrease in the Shannon diversity index of gut microbiota, which was instead observed in patients receiving the placebo. In addition, decreases in lymphocyte count and haemoglobin levels were observed only in the placebo group and not in the treated group, which was also characterised by an amelioration of the gut microbial profile, with an enrichment in beneficial bacteria and a preservation of species diversity. CONCLUSIONS Our data suggest that modulating the gut microbiota in acute disease through administration of a specific symbiotic formula could be a useful strategy in the frame of SARS-CoV-2 infections.
Collapse
Affiliation(s)
- Maria Letizia Giancola
- National Institute for Infectious Diseases, INMI “Lazzaro Spallanzani”, IRCCS, 00149 Rome, Italy; (M.L.G.); (A.M.); (A.B.); (P.D.M.); (C.D.G.); (G.G.); (C.F.)
| | - Andrea Fontana
- Biostatistic Unit, Fondazione-IRCCS “Casa Sollievo della Sofferenza” Hospital, 71013 San Giovanni Rotondo, FG, Italy; (A.F.); (M.C.)
| | - Concetta Panebianco
- Gastroenterology Unit, Fondazione-IRCCS “Casa Sollievo della Sofferenza” Hospital, Opera di San Pio da Pietrelcina, 71013 San Giovanni Rotondo, FG, Italy; (C.P.); (G.C.)
| | - Antonio Mazzarelli
- National Institute for Infectious Diseases, INMI “Lazzaro Spallanzani”, IRCCS, 00149 Rome, Italy; (M.L.G.); (A.M.); (A.B.); (P.D.M.); (C.D.G.); (G.G.); (C.F.)
| | - Alessia Beccacece
- National Institute for Infectious Diseases, INMI “Lazzaro Spallanzani”, IRCCS, 00149 Rome, Italy; (M.L.G.); (A.M.); (A.B.); (P.D.M.); (C.D.G.); (G.G.); (C.F.)
| | - Patrizia De Marco
- National Institute for Infectious Diseases, INMI “Lazzaro Spallanzani”, IRCCS, 00149 Rome, Italy; (M.L.G.); (A.M.); (A.B.); (P.D.M.); (C.D.G.); (G.G.); (C.F.)
| | - Giovanna Cocomazzi
- Gastroenterology Unit, Fondazione-IRCCS “Casa Sollievo della Sofferenza” Hospital, Opera di San Pio da Pietrelcina, 71013 San Giovanni Rotondo, FG, Italy; (C.P.); (G.C.)
| | - Chiara De Giuli
- National Institute for Infectious Diseases, INMI “Lazzaro Spallanzani”, IRCCS, 00149 Rome, Italy; (M.L.G.); (A.M.); (A.B.); (P.D.M.); (C.D.G.); (G.G.); (C.F.)
| | - Germana Grassi
- National Institute for Infectious Diseases, INMI “Lazzaro Spallanzani”, IRCCS, 00149 Rome, Italy; (M.L.G.); (A.M.); (A.B.); (P.D.M.); (C.D.G.); (G.G.); (C.F.)
| | - Carla Fontana
- National Institute for Infectious Diseases, INMI “Lazzaro Spallanzani”, IRCCS, 00149 Rome, Italy; (M.L.G.); (A.M.); (A.B.); (P.D.M.); (C.D.G.); (G.G.); (C.F.)
| | - Giorgio Maria Baldini
- AO Consorziale Policlinico di Bari, Università Aldo Moro di Bari, 70121 Bari, BA, Italy;
| | - Viviana Contu
- Integrative Medicine Unit, Humanitas Gradenigo, Corso Regina Margherita 8/10, 10153 Torino, TO, Italy;
| | - Massimiliano Copetti
- Biostatistic Unit, Fondazione-IRCCS “Casa Sollievo della Sofferenza” Hospital, 71013 San Giovanni Rotondo, FG, Italy; (A.F.); (M.C.)
| | - Francesco Perri
- Gastroenterology Unit, Fondazione-IRCCS “Casa Sollievo della Sofferenza” Hospital, Opera di San Pio da Pietrelcina, 71013 San Giovanni Rotondo, FG, Italy; (C.P.); (G.C.)
| | - Emanuele Nicastri
- National Institute for Infectious Diseases, INMI “Lazzaro Spallanzani”, IRCCS, 00149 Rome, Italy; (M.L.G.); (A.M.); (A.B.); (P.D.M.); (C.D.G.); (G.G.); (C.F.)
| | - Valerio Pazienza
- Gastroenterology Unit, Fondazione-IRCCS “Casa Sollievo della Sofferenza” Hospital, Opera di San Pio da Pietrelcina, 71013 San Giovanni Rotondo, FG, Italy; (C.P.); (G.C.)
| |
Collapse
|
13
|
Chen Y, Li C, Wang X, Zhang CL, Ren ZG, Wang ZQ. Oral microbiota distinguishes patients with osteosarcoma from healthy controls. Front Cell Infect Microbiol 2024; 14:1383878. [PMID: 39055977 PMCID: PMC11269967 DOI: 10.3389/fcimb.2024.1383878] [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: 02/18/2024] [Accepted: 06/17/2024] [Indexed: 07/28/2024] Open
Abstract
Objective The human microbiota plays a key role in cancer diagnosis, pathogenesis, and treatment. However, osteosarcoma-associated oral microbiota alterations have not yet been unraveled. The aim of this study was to explore the characteristics of oral microbiota in osteosarcoma patients compared to healthy controls, and to identify potential microbiota as a diagnostic tool for osteosarcoma. Methods The oral microbiota was analyzed in osteosarcoma patients (n = 45) and matched healthy controls (n = 90) using 16S rRNA MiSeq sequencing technology. Results The microbial richness and diversity of the tongue coat were increased in osteosarcoma patients as estimated by the abundance-based coverage estimator indices, the Chao, and observed operational taxonomy units (OTUs). Principal component analysis delineated that the oral microbial community was significant differences between osteosarcoma patients and healthy controls. 14 genera including Rothia, Halomonas, Rhodococcus, and Granulicatella were remarkably reduced, whereas Alloprevotella, Prevotella, Selenomonas, and Campylobacter were enriched in osteosarcoma. Eventually, the optimal four OTUs were identified to construct a microbial classifier by the random forest model via a fivefold cross-validation, which achieved an area under the curve of 99.44% in the training group (30 osteosarcoma patients versus 60 healthy controls) and 87.33% in the test group (15 osteosarcoma patients versus 30 healthy controls), respectively. Notably, oral microbial markers validated strong diagnostic potential distinguishing osteosarcoma patients from healthy controls. Conclusion This study comprehensively characterizes the oral microbiota in osteosarcoma and reveals the potential efficacy of oral microbiota-targeted biomarkers as a noninvasive biological diagnostic tool for osteosarcoma.
Collapse
Affiliation(s)
- Yu Chen
- Department of Pathogen Biology, Medical College, Zhengzhou University, Zhengzhou, China
| | - Chao Li
- Department of Orthopaedic Surgery, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, China
| | - Xin Wang
- Department of Orthopaedic Surgery, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, China
| | - Chun Lei Zhang
- Department of Orthopaedic Surgery, Henan Provincial Chest Hospital, Zhengzhou University, Zhengzhou, China
| | - Zhi Gang Ren
- Department of Infectious Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhong Quan Wang
- Department of Pathogen Biology, Medical College, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
14
|
Missa KF, Diallo K, Bla KB, Tuo KJ, Gboko KDT, Tiémélé LS, Ouattara AF, Gragnon BG, Ngoi JM, Wilkinson RJ, Awandare GA, Bonfoh B. Association of symptomatic upper respiratory tract infections with the alteration of the oropharyngeal microbiome in a cohort of school children in Côte d'Ivoire. Front Microbiol 2024; 15:1412923. [PMID: 38993497 PMCID: PMC11238735 DOI: 10.3389/fmicb.2024.1412923] [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: 04/08/2024] [Accepted: 05/29/2024] [Indexed: 07/13/2024] Open
Abstract
Introduction The oropharyngeal microbiome plays an important role in protection against infectious agents when in balance. Despite use of vaccines and antibiotic therapy to prevent respiratory tract infections, they remain one of the major causes of mortality and morbidity in Low- and middle-income countries. Hence the need to explore other approaches to prevention by identifying microbial biomarkers that could be leveraged to modify the microbiota in order to enhance protection against pathogenic bacteria. The aim of this study was to analyze the oropharyngeal microbiome (OPM) of schoolchildren in Côte d'Ivoire presenting symptoms of upper respiratory tract infections (URTI) for better prevention strategy. Methods Primary schools' children in Korhogo (n = 37) and Abidjan (n = 39) were followed for six months with monthly oropharyngeal sampling. Clinical diagnostic of URT infection was performed and nucleic acid extracted from oropharyngeal swabs were used for 16S rRNA metagenomic analysis and RT-PCR. Results The clinical examination of children's throat in Abidjan and Korhogo identified respectively 17 (43.59%) and 15 (40.54%) participants with visible symptoms of URTIs, with 26 episodes of infection in Abidjan and 24 in Korhogo. Carriage of Haemophilus influenzae (12%), Streptococcus pneumoniae (6%) and SARS-CoV-2 (6%) was confirmed by PCR. A significant difference in alpha diversity was found between children colonized by S. pneumoniae and those that were not (p = 0.022). There was also a significant difference in alpha diversity between children colonised with H. influenzae and those who were not (p = 0.017). No significant difference was found for SARS-CoV-2. Sphingomonas, Ralstonia and Rothia were significantly enriched in non-carriers of S. pneumoniae; Actinobacillus was significantly enriched in non-carriers of H. influenzae; Actinobacillus and Porphyromonas were significantly enriched in non-carriers of SARS-CoV-2 (p < 0.001). Discussion Nearly 40% of children showed clinical symptoms of infection not related to geographical location. The OPM showed an imbalance during H. influenzae and S. pneumoniae carriage. This study provides a baseline understanding of microbiome markers in URTIs in children for future research, to develop targeted interventions aimed at restoring the microbial balance and reducing the symptoms associated with RTIs.
Collapse
Affiliation(s)
- Kouassi Firmin Missa
- Direction de la Recherche et du Développement, Centre Suisse de Recherches Scientifiques en Côte d'Ivoire, Abidjan, Côte d'Ivoire
- Laboratoire de Biologie et Santé, UFR Biosciences, Université Félix Houphouët Boigny de Cocody, Abidjan, Côte d'Ivoire
| | - Kanny Diallo
- Direction de la Recherche et du Développement, Centre Suisse de Recherches Scientifiques en Côte d'Ivoire, Abidjan, Côte d'Ivoire
- West African Centre for Cell Biology of Infectious Pathogens, Accra, Ghana
| | - Kouakou Brice Bla
- Laboratoire de Biologie et Santé, UFR Biosciences, Université Félix Houphouët Boigny de Cocody, Abidjan, Côte d'Ivoire
| | - Kolotioloman Jérémie Tuo
- Direction de la Recherche et du Développement, Centre Suisse de Recherches Scientifiques en Côte d'Ivoire, Abidjan, Côte d'Ivoire
- Laboratoire de Microbiologie, Biotechnologies et Bio-informatique, Institut National Polytechnique Félix Houphouët-Boigny, Yamoussoukro, Côte d'Ivoire
| | - Kossia Debia Thérèse Gboko
- Direction de la Recherche et du Développement, Centre Suisse de Recherches Scientifiques en Côte d'Ivoire, Abidjan, Côte d'Ivoire
| | - Laurent-Simon Tiémélé
- Direction de la Recherche et du Développement, Centre Suisse de Recherches Scientifiques en Côte d'Ivoire, Abidjan, Côte d'Ivoire
| | - Allassane Foungoye Ouattara
- Direction de la Recherche et du Développement, Centre Suisse de Recherches Scientifiques en Côte d'Ivoire, Abidjan, Côte d'Ivoire
- Laboratoire de Cytologie et Biologie Animale, Université Nangui Abrogoua, Abidjan, Côte d'Ivoire
| | - Biego Guillaume Gragnon
- Laboratoire de Cytologie et Biologie Animale, Université Nangui Abrogoua, Abidjan, Côte d'Ivoire
| | | | - Robert J Wilkinson
- Laboratoire National d'Appui au Développement Agricole, Laboratoire Régional de Korhogo, Korhogo, Côte d'Ivoire
- The Francis Crick Institute, London, United Kingdom
- Department of Infectious Disease, Imperial College London, London, United Kingdom
| | - Gordon A Awandare
- West African Centre for Cell Biology of Infectious Pathogens, Accra, Ghana
| | - Bassirou Bonfoh
- Direction de la Recherche et du Développement, Centre Suisse de Recherches Scientifiques en Côte d'Ivoire, Abidjan, Côte d'Ivoire
| |
Collapse
|
15
|
Chen J, Gao X, Liang J, Wu Q, Shen L, Zheng Y, Ma Y, Peng Y, He Y, Yin J. Association between gut microbiota dysbiosis and poor functional outcomes in acute ischemic stroke patients with COVID-19 infection. mSystems 2024; 9:e0018524. [PMID: 38700338 PMCID: PMC11237522 DOI: 10.1128/msystems.00185-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2024] [Accepted: 03/26/2024] [Indexed: 05/05/2024] Open
Abstract
Acute ischemic stroke (AIS) patients with active COVID-19 infection often have more severe symptoms and worse recovery. COVID-19 infection can cause gut microbiota dysbiosis, which is also a risk factor for poor outcomes in AIS patients. However, the association between gut microbiota and functional outcomes among AIS patients with COVID-19 infection has not been fully clarified yet. In this study, we performed 16S rRNA gene sequencing to characterize the gut microbial community among AIS patients with acute COVID-19 infection, AIS patients with post-acute COVID-19 infection, and AIS patients without COVID-19 infection. We found that AIS patients with acute COVID-19 experienced poorer recovery and significant gut dysbiosis, characterized by higher levels of Enterobacteriaceae and lower levels of Ruminococcaceae and Lachnospiraceae. Furthermore, a shorter time window (less than 28 days) between COVID-19 infection and stroke was identified as a risk factor for poor functional outcomes in AIS patients with COVID-19, and the enrichment of Enterobacteriaceae was indicated as a mediator in the relationship between infection time window and poor stroke outcomes. Our findings highlight the importance of early intervention after COVID-19 infection, especially by regulating the gut microbiota, which plays a role in the prognosis of AIS patients with COVID-19 infection.IMPORTANCEThe gut microbiota plays an important role in the association between respiratory system and cerebrovascular system through the gut-lung axis and gut-brain axis. However, the specific connection between gut bacteria and the functional outcomes of acute ischemic stroke (AIS) patients with COVID-19 is not fully understood yet. In our study, we observed a significant decrease in bacterial diversity and shifts in the abundance of key bacterial families in AIS patients with acute COVID-19 infection. Furthermore, we identified that the time window was a critical influence factor for stroke outcomes, and the enrichment of Enterobacteriaceae acted as a mediator in the relationship between the infection time window and poor stroke outcomes. Our research provides a new perspective on the complex interplay among AIS, COVID-19 infection, and gut microbiota dysbiosis. Moreover, recognizing Enterobacteriaceae as a potential mediator of poor stroke prognosis offers a novel avenue for future exploration and therapeutic interventions.
Collapse
Affiliation(s)
- Jiaying Chen
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
- Comprehensive Medical Treatment Ward, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xuxuan Gao
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Jingru Liang
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Qiheng Wu
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Linlin Shen
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Yifeng Zheng
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Yu Ma
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Yuping Peng
- Department of Neurosurgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Yan He
- Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
- Guangdong Provincial Clinical Research Center for Laboratory Medicine, Guangzhou, Guangdong, China
- State Key Laboratory of Organ Failure Research, Southern Medical University, Guangzhou, Guangdong, China
- Key Laboratory of Mental Health of the Ministry of Education, Guangzhou, Guangdong, China
| | - Jia Yin
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
16
|
Elgedawy GA, Samir M, Elabd NS, Elsaid HH, Enar M, Salem RH, Montaser BA, AboShabaan HS, Seddik RM, El-Askaeri SM, Omar MM, Helal ML. Metabolic profiling during COVID-19 infection in humans: Identification of potential biomarkers for occurrence, severity and outcomes using machine learning. PLoS One 2024; 19:e0302977. [PMID: 38814977 PMCID: PMC11139268 DOI: 10.1371/journal.pone.0302977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 04/15/2024] [Indexed: 06/01/2024] Open
Abstract
BACKGROUND After its emergence in China, the coronavirus SARS-CoV-2 has swept the world, leading to global health crises with millions of deaths. COVID-19 clinical manifestations differ in severity, ranging from mild symptoms to severe disease. Although perturbation of metabolism has been reported as a part of the host response to COVID-19 infection, scarce data exist that describe stage-specific changes in host metabolites during the infection and how this could stratify patients based on severity. METHODS Given this knowledge gap, we performed targeted metabolomics profiling and then used machine learning models and biostatistics to characterize the alteration patterns of 50 metabolites and 17 blood parameters measured in a cohort of 295 human subjects. They were categorized into healthy controls, non-severe, severe and critical groups with their outcomes. Subject's demographic and clinical data were also used in the analyses to provide more robust predictive models. RESULTS The non-severe and severe COVID-19 patients experienced the strongest changes in metabolite repertoire, whereas less intense changes occur during the critical phase. Panels of 15, 14, 2 and 2 key metabolites were identified as predictors for non-severe, severe, critical and dead patients, respectively. Specifically, arginine and malonyl methylmalonyl succinylcarnitine were significant biomarkers for the onset of COVID-19 infection and tauroursodeoxycholic acid were potential biomarkers for disease progression. Measuring blood parameters enhanced the predictive power of metabolic signatures during critical illness. CONCLUSIONS Metabolomic signatures are distinctive for each stage of COVID-19 infection. This has great translation potential as it opens new therapeutic and diagnostic prospective based on key metabolites.
Collapse
Affiliation(s)
- Gamalat A. Elgedawy
- Department of Clinical Biochemistry and Molecular Diagnostics, National Liver Institute, Menoufia University, Shebin El-Kom, Menoufia, Egypt
| | - Mohamed Samir
- Faculty of Veterinary Medicine, Department of Zoonoses, Zagazig University, Zagazig, Egypt
| | - Naglaa S. Elabd
- Faculty of Medicine, Department of Tropical Medicine, Menoufia University, Shebin El-Kom, Menoufia, Egypt
| | - Hala H. Elsaid
- Department of Clinical Biochemistry and Molecular Diagnostics, National Liver Institute, Menoufia University, Shebin El-Kom, Menoufia, Egypt
| | - Mohamed Enar
- Al Mahala Elkobra Fever Hospital, Al Mahala Elkobra, Egypt
| | - Radwa H. Salem
- Department of Clinical Microbiology and Immunology, National Liver Institute, Menoufia University, Shebin El-Kom, Menoufia, Egypt
| | - Belal A. Montaser
- Faculty of Medicine, Department of Clinical Pathology, Menoufia University, Shebin El-Kom, Menoufia, Egypt
| | - Hind S. AboShabaan
- Ph.D. of Biochemistry, National Liver Institute Hospital, Menoufia University, Shebin El-Kom, Menoufia, Egypt
| | - Randa M. Seddik
- Faculty of Medicine, Department of Tropical Medicine, Menoufia University, Shebin El-Kom, Menoufia, Egypt
| | - Shimaa M. El-Askaeri
- Department of Clinical Microbiology and Immunology, National Liver Institute, Menoufia University, Shebin El-Kom, Menoufia, Egypt
| | - Marwa M. Omar
- Faculty of Medicine, Department of Clinical Pathology, Menoufia University, Shebin El-Kom, Menoufia, Egypt
| | - Marwa L. Helal
- Department of Clinical Biochemistry and Molecular Diagnostics, National Liver Institute, Menoufia University, Shebin El-Kom, Menoufia, Egypt
| |
Collapse
|
17
|
Pusadkar V, Mazumder A, Azad A, Patil D, Azad RK. Deciphering Microbial Shifts in the Gut and Lung Microbiomes of COVID-19 Patients. Microorganisms 2024; 12:1058. [PMID: 38930440 PMCID: PMC11205787 DOI: 10.3390/microorganisms12061058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 05/17/2024] [Accepted: 05/21/2024] [Indexed: 06/28/2024] Open
Abstract
COVID-19, caused by SARS-CoV-2, results in respiratory and cardiopulmonary infections. There is an urgent need to understand not just the pathogenic mechanisms of this disease but also its impact on the physiology of different organs and microbiomes. Multiple studies have reported the effects of COVID-19 on the gastrointestinal microbiota, such as promoting dysbiosis (imbalances in the microbiome) following the disease's progression. Deconstructing the dynamic changes in microbiome composition that are specifically correlated with COVID-19 patients remains a challenge. Motivated by this problem, we implemented a biomarker discovery pipeline to identify candidate microbes specific to COVID-19. This involved a meta-analysis of large-scale COVID-19 metagenomic data to decipher the impact of COVID-19 on the human gut and respiratory microbiomes. Metagenomic studies of the gut and respiratory microbiomes of COVID-19 patients and of microbiomes from other respiratory diseases with symptoms similar to or overlapping with COVID-19 revealed 1169 and 131 differentially abundant microbes in the human gut and respiratory microbiomes, respectively, that uniquely associate with COVID-19. Furthermore, by utilizing machine learning models (LASSO and XGBoost), we demonstrated the power of microbial features in separating COVID-19 samples from metagenomic samples representing other respiratory diseases and controls (healthy individuals), achieving an overall accuracy of over 80%. Overall, our study provides insights into the microbiome shifts occurring in COVID-19 patients, shining a new light on the compositional changes.
Collapse
Affiliation(s)
- Vaidehi Pusadkar
- Department of Biological Sciences and BioDiscovery Institute, University of North Texas, Denton, TX 76203, USA;
| | - Anirudh Mazumder
- Texas Academy of Mathematics and Science, University of North Texas, Denton, TX 76203, USA
| | - Abhijay Azad
- Texas Academy of Mathematics and Science, University of North Texas, Denton, TX 76203, USA
| | - Deepti Patil
- Texas Academy of Mathematics and Science, University of North Texas, Denton, TX 76203, USA
| | - Rajeev K. Azad
- Department of Biological Sciences and BioDiscovery Institute, University of North Texas, Denton, TX 76203, USA;
| |
Collapse
|
18
|
Wei Y, Yu W, Zhang Z, Liu S, Xue J, Wu C, Gao Z, Guo S. Comparative analysis of oropharyngeal microbiota in healthcare workers post-COVID-19. Front Cell Infect Microbiol 2024; 14:1347345. [PMID: 38828262 PMCID: PMC11140064 DOI: 10.3389/fcimb.2024.1347345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 04/25/2024] [Indexed: 06/05/2024] Open
Abstract
Background To date, more than 770 million individuals have become coronavirus disease 2019 (COVID-19) convalescents worldwide. Emerging evidence highlights the influence of COVID-19 on the oral microbiome during both acute and convalescent disease phases. Front-line healthcare workers are at an elevated risk of exposure to viral infections, and the effects of COVID-19 on their oral microbiome remain relatively unexplored. Methods Oropharyngeal swab specimens, collected one month after a negative COVID-19 test from a cohort comprising 55 healthcare workers, underwent 16S rRNA sequencing. We conducted a comparative analysis between this post-COVID-19 cohort and the pre-infection dataset from the same participants. Community composition analysis, indicator species analysis, alpha diversity assessment, beta diversity exploration, and functional prediction were evaluated. Results The Shannon and Simpson indexes of the oral microbial community declined significantly in the post-COVID-19 group when compared with the pre-infection cohort. Moreover, there was clear intergroup clustering between the two groups. In the post-COVID-19 group, the phylum Firmicutes showed a significant increase. Further, there were clear differences in relative abundance of several bacterial genera in contrast with the pre-infection group, including Streptococcus, Gemella, Granulicatella, Capnocytophaga, Leptotrichia, Fusobacterium, and Prevotella. We identified Gemella enrichment in the post-COVID-19 group, potentially serving as a recovery period performance indicator. Functional prediction revealed lipopolysaccharide biosynthesis downregulation in the post-COVID-19 group, an outcome with host inflammatory response modulation and innate defence mechanism implications. Conclusion During the recovery phase of COVID-19, the oral microbiome diversity of front-line healthcare workers failed to fully return to its pre-infection state. Despite the negative COVID-19 test result one month later, notable disparities persisted in the composition and functional attributes of the oral microbiota.
Collapse
Affiliation(s)
- Yue Wei
- Nursing of school, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Wenyi Yu
- Department of Respiratory and Critical Care Medicine, Peking University People’s Hospital, Beijing, China
| | - Zhixia Zhang
- Nursing Department, Linfen Central Hospital, Shanxi, China
| | - Siqin Liu
- Department of Prosthodontics, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jianbo Xue
- Department of Respiratory and Critical Care Medicine, Peking University People’s Hospital, Beijing, China
| | - Chunyan Wu
- Nursing of school, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Zhancheng Gao
- Department of Respiratory and Critical Care Medicine, Peking University People’s Hospital, Beijing, China
- Institute of Chest and Lung Diseases, Shanxi Medical University, Linfen, Shanxi, China
| | - Shuming Guo
- Nursing of school, Shanxi Medical University, Taiyuan, Shanxi, China
- Nursing Department, Linfen Central Hospital, Shanxi, China
| |
Collapse
|
19
|
Liu S, Wang B, Chen T, Wang H, Liu J, Zhao X, Zhang Y. Two new and effective food-extracted immunomodulatory agents exhibit anti-inflammatory response activity in the hACE2 acute lung injury murine model of COVID-19. Front Immunol 2024; 15:1374541. [PMID: 38807598 PMCID: PMC11130445 DOI: 10.3389/fimmu.2024.1374541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 04/25/2024] [Indexed: 05/30/2024] Open
Abstract
Objective The coronavirus disease 2019 (COVID-19) spread rapidly and claimed millions of lives worldwide. Acute respiratory distress syndrome (ARDS) is the major cause of COVID-19-associated deaths. Due to the limitations of current drugs, developing effective therapeutic options that can be used rapidly and safely in clinics for treating severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) infections is necessary. This study aims to investigate the effects of two food-extracted immunomodulatory agents, ajoene-enriched garlic extract (AGE) and cruciferous vegetables-extracted sulforaphane (SFN), on anti-inflammatory and immune responses in a SARS-CoV-2 acute lung injury mouse model. Methods In this study, we established a mouse model to mimic the SARS-CoV-2 infection acute lung injury model via intratracheal injection of polyinosinic:polycytidylic acid (poly[I:C]) and SARS-CoV-2 recombinant spike protein (SP). After the different agents treatment, lung sections, bronchoalveolar lavage fluid (BALF) and fresh faeces were harvested. Then, H&E staining was used to examine symptoms of interstitial pneumonia. Flow cytometry was used to examine the change of immune cell populations. Multiplex cytokines assay was used to examine the inflammatory cytokines.16S rDNA high-throughput sequencing was used to examine the change of gut microbiome. Results Our results showed that AGE and SFN significantly suppressed the symptoms of interstitial pneumonia, effectively inhibited the production of inflammatory cytokines, decreased the percentage of inflammatory cell populations, and elevated T cell populations in the mouse model. Furthermore, we also observed that the gut microbiome of genus Paramuribaculum were enriched in the AGE-treated group. Conclusion Here, for the first time, we observed that these two novel, safe, and relatively inexpensive immunomodulatory agents exhibited the same effects on anti-inflammatory and immune responses as neutralizing monoclonal antibodies (mAbs) against interleukin 6 receptor (IL-6R), which have been suggested for treating COVID-19 patients. Our results revealed the therapeutic ability of these two immunomodulatory agents in a mouse model of SARS-CoV-2 acute lung injury by promoting anti-inflammatory and immune responses. These results suggest that AGE and SFN are promising candidates for the COVID-19 treatment.
Collapse
Affiliation(s)
- Shasha Liu
- Biotherapy Center and Cancer Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Baiqiao Wang
- The First Clinical Medical College, Henan University of Chinese Medicine, Zhengzhou, China
| | - Tianran Chen
- Biotherapy Center and Cancer Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Hui Wang
- Biotherapy Center and Cancer Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jinbo Liu
- School of Life Sciences, Zhengzhou University, Zhengzhou, China
| | - Xuan Zhao
- Biotherapy Center and Cancer Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yi Zhang
- Biotherapy Center and Cancer Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- School of Life Sciences, Zhengzhou University, Zhengzhou, China
- School of Public Health, Zhengzhou University, Zhengzhou, China
- Engineering Key Laboratory for Cell Therapy of Henan Province, Zhengzhou, China
| |
Collapse
|
20
|
Geistlinger L, Mirzayi C, Zohra F, Azhar R, Elsafoury S, Grieve C, Wokaty J, Gamboa-Tuz SD, Sengupta P, Hecht I, Ravikrishnan A, Gonçalves RS, Franzosa E, Raman K, Carey V, Dowd JB, Jones HE, Davis S, Segata N, Huttenhower C, Waldron L. BugSigDB captures patterns of differential abundance across a broad range of host-associated microbial signatures. Nat Biotechnol 2024; 42:790-802. [PMID: 37697152 PMCID: PMC11098749 DOI: 10.1038/s41587-023-01872-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Accepted: 06/20/2023] [Indexed: 09/13/2023]
Abstract
The literature of human and other host-associated microbiome studies is expanding rapidly, but systematic comparisons among published results of host-associated microbiome signatures of differential abundance remain difficult. We present BugSigDB, a community-editable database of manually curated microbial signatures from published differential abundance studies accompanied by information on study geography, health outcomes, host body site and experimental, epidemiological and statistical methods using controlled vocabulary. The initial release of the database contains >2,500 manually curated signatures from >600 published studies on three host species, enabling high-throughput analysis of signature similarity, taxon enrichment, co-occurrence and coexclusion and consensus signatures. These data allow assessment of microbiome differential abundance within and across experimental conditions, environments or body sites. Database-wide analysis reveals experimental conditions with the highest level of consistency in signatures reported by independent studies and identifies commonalities among disease-associated signatures, including frequent introgression of oral pathobionts into the gut.
Collapse
Affiliation(s)
- Ludwig Geistlinger
- Center for Computational Biomedicine, Harvard Medical School, Boston, MA, USA
| | - Chloe Mirzayi
- Institute for Implementation Science in Population Health, City University of New York School of Public Health, New York, NY, USA
- Department of Epidemiology and Biostatistics, City University of New York School of Public Health, New York, NY, USA
| | - Fatima Zohra
- Institute for Implementation Science in Population Health, City University of New York School of Public Health, New York, NY, USA
- Department of Epidemiology and Biostatistics, City University of New York School of Public Health, New York, NY, USA
| | - Rimsha Azhar
- Institute for Implementation Science in Population Health, City University of New York School of Public Health, New York, NY, USA
- Department of Epidemiology and Biostatistics, City University of New York School of Public Health, New York, NY, USA
| | - Shaimaa Elsafoury
- Institute for Implementation Science in Population Health, City University of New York School of Public Health, New York, NY, USA
- Department of Epidemiology and Biostatistics, City University of New York School of Public Health, New York, NY, USA
| | - Clare Grieve
- Institute for Implementation Science in Population Health, City University of New York School of Public Health, New York, NY, USA
- Department of Epidemiology and Biostatistics, City University of New York School of Public Health, New York, NY, USA
| | - Jennifer Wokaty
- Institute for Implementation Science in Population Health, City University of New York School of Public Health, New York, NY, USA
- Department of Epidemiology and Biostatistics, City University of New York School of Public Health, New York, NY, USA
| | - Samuel David Gamboa-Tuz
- Institute for Implementation Science in Population Health, City University of New York School of Public Health, New York, NY, USA
- Department of Epidemiology and Biostatistics, City University of New York School of Public Health, New York, NY, USA
| | - Pratyay Sengupta
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology (IIT) Madras, Chennai, India
- Robert Bosch Centre for Data Science and Artificial Intelligence, Indian Institute of Technology (IIT) Madras, Chennai, India
- Centre for Integrative Biology and Systems mEdicine (IBSE), Indian Institute of Technology (IIT) Madras, Chennai, India
| | | | - Aarthi Ravikrishnan
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), Singapore, Republic of Singapore
| | - Rafael S Gonçalves
- Center for Computational Biomedicine, Harvard Medical School, Boston, MA, USA
| | - Eric Franzosa
- Infectious Disease and Microbiome Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Biostatistics, Harvard T. H. Chan School of Public Health, Boston, MA, USA
- Harvard Chan Microbiome in Public Health Center, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | - Karthik Raman
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology (IIT) Madras, Chennai, India
- Robert Bosch Centre for Data Science and Artificial Intelligence, Indian Institute of Technology (IIT) Madras, Chennai, India
- Centre for Integrative Biology and Systems mEdicine (IBSE), Indian Institute of Technology (IIT) Madras, Chennai, India
| | - Vincent Carey
- Channing Division of Network Medicine, Mass General Brigham, Harvard Medical School, Boston, MA, USA
| | - Jennifer B Dowd
- Leverhulme Centre for Demographic Science, University of Oxford, Oxford, UK
| | - Heidi E Jones
- Institute for Implementation Science in Population Health, City University of New York School of Public Health, New York, NY, USA
- Department of Epidemiology and Biostatistics, City University of New York School of Public Health, New York, NY, USA
| | - Sean Davis
- Departments of Biomedical Informatics and Medicine, University of Colorado Anschutz School of Medicine, Denver, CO, USA
| | - Nicola Segata
- Department CIBIO, University of Trento, Trento, Italy
- Istituto Europeo di Oncologia (IEO) IRCSS, Milan, Italy
| | - Curtis Huttenhower
- Infectious Disease and Microbiome Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Biostatistics, Harvard T. H. Chan School of Public Health, Boston, MA, USA
- Harvard Chan Microbiome in Public Health Center, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | - Levi Waldron
- Institute for Implementation Science in Population Health, City University of New York School of Public Health, New York, NY, USA.
- Department of Epidemiology and Biostatistics, City University of New York School of Public Health, New York, NY, USA.
- Department CIBIO, University of Trento, Trento, Italy.
| |
Collapse
|
21
|
Golzardi M, Hromić-Jahjefendić A, Šutković J, Aydin O, Ünal-Aydın P, Bećirević T, Redwan EM, Rubio-Casillas A, Uversky VN. The Aftermath of COVID-19: Exploring the Long-Term Effects on Organ Systems. Biomedicines 2024; 12:913. [PMID: 38672267 PMCID: PMC11048001 DOI: 10.3390/biomedicines12040913] [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: 04/03/2024] [Revised: 04/12/2024] [Accepted: 04/18/2024] [Indexed: 04/28/2024] Open
Abstract
BACKGROUND Post-acute sequelae of SARS-CoV-2 infection (PASC) is a complicated disease that affects millions of people all over the world. Previous studies have shown that PASC impacts 10% of SARS-CoV-2 infected patients of which 50-70% are hospitalised. It has also been shown that 10-12% of those vaccinated against COVID-19 were affected by PASC and its complications. The severity and the later development of PASC symptoms are positively associated with the early intensity of the infection. RESULTS The generated health complications caused by PASC involve a vast variety of organ systems. Patients affected by PASC have been diagnosed with neuropsychiatric and neurological symptoms. The cardiovascular system also has been involved and several diseases such as myocarditis, pericarditis, and coronary artery diseases were reported. Chronic hematological problems such as thrombotic endothelialitis and hypercoagulability were described as conditions that could increase the risk of clotting disorders and coagulopathy in PASC patients. Chest pain, breathlessness, and cough in PASC patients were associated with the respiratory system in long-COVID causing respiratory distress syndrome. The observed immune complications were notable, involving several diseases. The renal system also was impacted, which resulted in raising the risk of diseases such as thrombotic issues, fibrosis, and sepsis. Endocrine gland malfunction can lead to diabetes, thyroiditis, and male infertility. Symptoms such as diarrhea, nausea, loss of appetite, and taste were also among reported observations due to several gastrointestinal disorders. Skin abnormalities might be an indication of infection and long-term implications such as persistent cutaneous complaints linked to PASC. CONCLUSIONS Long-COVID is a multidimensional syndrome with considerable public health implications, affecting several physiological systems and demanding thorough medical therapy, and more study to address its underlying causes and long-term effects is needed.
Collapse
Affiliation(s)
- Maryam Golzardi
- Department of Genetics and Bioengineering, Faculty of Engineering and Natural Sciences, International University of Sarajevo, Hrasnicka Cesta 15, 71000 Sarajevo, Bosnia and Herzegovina; (M.G.); (J.Š.)
| | - Altijana Hromić-Jahjefendić
- Department of Genetics and Bioengineering, Faculty of Engineering and Natural Sciences, International University of Sarajevo, Hrasnicka Cesta 15, 71000 Sarajevo, Bosnia and Herzegovina; (M.G.); (J.Š.)
| | - Jasmin Šutković
- Department of Genetics and Bioengineering, Faculty of Engineering and Natural Sciences, International University of Sarajevo, Hrasnicka Cesta 15, 71000 Sarajevo, Bosnia and Herzegovina; (M.G.); (J.Š.)
| | - Orkun Aydin
- Department of Psychology, Faculty of Arts and Social Sciences, International University of Sarajevo, Hrasnicka Cesta 15, 71000 Sarajevo, Bosnia and Herzegovina; (O.A.); (P.Ü.-A.)
| | - Pinar Ünal-Aydın
- Department of Psychology, Faculty of Arts and Social Sciences, International University of Sarajevo, Hrasnicka Cesta 15, 71000 Sarajevo, Bosnia and Herzegovina; (O.A.); (P.Ü.-A.)
| | - Tea Bećirević
- Atrijum Polyclinic, 71000 Sarajevo, Bosnia and Herzegovina;
| | - Elrashdy M. Redwan
- Department of Biological Science, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia;
- Centre of Excellence in Bionanoscience Research, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Therapeutic and Protective Proteins Laboratory, Protein Research Department, Genetic Engineering and Biotechnology Research Institute, City of Scientific Research and Technological Applications (SRTA-City), New Borg EL-Arab, Alexandria 21934, Egypt
| | - Alberto Rubio-Casillas
- Autlan Regional Hospital, Health Secretariat, Autlan 48900, Jalisco, Mexico;
- Biology Laboratory, Autlan Regional Preparatory School, University of Guadalajara, Autlan 48900, Jalisco, Mexico
| | - Vladimir N. Uversky
- Department of Molecular Medicine and USF Health Byrd Alzheimer’s Research Institute, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| |
Collapse
|
22
|
Luo Y, Zhang Z, Ren J, Dou C, Wen J, Yang Y, Li X, Yan Z, Han Y. SARS-Cov-2 spike induces intestinal barrier dysfunction through the interaction between CEACAM5 and Galectin-9. Front Immunol 2024; 15:1303356. [PMID: 38686388 PMCID: PMC11056506 DOI: 10.3389/fimmu.2024.1303356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Accepted: 03/28/2024] [Indexed: 05/02/2024] Open
Abstract
Background Carcinoembryonic antigen-related cell adhesion molecule 5 (CEACAM5), as a typical tumor marker, has been found to exert immunomodulatory effects in many diseases. We previously reported the clinical and molecular evidences supporting that SARS-Cov-2 infected the gastrointestinal (GI) tract and found a reduction of CEACAM5 in COVID-19 patients' feces which associated with gut dysbiosis. Yet the role of CEACAM5 in GI infection is ill-defined. Methods Mice models were established through intraperitoneally injecting with recombinant viral spike-Fc to mimic the intestinal inflammation. We collected duodenum, jejunum, ileum and colon samples after 6h, 2 days, 4 days and 7 days of spike-Fc or control-Fc injection to perform proteomic analysis. Blood was collected from healthy donors and peripheral blood mononuclear cells (PBMC) were separated by density gradient centrifugation, then CD4+ T cells were isolated with magnetic beads and co-cultured with Caco-2 cells. Results In addition to intestinal CEACAM5, the expression of tight junction and the percent of CD4+ T lymphocytes were significantly decreased in spike-Fc group compared to control (p < 0.05), accompanied with increased level of inflammatory factors. The KEGG analysis revealed differentially expressed proteins were mainly enriched in the coronavirus disease (COVID-19), tight junction, focal adhesion, adherens junction and PI3K-Akt signaling pathway. Protein-protein interaction (PPI) network analysis identified the interaction between CEACAM5 and Galectin-9 that was also verified by molecular docking and co-IP assay. We further confirmed a reduction of CEACAM5 in SARS-CoV-2 spike stimulated enterocytes could promote the expression of Galectin-9 protein in CD4+T cells. Then it gave rise to the increasing release of inflammatory factors and increased apoptosis of CD4+T cells by inhibition of PI3K/AKT/mTOR pathway. Ultimately intestinal barrier dysfunction happened. Conclusion Our results indicated that CEACAM5 overexpression and Galectin-9 knockdown played a protective role in intestinal barrier injury upon spike-Fc stimulation. Collectively, our findings identified firstly that SARS-CoV-2 spike induced intestinal barrier dysfunction through the interaction between CEACAM5 and Galectin-9. The result provides potential therapeutic targets in intestinal barrier dysfunction for treating severe COVID patients.
Collapse
Affiliation(s)
- Yingshu Luo
- Department of Gastroenterology, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong, China
| | - Zhenling Zhang
- Department of Gastroenterology, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong, China
| | - Jiangnan Ren
- Department of Gastroenterology, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong, China
| | - Chunxu Dou
- Department of Gastroenterology, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong, China
| | - Jiancheng Wen
- Department of Gastroenterology, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong, China
| | - Yang Yang
- Department of Gastroenterology, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong, China
| | - Xiaofeng Li
- Department of Gastroenterology, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong, China
| | - Zhixiang Yan
- Guangdong Provincial Key Laboratory of Biomedical Imaging and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong, China
| | - Yanzhi Han
- Department of Gastroenterology, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong, China
| |
Collapse
|
23
|
Nawaz AD, Haider MZ, Akhtar S. COVID-19 and Alzheimer's disease: Impact of lockdown and other restrictive measures during the COVID-19 pandemic. BIOMOLECULES & BIOMEDICINE 2024; 24:219-229. [PMID: 38078809 PMCID: PMC10950341 DOI: 10.17305/bb.2023.9680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Revised: 11/12/2023] [Accepted: 11/28/2023] [Indexed: 03/14/2024]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection initially results in respiratory distress symptoms but can also lead to central nervous system (CNS) and neurological manifestations, significantly impacting coronavirus disease 2019 (COVID-19) patients with neurodegenerative diseases. Additionally, strict lockdown measures introduced to curtail the spread of COVID-19 have raised concerns over the wellbeing of patients with dementia and/or Alzheimer's disease. The aim of this review was to discuss the overlapping molecular pathologies and the potential bidirectional relationship between COVID-19 and Alzheimer's dementia, as well as the impact of lockdown/restriction measures on the neuropsychiatric symptoms (NPS) of patients with Alzheimer's dementia. Furthermore, we aimed to assess the impact of lockdown measures on the NPS of caregivers, exploring its potential effects on the quality and extent of care they provide to dementia patients.We utilized the PubMed and Google Scholar databases to search for articles on COVID-19, dementia, Alzheimer's disease, lockdown, and caregivers. Our review highlights that patients with Alzheimer's disease face an increased risk of COVID-19 infection and complications. Additionally, these patients are likely to experience greater cognitive decline. It appears that these issues are primarily caused by the SARS-CoV-2 infection and appear to be further exacerbated by restrictive/lockdown measures. Moreover, lockdown measures introduced during the pandemic have negatively impacted both the NPSs of caregivers and their perception of the wellbeing of their Alzheimer's patients. Thus, additional safeguard measures, along with pharmacological and non-pharmacological approaches, are needed to protect the wellbeing of dementia patients and their caregivers in light of this and possible future pandemics.
Collapse
Affiliation(s)
| | | | - Saghir Akhtar
- College of Medicine, QU Health, Qatar University, Doha, Qatar
| |
Collapse
|
24
|
Ahmadi Badi S, Kariman A, Bereimipour A, Shojaie S, Aghsadeghi M, Khatami S, Masotti A. Association Between Altered Microbiota Composition and Immune System-Related Genes in COVID-19 Infection. Mol Biotechnol 2024:10.1007/s12033-024-01096-8. [PMID: 38456962 DOI: 10.1007/s12033-024-01096-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Accepted: 01/18/2024] [Indexed: 03/09/2024]
Abstract
Microbiota and immunity affect the host's susceptibility to SARS-CoV-2 infection and the severity of COVID-19. This study aimed to identify significant alterations in the microbiota composition, immune signaling pathways, their potential association, and candidate microRNA in COVID-19 patients using an in silico study model. Enrichment online databases and Python programming were utilized to analyze GSE164805, GSE180594, and GSE182279, as well as NGS data of microbiota composition (PRJNA650244 and PRJNA660302) associated with COVID-19, employing amplicon-based/marker gene sequencing methods. C1, TNF, C2, IL1, and CFH genes were found to have a significant impact on immune signaling pathways. Additionally, we observed a notable decrease in Bacteroides spp. and Faecalibacterium sp., while Escherichia coli, Streptococcus spp., and Akkermansia muciniphila showed increased abundance in COVID-19. Notably, A. muciniphila demonstrated an association with immunity through C1 and TNF, while Faecalibacterium sp. was linked to C2 and IL1. The correlation between E. coli and CFH, as well as IL1 and Streptococcus spp. with C2, was identified. hsa-let-7b-5p was identified as a potential candidate that may be involved in the interaction between the microbiota composition, immune response, and COVID-19. In conclusion, integrative in silico analysis shows that these microbiota members are potentially crucial in the immune responses against COVID-19.
Collapse
Affiliation(s)
- Sara Ahmadi Badi
- Biochemistry Department, Pasteur Institute of Iran, Tehran, Iran.
- Pediatric Gastroenterology and Hepatology Research Center, Pediatrics Center of Excellence, Children's Medical Center, Tehran University of Medical Science, Tehran, Iran.
| | - Arian Kariman
- Pediatric Gastroenterology and Hepatology Research Center, Pediatrics Center of Excellence, Children's Medical Center, Tehran University of Medical Science, Tehran, Iran
| | - Ahmad Bereimipour
- Biological Sciences and BioDiscovery Institute, University of North Texas, Denton, TX, USA
| | - Shima Shojaie
- Pediatric Gastroenterology and Hepatology Research Center, Pediatrics Center of Excellence, Children's Medical Center, Tehran University of Medical Science, Tehran, Iran
| | | | - Shohreh Khatami
- Biochemistry Department, Pasteur Institute of Iran, Tehran, Iran
| | - Andrea Masotti
- Research Laboratories, Bambino Gesù Children's Hospital-IRCCS, Rome, Italy
| |
Collapse
|
25
|
Brzychczy-Sroka B, Talaga-Ćwiertnia K, Sroka-Oleksiak A, Gurgul A, Zarzecka-Francica E, Ostrowski W, Kąkol J, Drożdż K, Brzychczy-Włoch M, Zarzecka J. Standardization of the protocol for oral cavity examination and collecting of the biological samples for microbiome research using the next-generation sequencing (NGS): own experience with the COVID-19 patients. Sci Rep 2024; 14:3717. [PMID: 38355866 PMCID: PMC10867075 DOI: 10.1038/s41598-024-53992-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 02/07/2024] [Indexed: 02/16/2024] Open
Abstract
To date, publications have shown that compositions of oral microbiota differ depending on their habitats (e.g. tongue, tonsils, pharynx). The absence of set standards for the choice of the areas and conditions of material collection makes the oral microbiome one of the most difficult environments for a comparative analysis with other researchers, which is a meaningful limitation during an assessment of the potential effects of microorganisms as biomarkers in the courses of various human diseases. Therefore, standardisation of basic conditions of a dental examination and collection of material for the next generation sequencing (NGS) is worth attempting. The standardisation of the dental exam and collection of the clinical materials: saliva, swab from the tongue ridge, hard palate, palatine tonsils and oropharynx, supragingival plaque and subgingival plaque. Protocol involved the patients (n = 60), assigned to 3 groups: I-COVID-19 convalescents who received antibiotics, n = 17, II-COVID-19 convalescents, n = 23 and III-healthy individuals, n = 20. The collected biological samples were used to conduct NGS (16S rRNA). The conditions of patient preparation for collecting biological materials as well as the schedule of dental examination, were proposed. Based on the research conducted, we have indicated the dental indicators that best differentiate the group of COVID-19 patients (groups I and II) from healthy people (group III). These include the DMFT, D and BOP indices. The use of alpha and beta diversity analysis provided an overall insight into the diversity of microbial communities between specific niches and patient groups. The most different diversity between the studied group of patients (group II) and healthy people (group III) was noted in relation to the supragingival plaque. The order of activities during the dental exam as well as while collecting and securing clinical materials is particularly important to avoid technical errors and material contamination which may result in erroneous conclusions from the analyses of the results of sensitive tests such as the NGS. It has been shown that the dental indices: DMFT, D number, PI and BOP are the best prognostic parameters to assess the oral health. Based on beta diversity the most sensitive niche and susceptible to changes in the composition of the microbiota is the supragingival plaque. The procedures developed by our team can be applied as ready-to-use forms in studies conducted by other researchers.
Collapse
Affiliation(s)
- Barbara Brzychczy-Sroka
- Department of Conservative Dentistry with Endodontics, Institute of Dentistry, Jagiellonian University Medical College, Kraków, Poland
| | - Katarzyna Talaga-Ćwiertnia
- Chair of Microbiology, Department of Molecular Medical Microbiology, Faculty of Medicine, Jagiellonian University Medical College, Czysta 18, 31-121, Kraków, Poland.
| | - Agnieszka Sroka-Oleksiak
- Chair of Microbiology, Department of Molecular Medical Microbiology, Faculty of Medicine, Jagiellonian University Medical College, Czysta 18, 31-121, Kraków, Poland
| | - Artur Gurgul
- Center for Experimental and Innovative Medicine, The University of Agriculture in Kraków, Kraków, Poland
| | - Elżbieta Zarzecka-Francica
- Department of Prosthodontics and Orthodontics, Institute of Dentistry, Jagiellonian University Medical College, Kraków, Poland
| | - Wojciech Ostrowski
- Department of Conservative Dentistry with Endodontics, Institute of Dentistry, Jagiellonian University Medical College, Kraków, Poland
| | - Janusz Kąkol
- University Hospital in Cracow, Temporary COVID Ward No. 1, Kraków, Poland
| | - Kamil Drożdż
- Chair of Microbiology, Department of Molecular Medical Microbiology, Faculty of Medicine, Jagiellonian University Medical College, Czysta 18, 31-121, Kraków, Poland
| | - Monika Brzychczy-Włoch
- Chair of Microbiology, Department of Molecular Medical Microbiology, Faculty of Medicine, Jagiellonian University Medical College, Czysta 18, 31-121, Kraków, Poland
| | - Joanna Zarzecka
- Department of Conservative Dentistry with Endodontics, Institute of Dentistry, Jagiellonian University Medical College, Kraków, Poland
| |
Collapse
|
26
|
Ganesan SM, Peter TK, Withanage MHH, Boksa F, Zeng E, Martinez A, Dabdoub SM, Dhingra K, Hernandez-Kapila Y. COVID-19 associated oral and oropharyngeal microbiome: Systematic review and meta-analysis. Periodontol 2000 2024; 94:603-626. [PMID: 37277934 DOI: 10.1111/prd.12489] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Accepted: 04/01/2023] [Indexed: 06/07/2023]
Abstract
Three years into the coronavirus disease 2019 (COVID-19) pandemic, there are still growing concerns with the emergence of different variants, unknown long- and short-term effects of the virus, and potential biological mechanisms underlying etiopathogenesis and increased risk for morbidity and mortality. The role of the microbiome in human physiology and the initiation and progression of several oral and systemic diseases have been actively studied in the past decade. With the proof of viral transmission, carriage, and a potential role in etiopathogenesis, saliva and the oral environment have been a focus of COVID-19 research beyond diagnostic purposes. The oral environment hosts diverse microbial communities and contributes to human oral and systemic health. Several investigations have identified disruptions in the oral microbiome in COVID-19 patients. However, all these studies are cross-sectional in nature and present heterogeneity in study design, techniques, and analysis. Therefore, in this undertaking, we (a) systematically reviewed the current literature associating COVID-19 with changes in the microbiome; (b) performed a re-analysis of publicly available data as a means to standardize the analysis, and (c) reported alterations in the microbial characteristics in COVID-19 patients compared to negative controls. Overall, we identified that COVID-19 is associated with oral microbial dysbiosis with significant reduction in diversity. However, alterations in specific bacterial members differed across the study. Re-analysis from our pipeline shed light on Neisseria as the potential key microbial member associated with COVID-19.
Collapse
Affiliation(s)
- Sukirth M Ganesan
- Department of Periodontics, The University of Iowa College of Dentistry and Dental Clinics, Iowa, USA
- Iowa Institute for Oral Health Research, The University of Iowa College of Dentistry and Dental Clinics, Iowa City, Iowa, USA
| | - Tabitha K Peter
- Division of Biostatistics and Computational Biology, The University of Iowa College of Dentistry and Dental Clinics, Iowa City, Iowa, USA
| | - Miyuraj H H Withanage
- Division of Biostatistics and Computational Biology, The University of Iowa College of Dentistry and Dental Clinics, Iowa City, Iowa, USA
| | - Frank Boksa
- Department of Periodontics, The University of Iowa College of Dentistry and Dental Clinics, Iowa, USA
| | - Erliang Zeng
- Iowa Institute for Oral Health Research, The University of Iowa College of Dentistry and Dental Clinics, Iowa City, Iowa, USA
- Division of Biostatistics and Computational Biology, The University of Iowa College of Dentistry and Dental Clinics, Iowa City, Iowa, USA
- Department of Preventive and Community Dentistry, The University of Iowa College of Dentistry and Dental Clinics, Iowa City, Iowa, USA
| | - April Martinez
- Division of Periodontology, University of California at San Francisco School of Dentistry, San Francisco, California, USA
| | - Shareef M Dabdoub
- Department of Periodontics, The University of Iowa College of Dentistry and Dental Clinics, Iowa, USA
- Iowa Institute for Oral Health Research, The University of Iowa College of Dentistry and Dental Clinics, Iowa City, Iowa, USA
- Division of Biostatistics and Computational Biology, The University of Iowa College of Dentistry and Dental Clinics, Iowa City, Iowa, USA
| | - Kunaal Dhingra
- Periodontology Division, Centre for Dental Education and Research, All India Institute of Medical Sciences, New Delhi, India
| | - Yvonne Hernandez-Kapila
- Section of Biosystems and Function Periodontics, University of California at Los Angeles School of Dentistry, Los Angeles, California, USA
| |
Collapse
|
27
|
Vakili B, Shoaei P, Shahzamani K, Siadat SD, Shojaei H, Esfandiari Z, Nasri E, Shabani S, Zamani Moghadam A, Ataei B. Gut-Lung Microbiota Characterization in Patients with Non-Small Cell Lung Carcinoma and COVID-19 Coinfection. ARCHIVES OF IRANIAN MEDICINE 2024; 27:62-71. [PMID: 38619029 PMCID: PMC11017262 DOI: 10.34172/aim.2024.11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 12/30/2023] [Indexed: 04/16/2024]
Abstract
BACKGROUND Non-small cell lung cancer (NSCLC) patients with COVID-19 have an excessive chance of morbidity and mortality. The fecal-nasopharyngeal microbiota compositions of NSCLC patients were assessed in this study. METHODS In total, 234 samples were collected from 17 NSCLC patients infected with COVID-19, 20 NSCLC patients without confirmed COVID-19, 40 non NSCLC patients with COVID-19, and 40 healthy individuals. RESULTS In lung microbiota, the abundance of Streptococcus spp. in NSCLC patients with confirmed COVID-19 was significantly higher than the two control groups. Pseudomonas aeruginosa and Staphylococcus aureus were listed as the most frequent pulmonary bacterial groups that colonized COVID-19 patients. In fecal specimens, the numbers of Bacteroidetes, Firmicutes, and Actinobacteria phyla were significantly higher amongst NSCLC patients with COVID-19. NSCLC patients infected with COVID-19 showed lower levels of Lactobacillus spp., Akkermansia muciniphila, and Bifidobacterium spp. The counts of Streptococcus spp., in NSCLC patients with COVID-19 were significantly higher than those of healthy individuals (8.49±0.70 log CFU/g wet feces vs 8.49±0.70 log CFU/g wet feces). Prevotella spp. were enriched in the gut and respiratory tracts of COVID-19 patient groups. The unbiased analysis showed an increment in Enterococcus spp., Streptococcus spp., and Prevotella spp. CONCLUSION Eventually, it was found that compared to control groups, COVID-19 patients with NSCLC showed diminished gut bacteria diversity and increase in Lactobacillus spp., A. muciniphila, and Bifidobacterium spp. The overgrowth of Enterococcus spp., Streptococcus spp., and Prevotella spp. could be potential predictive biomarkers in the gut-lung axis of NSCLC patients with COVID-19.
Collapse
Affiliation(s)
- Bahareh Vakili
- Infectious Diseases and Tropical Medicine Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Parisa Shoaei
- Nosocomial Infection Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Kiana Shahzamani
- Hepatitis Research Center, School of Medicine, Lorestan University of Medical Sciences, Khoramabad, Iran
| | - Seyed Davar Siadat
- Department of Mycobacteriology and Pulmonary Research, Pasteur Institute of Iran, Tehran, Iran
- Microbiology Research Center (MRC), Pasteur Institute of Iran, Tehran, Iran
| | - Hasan Shojaei
- Department of Microbiology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Zahra Esfandiari
- Department of Food Science and Technology, Nutrition and Food Security Research Center, School of Nutrition and Food Science, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Elahe Nasri
- Infectious Diseases and Tropical Medicine Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Shiva Shabani
- Department of Infectious Diseases, School of Medicine, Arak University of Medical Sciences, Arak, Iran
| | - Ali Zamani Moghadam
- Infectious Diseases and Tropical Medicine Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Behrooz Ataei
- Infectious Diseases and Tropical Medicine Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
28
|
Álvarez-Santacruz C, Tyrkalska SD, Candel S. The Microbiota in Long COVID. Int J Mol Sci 2024; 25:1330. [PMID: 38279329 PMCID: PMC10816132 DOI: 10.3390/ijms25021330] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 01/19/2024] [Accepted: 01/20/2024] [Indexed: 01/28/2024] Open
Abstract
Interest in the coronavirus disease 2019 (COVID-19) has progressively decreased lately, mainly due to the great effectivity of vaccines. Furthermore, no new severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants able to circumvent the protection of these vaccines, while presenting high transmissibility and/or lethality, have appeared. However, long COVID has emerged as a huge threat to human health and economy globally. The human microbiota plays an important role in health and disease, participating in the modulation of innate and adaptive immune responses. Thus, multiple studies have found that the nasopharyngeal microbiota is altered in COVID-19 patients, with these changes associated with the onset and/or severity of the disease. Nevertheless, although dysbiosis has also been reported in long COVID patients, mainly in the gut, little is known about the possible involvement of the microbiota in the development of this disease. Therefore, in this work, we aim to fill this gap in the knowledge by discussing and comparing the most relevant studies that have been published in this field up to this point. Hence, we discuss that the relevance of long COVID has probably been underestimated, and that the available data suggest that the microbiota could be playing a pivotal role on the pathogenesis of the disease. Further research to elucidate the involvement of the microbiota in long COVID will be essential to explore new therapeutic strategies based on manipulation of the microbiota.
Collapse
Affiliation(s)
| | - Sylwia D. Tyrkalska
- Departamento de Biología Celular e Histología, Facultad de Biología, Universidad de Murcia, 30100 Murcia, Spain;
- Instituto Murciano de Investigación Biosanitaria (IMIB)-Pascual Parrilla, 30120 Murcia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Sergio Candel
- Departamento de Biología Celular e Histología, Facultad de Biología, Universidad de Murcia, 30100 Murcia, Spain;
- Instituto Murciano de Investigación Biosanitaria (IMIB)-Pascual Parrilla, 30120 Murcia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
29
|
Zhang Y, Ma Y, Sun W, Zhou X, Wang R, Xie P, Dai L, Gao Y, Li J. Exploring gut-lung axis crosstalk in SARS-CoV-2 infection: Insights from a hACE2 mouse model. J Med Virol 2024; 96:e29336. [PMID: 38193530 DOI: 10.1002/jmv.29336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Revised: 11/27/2023] [Accepted: 12/11/2023] [Indexed: 01/10/2024]
Abstract
Based on the forefront of clinical research, there is a growing recognition that the gut microbiota, which plays a pivotal role in shaping both the innate and adaptive immune systems, may significantly contribute to the pathogenesis of coronavirus disease 2019 (COVID-19). Although an association between altered gut microbiota and COVID-19 pathogenesis has been established, the causative mechanisms remain incompletely understood. Additionally, the validation of the precise functional alterations within the gut microbiota relevant to COVID-19 pathogenesis has been limited by a scarcity of suitable animal experimental models. In the present investigation, we employed a newly developed humanized ACE2 knock-in (hACE2-KI) mouse model, capable of recapitulating critical aspects of pulmonary and intestinal infection, to explore the modifications in the gut microbiota following severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. Examination of fecal samples using 16S rRNA gene profiling unveiled a notable reduction in species richness and conspicuous alterations in microbiota composition at 6 days postinfection (dpi). These alterations were primarily characterized by a decline in beneficial bacterial species and an escalation in certain opportunistic pathogens. Moreover, our analysis entailed a correlation study between the gut microbiota and plasma cytokine concentrations, revealing the potential involvement of the Lachnospiraceae_NK4A136_group and unclassified_f_Lachnospiraceae genera in attenuating hyperinflammatory responses triggered by the infection. Furthermore, integration of gut microbiota data with RNA-seq analysis results suggested that the increased presence of Staphylococcus in fecal samples may signify the potential for bacterial coinfection in lung tissues via gut translocation. In summary, our hACE2-KI mouse model effectively recapitulated the observed alterations in the gut microbiota during SARS-CoV-2 infection. This model presents a valuable tool for elucidating gut microbiota-targeted strategies aimed at mitigating COVID-19.
Collapse
Affiliation(s)
- Yu Zhang
- Department of Biosafety, School of Basic Medicine, Army Medical University, Chongqing, China
| | - Yifang Ma
- Department of Biosafety, School of Basic Medicine, Army Medical University, Chongqing, China
| | - Weiyang Sun
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Xiaoyang Zhou
- Department of Biosafety, School of Basic Medicine, Army Medical University, Chongqing, China
| | - Ruixuan Wang
- Department of Biosafety, School of Basic Medicine, Army Medical University, Chongqing, China
| | - Peng Xie
- Department of Biosafety, School of Basic Medicine, Army Medical University, Chongqing, China
| | - Lu Dai
- Department of Biosafety, School of Basic Medicine, Army Medical University, Chongqing, China
| | - Yuwei Gao
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Jintao Li
- Department of Biosafety, School of Basic Medicine, Army Medical University, Chongqing, China
| |
Collapse
|
30
|
Mir TUG, Manhas S, Khurshid Wani A, Akhtar N, Shukla S, Prakash A. Alterations in microbiome of COVID-19 patients and its impact on forensic investigations. Sci Justice 2024; 64:81-94. [PMID: 38182316 DOI: 10.1016/j.scijus.2023.12.002] [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: 02/19/2023] [Revised: 11/12/2023] [Accepted: 12/11/2023] [Indexed: 01/07/2024]
Abstract
The human microbiome is vital for maintaining human health and has garnered substantial attention in recent years, particularly in the context of the coronavirus disease 2019 (COVID-19) outbreak. Studies have underscored significant alterations in the microbiome of COVID-19 patients across various body niches, including the gut, respiratory tract, oral cavity, skin, and vagina. These changes manifest as shifts in microbiota composition, characterized by an increase in opportunistic pathogens and a decrease in beneficial commensal bacteria. Such microbiome transformations may play a pivotal role in influencing the course and severity of COVID-19, potentially contributing to the inflammatory response. This ongoing relationship between COVID-19 and the human microbiome serves as a compelling subject of research, underscoring the necessity for further investigations into the underlying mechanisms and their implications for patient health. Additionally, these alterations in the microbiome may have significant ramifications for forensic investigations, given the microbiome's potential in establishing individual characteristics. Consequently, changes in the microbiome could introduce a level of complexity into forensic determinations. As research progresses, a more profound understanding of the human microbiome within the context of COVID-19 may offer valuable insights into disease prevention, treatment strategies, and its potential applications in forensic science. Consequently, this paper aims to provide an overarching review of microbiome alterations due to COVID-19 and the associated impact on forensic applications, bridging the gap between the altered microbiome of COVID-19 patients and the challenges forensic investigations may encounter when analyzing this microbiome as a forensic biomarker.
Collapse
Affiliation(s)
- Tahir Ul Gani Mir
- School of Bioengineering and Biosciences, Lovely Professional University, Phagwara 144411, Punjab, India; State Forensic Science Laboratory, Srinagar, Jammu and Kashmir 190001, India.
| | - Sakshi Manhas
- School of Bioengineering and Biosciences, Lovely Professional University, Phagwara 144411, Punjab, India
| | - Atif Khurshid Wani
- School of Bioengineering and Biosciences, Lovely Professional University, Phagwara 144411, Punjab, India
| | - Nahid Akhtar
- School of Bioengineering and Biosciences, Lovely Professional University, Phagwara 144411, Punjab, India
| | - Saurabh Shukla
- School of Bioengineering and Biosciences, Lovely Professional University, Phagwara 144411, Punjab, India.
| | - Ajit Prakash
- Department of Biochemistry and Biophysics, University of North Carolina, Chapel Hill, NC 27599, USA
| |
Collapse
|
31
|
Bezinelli LM, Corrêa L, Beyerstedt S, Franco ML, Rangel ÉB, Benítez CG, Hamerschlak N, Pinho JR, Heller D, Eduardo FP. Reduction of SARS-CoV-2 viral load in saliva after rinsing with mouthwashes containing cetylpyridinium chloride: a randomized clinical study. PeerJ 2023; 11:e15080. [PMID: 38130922 PMCID: PMC10734404 DOI: 10.7717/peerj.15080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 02/24/2023] [Indexed: 12/23/2023] Open
Abstract
Background Symptomatic patients with COVID-19 typically have a high SARS-CoV-2 viral load in their saliva. Procedures to reduce the viral load in their oral cavity are important for mitigating the viral transmission. Methods This randomized clinical trial investigated the impact of two mouthwashes (0.075% cetylpyridinium chloride plus 0.28% zinc lactate (CPC+Zn) (n = 32), and 0.075% cetylpyridinium chloride (CPC) (n = 31)) on the viral load of SARS-CoV-2 in saliva when compared to the distilled water negative control (n = 32). Saliva was collected before (T0) and after (5 min, T1; 30 min, T2; and 60 min, T3) the intervention. Viral load in saliva was measured by qRT-PCR assays. The data in both groups was normalized for T0 and Negative Control, resulting in fold change values. Results CPC+Zn oral solution reduced the viral load in saliva by 6.34-fold at T1, 3.6-fold at T2 and 1.9-fold at T3. Rinsing with the CPC mouthwash reduced the viral load in saliva by 2.5-fold at T1, 1.9-fold at T2 and 2.0-fold at T3. Conclusion CPC+Zn mouthwash or with the CPC mouthwash reduced the viral load in saliva of COVID-19 patients immediately after rinsing. These reductions extended up to 60 min.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Debora Heller
- Hospital Israelita Albert Einstein, Sao Paulo, Brazil
- Universidade Cruzeiro do Sul, Sao Paulo, Brazil
- University of Texas at San Antonio, San Antonio, TX, United States of America
| | | |
Collapse
|
32
|
He KY, Lei XY, Zhang L, Wu DH, Li JQ, Lu LY, Laila UE, Cui CY, Xu ZX, Jian YP. Development and management of gastrointestinal symptoms in long-term COVID-19. Front Microbiol 2023; 14:1278479. [PMID: 38156008 PMCID: PMC10752947 DOI: 10.3389/fmicb.2023.1278479] [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: 08/16/2023] [Accepted: 11/20/2023] [Indexed: 12/30/2023] Open
Abstract
Background Emerging evidence reveals that SARS-CoV-2 possesses the capability to disrupt the gastrointestinal (GI) homeostasis, resulting in the long-term symptoms such as loss of appetite, diarrhea, gastroesophageal reflux, and nausea. In the current review, we summarized recent reports regarding the long-term effects of COVID-19 (long COVID) on the gastrointestine. Objective To provide a narrative review of abundant clinical evidence regarding the development and management of long-term GI symptoms in COVID-19 patients. Results Long-term persistent digestive symptoms are exhibited in a majority of long-COVID patients. SARS-CoV-2 infection of intestinal epithelial cells, cytokine storm, gut dysbiosis, therapeutic drugs, psychological factors and exacerbation of primary underlying diseases lead to long-term GI symptoms in COVID-19 patients. Interventions like probiotics, prebiotics, fecal microbiota transplantation, and antibiotics are proved to be beneficial in preserving intestinal microecological homeostasis and alleviating GI symptoms. Conclusion Timely diagnosis and treatment of GI symptoms in long-COVID patients hold great significance as they may contribute to the mitigation of severe conditions and ultimately lead to the improvement of outcomes of the patients.
Collapse
Affiliation(s)
- Kai-Yue He
- School of Life Sciences, Henan University, Kaifeng, China
| | - Xin-Yuan Lei
- School of Life Sciences, Henan University, Kaifeng, China
| | - Lei Zhang
- School of Life Sciences, Henan University, Kaifeng, China
| | - Dan-Hui Wu
- School of Life Sciences, Henan University, Kaifeng, China
| | - Jun-Qi Li
- School of Life Sciences, Henan University, Kaifeng, China
| | - Li-Yuan Lu
- School of Life Sciences, Henan University, Kaifeng, China
| | - Umm E. Laila
- School of Life Sciences, Henan University, Kaifeng, China
| | - Cui-Yun Cui
- Department of Blood Transfusion, Henan Provincial People’s Hospital, Zhengzhou, Henan, China
| | - Zhi-Xiang Xu
- School of Life Sciences, Henan University, Kaifeng, China
| | - Yong-Ping Jian
- School of Life Sciences, Henan University, Kaifeng, China
| |
Collapse
|
33
|
Ling Z, Cheng Y, Liu X, Yan X, Wu L, Shao L, Gao J, Lei W, Song Q, Zhao L, Jin G. Altered oral microbiota and immune dysfunction in Chinese elderly patients with schizophrenia: a cross-sectional study. Transl Psychiatry 2023; 13:383. [PMID: 38071192 PMCID: PMC10710460 DOI: 10.1038/s41398-023-02682-1] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 11/24/2023] [Accepted: 11/27/2023] [Indexed: 12/18/2023] Open
Abstract
Schizophrenia (SZ) is a complex psychiatric neurodevelopmental disorder with uncertain etiology and pathogenesis. Increasing evidence has recognized the key role of the gut microbiota in SZ. However, few studies have investigated the potential link between oral microbiota and SZ. We studied the tongue coating microbiota and inflammatory profiles of 118 elderly SZ patients and 97 age-matched healthy controls using Illumina MiSeq sequencing and multiplex immunoassays, respectively. Reduced α-diversity, along with a significant difference in β-diversity, were observed in patients with SZ. We have identified SZ-associated oral dysbiosis, characterized by increased Streptococcus and Fusobacterium, as well as decreased Prevotella and Veillonella. These differential genera could potentially serve as biomarkers for SZ, either alone or in combination. Additionally, an elevated Streptococcus/Prevotella ratio could indicate oral dysbiosis. These differential genera formed two distinct clusters: Streptococcus-dominated and Prevotella-dominated, which exhibited different correlations with the altered immunological profiles. Furthermore, we also observed disruptions in the inferred microbiota functions in SZ-associated microbiota, particularly in lipid and amino acid metabolism. Our study provides novel insights into the characteristics of tongue coating microbiota and its associations with immunological disturbances in elderly SZ patients, which offer new targets for the diagnosis and treatment of SZ in the elderly.
Collapse
Affiliation(s)
- Zongxin Ling
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, the First Affiliated Hospital, School of Medicine, Zhejiang University, 310003, Hangzhou, Zhejiang, China.
- Jinan Microecological Biomedicine Shandong Laboratory, 250000, Jinan, Shandong, China.
| | - Yiwen Cheng
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, the First Affiliated Hospital, School of Medicine, Zhejiang University, 310003, Hangzhou, Zhejiang, China
- Jinan Microecological Biomedicine Shandong Laboratory, 250000, Jinan, Shandong, China
| | - Xia Liu
- Department of Intensive Care Unit, the First Affiliated Hospital, School of Medicine, Zhejiang University, 310003, Hangzhou, Zhejiang, China
| | - Xiumei Yan
- Department of Laboratory Medicine, Lishui Second People's Hospital, 323000, Lishui, Zhejiang, China
| | - Lingbin Wu
- Department of Laboratory Medicine, Lishui Second People's Hospital, 323000, Lishui, Zhejiang, China
| | - Li Shao
- School of Clinical Medicine, Institute of Hepatology and Metabolic Diseases, Hangzhou Normal University, The Affiliated Hospital of Hangzhou Normal University, 310015, Hangzhou, Zhejiang, China
| | - Jie Gao
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, the First Affiliated Hospital, School of Medicine, Zhejiang University, 310003, Hangzhou, Zhejiang, China
| | - Wenhui Lei
- Jinan Microecological Biomedicine Shandong Laboratory, 250000, Jinan, Shandong, China
- School of Basic Medicine, Shandong First Medical University, 250000, Jinan, Shandong, China
| | - Qinghai Song
- Department of Psychiatry, Lishui Second People's Hospital, 323000, Lishui, Zhejiang, China
| | - Longyou Zhao
- Department of Laboratory Medicine, Lishui Second People's Hospital, 323000, Lishui, Zhejiang, China.
| | - Guolin Jin
- Department of Psychiatry, Lishui Second People's Hospital, 323000, Lishui, Zhejiang, China.
| |
Collapse
|
34
|
Zhou J, Yang X, Yang Y, Wei Y, Lu D, Xie Y, Liang H, Cui P, Ye L, Huang J. Human microbiota dysbiosis after SARS-CoV-2 infection have the potential to predict disease prognosis. BMC Infect Dis 2023; 23:841. [PMID: 38031010 PMCID: PMC10685584 DOI: 10.1186/s12879-023-08784-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Accepted: 11/02/2023] [Indexed: 12/01/2023] Open
Abstract
BACKGROUND The studies on SARS-CoV-2 and human microbiota have yielded inconsistent results regarding microbiota α-diversity and key microbiota. To address these issues and explore the predictive ability of human microbiota for the prognosis of SARS-CoV-2 infection, we conducted a reanalysis of existing studies. METHODS We reviewed the existing studies on SARS-CoV-2 and human microbiota in the Pubmed and Bioproject databases (from inception through October 29, 2021) and extracted the available raw 16S rRNA sequencing data of human microbiota. Firstly, we used meta-analysis and bioinformatics methods to reanalyze the raw data and evaluate the impact of SARS-CoV-2 on human microbial α-diversity. Secondly, machine learning (ML) was employed to assess the ability of microbiota to predict the prognosis of SARS-CoV-2 infection. Finally, we aimed to identify the key microbiota associated with SARS-CoV-2 infection. RESULTS A total of 20 studies related to SARS-CoV-2 and human microbiota were included, involving gut (n = 9), respiratory (n = 11), oral (n = 3), and skin (n = 1) microbiota. Meta-analysis showed that in gut studies, when limiting factors were studies ruled out the effect of antibiotics, cross-sectional and case-control studies, Chinese studies, American studies, and Illumina MiSeq sequencing studies, SARS-CoV-2 infection was associated with down-regulation of microbiota α-diversity (P < 0.05). In respiratory studies, SARS-CoV-2 infection was associated with down-regulation of α-diversity when the limiting factor was V4 sequencing region (P < 0.05). Additionally, the α-diversity of skin microbiota was down-regulated at multiple time points following SARS-CoV-2 infection (P < 0.05). However, no significant difference in oral microbiota α-diversity was observed after SARS-CoV-2 infection. ML models based on baseline respiratory (oropharynx) microbiota profiles exhibited the ability to predict outcomes (survival and death, Random Forest, AUC = 0.847, Sensitivity = 0.833, Specificity = 0.750) after SARS-CoV-2 infection. The shared differential Prevotella and Streptococcus in the gut, respiratory tract, and oral cavity was associated with the severity and recovery of SARS-CoV-2 infection. CONCLUSIONS SARS-CoV-2 infection was related to the down-regulation of α-diversity in the human gut and respiratory microbiota. The respiratory microbiota had the potential to predict the prognosis of individuals infected with SARS-CoV-2. Prevotella and Streptococcus might be key microbiota in SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Jie Zhou
- Guangxi Key Laboratory of AIDS Prevention and Treatment & School of Public Health, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Universities Key Laboratory of Prevention and Control of Highly Prevalent Disease, Nanning, Guangxi, China
| | - Xiping Yang
- Guangxi Key Laboratory of AIDS Prevention and Treatment & School of Public Health, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Universities Key Laboratory of Prevention and Control of Highly Prevalent Disease, Nanning, Guangxi, China
| | - Yuecong Yang
- Guangxi Key Laboratory of AIDS Prevention and Treatment & School of Public Health, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Universities Key Laboratory of Prevention and Control of Highly Prevalent Disease, Nanning, Guangxi, China
| | - Yiru Wei
- Guangxi Key Laboratory of AIDS Prevention and Treatment & School of Public Health, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Universities Key Laboratory of Prevention and Control of Highly Prevalent Disease, Nanning, Guangxi, China
| | - Dongjia Lu
- Guangxi Key Laboratory of AIDS Prevention and Treatment & School of Public Health, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Universities Key Laboratory of Prevention and Control of Highly Prevalent Disease, Nanning, Guangxi, China
| | - Yulan Xie
- Guangxi Key Laboratory of AIDS Prevention and Treatment & School of Public Health, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Universities Key Laboratory of Prevention and Control of Highly Prevalent Disease, Nanning, Guangxi, China
| | - Hao Liang
- Guangxi Key Laboratory of AIDS Prevention and Treatment & School of Public Health, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Universities Key Laboratory of Prevention and Control of Highly Prevalent Disease, Nanning, Guangxi, China
- Life Science Institute, Guangxi Medical University, Nanning, Guangxi, China
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-Constructed By the Province and Ministry, Nanning, Guangxi, China
| | - Ping Cui
- Guangxi Key Laboratory of AIDS Prevention and Treatment & School of Public Health, Guangxi Medical University, Nanning, Guangxi, China
- Life Science Institute, Guangxi Medical University, Nanning, Guangxi, China
| | - Li Ye
- Guangxi Key Laboratory of AIDS Prevention and Treatment & School of Public Health, Guangxi Medical University, Nanning, Guangxi, China.
- Guangxi Universities Key Laboratory of Prevention and Control of Highly Prevalent Disease, Nanning, Guangxi, China.
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-Constructed By the Province and Ministry, Nanning, Guangxi, China.
| | - Jiegang Huang
- Guangxi Key Laboratory of AIDS Prevention and Treatment & School of Public Health, Guangxi Medical University, Nanning, Guangxi, China.
- Guangxi Universities Key Laboratory of Prevention and Control of Highly Prevalent Disease, Nanning, Guangxi, China.
| |
Collapse
|
35
|
Lian X, Liu Z, Wu T, Lou J, Chen Y, Liu S, Jin L, Li S, Lian Y, Jiang Y, Ren Z. Oral microbiome alterations in epilepsy and after seizure control. Front Microbiol 2023; 14:1277022. [PMID: 38107849 PMCID: PMC10721976 DOI: 10.3389/fmicb.2023.1277022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Accepted: 10/31/2023] [Indexed: 12/19/2023] Open
Abstract
Background The existing diagnostic methods of epilepsy such as history collection and electroencephalogram have great limitations in practice, so more reliable and less difficult diagnostic methods are needed. Methods By characterizing oral microbiota in patients diagnosed with epilepsy (EPs) and patients whose seizures were under control (EPRs), we sought to discover biomarkers for different disease states. 16S rRNA gene sequencing was performed on 480 tongue swabs [157 EPs, 22 EPRs, and 301 healthy controls (HCs)]. Results Compared with normal individuals, patients with epilepsy exhibit increased alpha diversity in their oral microbiota, and the oral microbial communities of the two groups demonstrate significant beta diversity differences. EPs exhibit a significant increase in the abundance of 26 genera, including Streptococcus, Granulicatella, and Kluyvera, while the abundance of 14 genera, including Peptostreptococcus, Neisseria, and Schaalia, is significantly reduced. The area under the receiver operating characteristic curve (AUC) of oral microbial markers in the training cohort and validation cohort was 98.85% and 97.23%, respectively. Importantly, the AUC of the biomarker set achieved 92.44% of additional independent validation sets. In addition, EPRs also have their own unique oral community. Conclusion This study describes the characterization of the oral microbiome in EP and EPR and demonstrates the potential of the specific microbiome as a non-invasive diagnostic tool for epilepsy.
Collapse
Affiliation(s)
- Xiaolei Lian
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- The Academy of Medical Sciences of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Zhenguo Liu
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Department of Infectious Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Tianwen Wu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jiamin Lou
- Department of Infectious Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yuan Chen
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Shanshuo Liu
- Department of Infectious Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Limin Jin
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Shuang Li
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yajun Lian
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yan Jiang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhigang Ren
- Department of Infectious Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
36
|
Shen J, Wang S, Xia H, Han S, Wang Q, Wu Z, Zhuge A, Li S, Chen H, Lv L, Chen Y, Li L. Akkermansia muciniphila attenuated lipopolysaccharide-induced acute lung injury by modulating the gut microbiota and SCFAs in mice. Food Funct 2023; 14:10401-10417. [PMID: 37955584 DOI: 10.1039/d3fo04051h] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2023]
Abstract
Gut microbiota are closely related to lipopolysaccharide (LPS)-induced acute lung injury (ALI). Akkermansia muciniphila (A. muciniphila) maintains the intestinal barrier function and regulates the balance of reduced glutathione/oxidized glutathione. However, it may be useful as a treatment strategy for LPS-induced lung injury. Our study aimed to explore whether A. muciniphila could improve lung injury by affecting the gut microbiota. The administration of A. muciniphila effectively attenuated lung injury tissue damage and significantly decreased the oxidative stress and inflammatory reaction induced by LPS, with lower levels of myeloperoxidase (MDA), enhanced superoxide dismutase (SOD) activity, decreased pro-inflammatory cytokine levels, and reduced macrophage and neutrophil infiltration. Moreover, A. muciniphila maintained the intestinal barrier function, reshaped the disordered microbial community, and promoted the secretion of short-chain fatty acids (SCFAs). A. muciniphila significantly downregulated the expression of TLR2, MyD88 and NF-kappa B (P < 0.05). Butyrate supplementation demonstrated a significant improvement in the inflammatory response (P < 0.05) and mitigation of histopathological damage in mice with ALI, thereby restoring the intestinal butyric acid concentration. In conclusion, our findings indicate that A. muciniphila inhibits the accumulation of inflammatory cytokines and attenuates the activation of the TLR2/Myd88/NF-κB pathway due to exerting anti-inflammatory effects through butyrate. This study provides an experimental foundation for the potential application of A. muciniphila and butyrate in the prevention and treatment of ALI.
Collapse
Affiliation(s)
- Jian Shen
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Centre for Infectious Diseases, Collaborative Innovation Centre for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou City 310003, China.
| | - Shuting Wang
- Department of Gastroenterology, The Second Affiliated Hospital of Nanchang University, Jiangxi, Nanchang 330006, China
| | - He Xia
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Centre for Infectious Diseases, Collaborative Innovation Centre for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou City 310003, China.
| | - Shengyi Han
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Centre for Infectious Diseases, Collaborative Innovation Centre for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou City 310003, China.
| | - Qiangqiang Wang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Centre for Infectious Diseases, Collaborative Innovation Centre for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou City 310003, China.
| | - Zhengjie Wu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Centre for Infectious Diseases, Collaborative Innovation Centre for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou City 310003, China.
| | - Aoxiang Zhuge
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Centre for Infectious Diseases, Collaborative Innovation Centre for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou City 310003, China.
| | - Shengjie Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Centre for Infectious Diseases, Collaborative Innovation Centre for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou City 310003, China.
| | - Hui Chen
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Centre for Infectious Diseases, Collaborative Innovation Centre for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou City 310003, China.
| | - Longxian Lv
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Centre for Infectious Diseases, Collaborative Innovation Centre for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou City 310003, China.
| | - Yanfei Chen
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Centre for Infectious Diseases, Collaborative Innovation Centre for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou City 310003, China.
| | - Lanjuan Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Centre for Infectious Diseases, Collaborative Innovation Centre for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou City 310003, China.
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, China
| |
Collapse
|
37
|
Liang Z, Wang N, Fan C, Shang L, Zhang Y, Gao C, Luo J. Disturbance of Adaptive Immunity System Was Accompanied by a Decrease in Plasma Short-Chain Fatty Acid for Patients Hospitalized During SARS-CoV-2 Infection After COVID-19 Vaccination. J Inflamm Res 2023; 16:5261-5272. [PMID: 38026252 PMCID: PMC10656857 DOI: 10.2147/jir.s434860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 11/07/2023] [Indexed: 12/01/2023] Open
Abstract
Introduction Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection can lead to disorders of immune function and a decrease in the diversity of intestinal flora. We aimed to explore the changes of circulating immune cell subsets and the plasma level of intestinal short-chain fatty acids (SCFAs) in patients with Coronavirus disease 2019 (COVID-19), further understanding the pathogenesis of COVID-19. Methods The study included 83 newly diagnosed COVID-19 patients and 39 non-COVID-19 controls. All have completed a full course of vaccination against SARS-CoV-2. The levels of peripheral lymphocyte subsets and plasma cytokines were detected by flow cytometry. Targeted metabolomics was used to explore the level of SCFAs in plasma. Results Compared with the non-COVID-19 group, COVID-19 patients showed a decrease in CD19+B cells, CD4+T cells, CD8+T cells, NK cells, CD4+CD8+T cells and CD4-CD8-T cells (all p<0.001) and concomitantly an increase in sIL-2R, IL-6 and IL-10 (all p<0.005). These alterations were more pronounced in those critical patients. In addition, COVID-19 patients had lower levels of propanoic acid (PA), butyric acid (BA), isobutyric acid (IBA) and isohexanoic acid (ICA) (all p<0.01). Among them, the level of ICA is positively correlated with the absolute number of immune cells. Conclusion Our study suggests the immune cell subsets in COVID-19 patients who had completed vaccination were still severely disturbed and concomitantly lower SCFAs, especially in severe patients with poor prognosis. Lower levels of plasma SCFAs may contribute to lymphopenia in COVID-19. The potential relationship between plasma SCFAs and immune cell reduction provides a new direction for the treatment of COVID-19.
Collapse
Affiliation(s)
- Zhaojun Liang
- Division of Rheumatology, Department of Medicine, the Second Hospital of Shanxi Medical University, Taiyuan, People’s Republic of China
- Shanxi Key Laboratory for Immunomicroecology, the Second Hospital of Shanxi Medical University, Taiyuan, People’s Republic of China
| | - Nan Wang
- Division of Rheumatology, Department of Medicine, the Second Hospital of Shanxi Medical University, Taiyuan, People’s Republic of China
- Shanxi Key Laboratory for Immunomicroecology, the Second Hospital of Shanxi Medical University, Taiyuan, People’s Republic of China
| | - Chunxue Fan
- Division of Rheumatology, Department of Medicine, the Second Hospital of Shanxi Medical University, Taiyuan, People’s Republic of China
- Shanxi Key Laboratory for Immunomicroecology, the Second Hospital of Shanxi Medical University, Taiyuan, People’s Republic of China
| | - Lili Shang
- Division of Rheumatology, Department of Medicine, the Second Hospital of Shanxi Medical University, Taiyuan, People’s Republic of China
- Shanxi Key Laboratory for Immunomicroecology, the Second Hospital of Shanxi Medical University, Taiyuan, People’s Republic of China
| | - Yaping Zhang
- Division of Rheumatology, Department of Medicine, the Second Hospital of Shanxi Medical University, Taiyuan, People’s Republic of China
- Shanxi Key Laboratory for Immunomicroecology, the Second Hospital of Shanxi Medical University, Taiyuan, People’s Republic of China
| | - Chong Gao
- Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Jing Luo
- Division of Rheumatology, Department of Medicine, the Second Hospital of Shanxi Medical University, Taiyuan, People’s Republic of China
- Shanxi Key Laboratory for Immunomicroecology, the Second Hospital of Shanxi Medical University, Taiyuan, People’s Republic of China
| |
Collapse
|
38
|
Krasaewes K, Chaiwarith R, Chattipakorn N, Chattipakorn SC. Profiles of gut microbiota associated with clinical outcomes in patients with different stages of SARS-CoV-2 infection. Life Sci 2023; 332:122136. [PMID: 37783267 DOI: 10.1016/j.lfs.2023.122136] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Revised: 08/29/2023] [Accepted: 09/29/2023] [Indexed: 10/04/2023]
Abstract
The correlation between SARS-CoV-2 infection and gut microbiota has been a subject of growing interest in recent research endeavors. It is postulated that SARS-CoV-2 might lead to gut dysbiosis by affecting the gut-lung axis and reducing the production of antimicrobial peptides in the gastrointestinal tract. Our comprehensive review of both in vivo and clinical studies has revealed a consistent decline in alpha diversity and increased dissimilarity in beta diversity of gut microbiota in comparison to healthy populations, observed during both the acute and post-infection phases of COVID-19. Furthermore, there is a notable reduction in the number of beneficial butyrate-producing bacteria, alongside an upsurge in opportunistic bacteria. Concomitantly, the functional and metabolic characteristics of gut microbiota are significantly altered. Consequently, COVID-19 patients exhibit a heightened inflammatory state, which has been linked to the severity of the disease in the acute phase and the occurrence of post-acute COVID-19 syndrome (PACS) in the post-infection phase. Notably, certain specific gut microbiota species have emerged as potential candidates for aiding in the diagnosis, prediction of disease severity, or treatment of severe cases of COVID-19. This review also underscores the significance of gut microbiota in the context of post-acute COVID-19 syndrome (PACS) and offers valuable insights into possible biomarkers for diagnosis and therapeutic targets for PACS in the future.
Collapse
Affiliation(s)
- Kawisara Krasaewes
- Division of Infectious Disease and Tropical Medicine, Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Romanee Chaiwarith
- Division of Infectious Disease and Tropical Medicine, Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Nipon Chattipakorn
- Neuroelectrophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Siriporn C Chattipakorn
- Neuroelectrophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand; Department of Oral Biology and Diagnostic Sciences, Faculty of Dentistry, Chiang Mai University, Chiang Mai 50200, Thailand.
| |
Collapse
|
39
|
Liu S, Zhao Y, Feng X, Xu H. SARS-CoV-2 infection threatening intestinal health: A review of potential mechanisms and treatment strategies. Crit Rev Food Sci Nutr 2023; 63:12578-12596. [PMID: 35894645 DOI: 10.1080/10408398.2022.2103090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The outbreak of the COVID-19 pandemic has brought great problems to mankind, including economic recession and poor health. COVID-19 patients are frequently reported with gastrointestinal symptoms such as diarrhea and vomiting in clinical diagnosis. Maintaining intestinal health is the key guarantee to maintain the normal function of multiple organs, otherwise it will be a disaster. Therefore, the purpose of this review was deeply understanded the potential mechanism of SARS-CoV-2 infection threatening intestinal health and put forward reasonable treatment strategies. Combined with the existing researches, we summarized the mechanism of SARS-CoV-2 infection threatening intestinal health, including intestinal microbiome disruption, intestinal barrier dysfunction, intestinal oxidative stress and intestinal cytokine storm. These adverse intestinal events may affect other organs through the circulatory system or aggravate the course of the disease. Typically, intestinal disadvantage may promote the progression of SARS-CoV-2 through the gut-lung axis and increase the disease degree of COVID-19 patients. In view of the lack of specific drugs to inhibit SARS-CoV-2 replication, the current review described new strategies of probiotics, prebiotics, postbiotics and nutrients to combat SARS-CoV-2 infection and maintain intestinal health. To provide new insights for the prevention and treatment of gastrointestinal symptoms and pneumonia in patients with COVID-19.
Collapse
Affiliation(s)
- Shanji Liu
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, China
| | - Yu Zhao
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, China
| | - Xiaoyan Feng
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, China
| | - Hengyi Xu
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, China
| |
Collapse
|
40
|
Abdulan IM, Feller V, Oancea A, Maștaleru A, Alexa AI, Negru R, Cumpăt CM, Leon MM. Evolution of Cardiovascular Risk Factors in Post-COVID Patients. J Clin Med 2023; 12:6538. [PMID: 37892676 PMCID: PMC10607829 DOI: 10.3390/jcm12206538] [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: 08/17/2023] [Revised: 10/01/2023] [Accepted: 10/13/2023] [Indexed: 10/29/2023] Open
Abstract
(1) Background: SARS-CoV-2 infection has been a subject of extensive discussion in the medical field, particularly in relation to the risk factors and effective treatment strategies for reducing the negative health outcomes associated with the virus. However, researchers indicate that individuals in the recovery phase after COVID-19 experience a range of symptoms that significantly impact their overall well-being and quality of life. At present, there is insufficient evidence to substantiate the claim that patients in the post-acute phase of COVID-19 are at an elevated risk of developing new-onset hypertension or even metabolic syndrome. The current study aimed to assess the risk of cardiovascular diseases after COVID-19 and the optimal treatment of these conditions. (2) Methods: This research was conducted at the Cardiovascular Rehabilitation Clinic of the Iasi Clinical Rehabilitation Hospital (Romania) between the 1st of September and 31st of December 2022. From a total of 551 patients hospitalized in that period, 70 patients with multiple comorbidities were selected. This study included patients over 18 years old who were diagnosed with COVID-19 within the past 30 days. (3) Results: The included patients were mostly women (62.9%) from the urban area (61.4%). Comparing the post-COVID-19 period to the pre-COVID-19 one, it was observed that the risk of hypertension increased from 69.57% to 90% among the subjects (p = 0.005). Risk factors for the new onset of hypertension were identified as age, female gender, and an elevated body mass index. Moreover, the number of patients with dyslipidemia doubled, and a higher body mass index was noted. (4) Conclusions: Our findings suggest that patients affected by COVID-19 are at an increased risk of developing hypertension and related disorders.
Collapse
Affiliation(s)
- Irina Mihaela Abdulan
- Department of Medical Specialties I, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (I.M.A.); (R.N.); (C.M.C.); (M.M.L.)
- Clinical Rehabilitation Hospital, 700661 Iasi, Romania;
| | | | - Andra Oancea
- Department of Medical Specialties I, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (I.M.A.); (R.N.); (C.M.C.); (M.M.L.)
- Clinical Rehabilitation Hospital, 700661 Iasi, Romania;
| | - Alexandra Maștaleru
- Department of Medical Specialties I, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (I.M.A.); (R.N.); (C.M.C.); (M.M.L.)
- Clinical Rehabilitation Hospital, 700661 Iasi, Romania;
| | - Anisia Iuliana Alexa
- Department of Surgery II, Discipline of Ophthalmology, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania;
| | - Robert Negru
- Department of Medical Specialties I, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (I.M.A.); (R.N.); (C.M.C.); (M.M.L.)
| | - Carmen Marinela Cumpăt
- Department of Medical Specialties I, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (I.M.A.); (R.N.); (C.M.C.); (M.M.L.)
- Clinical Rehabilitation Hospital, 700661 Iasi, Romania;
| | - Maria Magdalena Leon
- Department of Medical Specialties I, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (I.M.A.); (R.N.); (C.M.C.); (M.M.L.)
- Clinical Rehabilitation Hospital, 700661 Iasi, Romania;
| |
Collapse
|
41
|
Huang G, Mao Y, Zhang W, Luo Q, Xie R, Huang D, Liang Y. Explore the changes of intestinal flora in patients with coronavirus disease 2019 based on bioinformatics. Front Cell Infect Microbiol 2023; 13:1265028. [PMID: 37900316 PMCID: PMC10611479 DOI: 10.3389/fcimb.2023.1265028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Accepted: 09/26/2023] [Indexed: 10/31/2023] Open
Abstract
Background Studies have revealed that there were significant changes in intestinal flora composition in patients with coronavirus disease 2019 (COVID-19) compared to non-COVID-19 patients, regardless of whether they were treated with medication. Therefore, a comprehensive study of the intestinal flora of COVID-19 patients is needed to further understand the mechanisms of COVID-19 development. Methods In total, 20 healthy samples and 20 COVID-19 samples were collected in this study. Firstly, alpha diversity and beta diversity were analyzed to assess whether there were difference in species richness and diversity as well as species composition between COVID-19 and control groups. The observed features index, Evenness index, PD index, and Shannon index were utilized to measure alpha diversity. The principal coordinates analysis (PCoA) and non-metric multidimensional scaling (NMDS) were performed to analyzed beta diversity. Linear discriminant analysis Effect Size (LEfSe) was utilized to analyze the variability in the abundance of bacterial taxa from different classification levels. The random forest (RF), Least absolute shrinkage and selection operator (LASSO), and univariate logistic regression were utilized to identify key Amplicon Sequence Variant (ASVs). Finally, the relevant networks of bacterial taxa were created in COVID-19 and control groups, separately. Results There were more species in the control group than in COVID-19 group. The observed features index, Shannon index, and Evenness index in the control groups were markedly higher than in the COVID-19 group. Therefore, there were marked variations in bacterial taxa composition between the COVID-19 and control groups. The nine bacterial taxa were significantly more abundant in the COVID-19 group, such as g-Streptococcus, f-Streptococcaceae, o-Lactobacillales, c-Bacilli and so on. In the control group, 26 bacterial taxa were significantly more abundant, such as c-Clostrjdia, o-Oscillospirales, f-Ruminococcaceae, etc. The 5 key ASVs were obtained through taking the intersection of the characteristic ASVs obtained by the three algorithms, namely ASV6, ASV53, ASV92, ASV96, and ASV105, which had diagnostic value for COVID-19. The relevance network in the control group was more complex compared to the COVID-19 group. Conclusion Our findings provide five key ASVs for diagnosis of COVID-19, providing a scientific reference for further studies of COVID-19.
Collapse
Affiliation(s)
- Gangding Huang
- Department of Gastroenterology, the Fifth Affiliated Hospital of Guangxi Medical University, Nanning, China
| | | | | | | | | | | | | |
Collapse
|
42
|
Meslier V, Menozzi E, David A, Morabito C, Lucas Del Pozo S, Famechon A, North J, Quinquis B, Koletsi S, Macnaughtan J, Mezabrovschi R, Ehrlich SD, Schapira AHV, Almeida M. Evaluation of an Adapted Semi-Automated DNA Extraction for Human Salivary Shotgun Metagenomics. Biomolecules 2023; 13:1505. [PMID: 37892187 PMCID: PMC10604855 DOI: 10.3390/biom13101505] [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: 08/29/2023] [Revised: 09/29/2023] [Accepted: 10/05/2023] [Indexed: 10/29/2023] Open
Abstract
Recent attention has highlighted the importance of oral microbiota in human health and disease, e.g., in Parkinson's disease, notably using shotgun metagenomics. One key aspect for efficient shotgun metagenomic analysis relies on optimal microbial sampling and DNA extraction, generally implementing commercial solutions developed to improve sample collection and preservation, and provide high DNA quality and quantity for downstream analysis. As metagenomic studies are today performed on a large number of samples, the next evolution to increase study throughput is with DNA extraction automation. In this study, we proposed a semi-automated DNA extraction protocol for human salivary samples collected with a commercial kit, and compared the outcomes with the DNA extraction recommended by the manufacturer. While similar DNA yields were observed between the protocols, our semi-automated DNA protocol generated significantly higher DNA fragment sizes. Moreover, we showed that the oral microbiome composition was equivalent between DNA extraction methods, even at the species level. This study demonstrates that our semi-automated protocol is suitable for shotgun metagenomic analysis, while allowing for improved sample treatment logistics with reduced technical variability and without compromising the structure of the oral microbiome.
Collapse
Affiliation(s)
- Victoria Meslier
- MetaGenoPolis, INRAE, Université Paris-Saclay, 78350 Jouy-en-Josas, France (C.M.)
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA (S.K.); (R.M.); (S.D.E.)
| | - Elisa Menozzi
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA (S.K.); (R.M.); (S.D.E.)
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, University College London (UCL), London WC1E 6BT, UK
| | - Aymeric David
- MetaGenoPolis, INRAE, Université Paris-Saclay, 78350 Jouy-en-Josas, France (C.M.)
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA (S.K.); (R.M.); (S.D.E.)
| | - Christian Morabito
- MetaGenoPolis, INRAE, Université Paris-Saclay, 78350 Jouy-en-Josas, France (C.M.)
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA (S.K.); (R.M.); (S.D.E.)
| | - Sara Lucas Del Pozo
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA (S.K.); (R.M.); (S.D.E.)
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, University College London (UCL), London WC1E 6BT, UK
| | - Alexandre Famechon
- MetaGenoPolis, INRAE, Université Paris-Saclay, 78350 Jouy-en-Josas, France (C.M.)
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA (S.K.); (R.M.); (S.D.E.)
| | - Janet North
- Research Department of Hematology, Cancer Institute, University College London (UCL), London WC1E 6BT, UK
| | - Benoit Quinquis
- MetaGenoPolis, INRAE, Université Paris-Saclay, 78350 Jouy-en-Josas, France (C.M.)
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA (S.K.); (R.M.); (S.D.E.)
| | - Sofia Koletsi
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA (S.K.); (R.M.); (S.D.E.)
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, University College London (UCL), London WC1E 6BT, UK
| | - Jane Macnaughtan
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA (S.K.); (R.M.); (S.D.E.)
- Liver Failure Group, Institute for Liver and Digestive Health, University College London, London WC1E 6BT, UK
| | - Roxana Mezabrovschi
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA (S.K.); (R.M.); (S.D.E.)
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, University College London (UCL), London WC1E 6BT, UK
| | - S. Dusko Ehrlich
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA (S.K.); (R.M.); (S.D.E.)
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, University College London (UCL), London WC1E 6BT, UK
| | - Anthony H. V. Schapira
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA (S.K.); (R.M.); (S.D.E.)
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, University College London (UCL), London WC1E 6BT, UK
| | - Mathieu Almeida
- MetaGenoPolis, INRAE, Université Paris-Saclay, 78350 Jouy-en-Josas, France (C.M.)
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA (S.K.); (R.M.); (S.D.E.)
| |
Collapse
|
43
|
Brzychczy- Sroka B, Talaga-Ćwiertnia K, Sroka-Oleksiak A, Gurgul A, Zarzecka-Francica E, Ostrowski W, Kąkol J, Zarzecka J, Brzychczy-Włoch M. Oral microbiota study of the patients after hospitalisation for COVID-19, considering selected dental indices and antibiotic therapy using the next generation sequencing method (NGS). J Oral Microbiol 2023; 15:2264591. [PMID: 37840855 PMCID: PMC10569355 DOI: 10.1080/20002297.2023.2264591] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 09/24/2023] [Indexed: 10/17/2023] Open
Abstract
Background Poor oral hygiene and the increased incidence and severity of periodontitis may exacerbate SARS-CoV-2 infection. The aim was to evaluate the oral microbiota of 60 participants divided into groups: COVID-19 convalescents who received antibiotics during hospitalization (I), COVID-19 convalescents without antibiotic therapy (II) and healthy individuals (III). Materials and Methods Dental examination was conducted, and oral health status was evaluated using selected dental indexes. Clinical samples (saliva, dorsal swabs, supragingival and subgingival plaque) were collected and used for metagenomic library to the next-generation sequencing (NGS) preparation. Results Each of the clinical materials in particular groups of patients showed a statistically significant and quantitatively different bacterial composition. Patients from group I showed significantly worse oral health, reflected by higher average values of dental indexes and also a higher percentage of Veillonella, Tannerella, Capnocytophaga and Selenomonas genera in comparison to other groups. Additionally, a statistically significant decrease in the amount of Akkermansia type in both groups with COVID-19 was observed for all materials. Conclusions The primary factor affecting the composition of oral microbiota was not the SARS-CoV-2 infection itself, but the use of antibiotic therapy. The increased percentage of pro-inflammatory pathogens observed in COVID-19 patients underscores the importance of preventing periodontal disease and improving oral hygiene in the future.
Collapse
Affiliation(s)
- Barbara Brzychczy- Sroka
- Department of Conservative Dentistry with Endodontics, Institute of Dentistry, Jagiellonian University Medical College, Kraków, Poland
| | - Katarzyna Talaga-Ćwiertnia
- Department of Molecular Medical Microbiology, Chair of Microbiology, Faculty of Medicine, Jagiellonian University Medical College, Kraków, Poland
| | - Agnieszka Sroka-Oleksiak
- Department of Molecular Medical Microbiology, Chair of Microbiology, Faculty of Medicine, Jagiellonian University Medical College, Kraków, Poland
| | - Artur Gurgul
- Center for Experimental and Innovative Medicine, The University of Agriculture in Kraków, Kraków, Poland
| | - Elżbieta Zarzecka-Francica
- Department of Prosthodontics and Orthodontics, Institute of Dentistry, Jagiellonian University Medical College, Kraków, Poland
| | - Wojciech Ostrowski
- Department of Conservative Dentistry with Endodontics, Institute of Dentistry, Jagiellonian University Medical College, Kraków, Poland
| | | | - Joanna Zarzecka
- Department of Conservative Dentistry with Endodontics, Institute of Dentistry, Jagiellonian University Medical College, Kraków, Poland
| | - Monika Brzychczy-Włoch
- Department of Molecular Medical Microbiology, Chair of Microbiology, Faculty of Medicine, Jagiellonian University Medical College, Kraków, Poland
| |
Collapse
|
44
|
Jangi S. Importance of Diversity: Inclusion of Duodenal Sampling Enhances the Study of Microbial Composition During the COVID-19 Pandemic. Dig Dis Sci 2023; 68:3838-3840. [PMID: 37578564 DOI: 10.1007/s10620-023-08067-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/26/2023] [Indexed: 08/15/2023]
|
45
|
Albóniga OE, Moreno E, Martínez-Sanz J, Vizcarra P, Ron R, Díaz-Álvarez J, Rosas Cancio-Suarez M, Sánchez-Conde M, Galán JC, Angulo S, Moreno S, Barbas C, Serrano-Villar S. Differential abundance of lipids and metabolites related to SARS-CoV-2 infection and susceptibility. Sci Rep 2023; 13:15124. [PMID: 37704651 PMCID: PMC10500013 DOI: 10.1038/s41598-023-40999-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 08/20/2023] [Indexed: 09/15/2023] Open
Abstract
The mechanisms driving SARS-CoV-2 susceptibility remain poorly understood, especially the factors determining why unvaccinated individuals remain uninfected despite high-risk exposures. To understand lipid and metabolite profiles related with COVID-19 susceptibility and disease progression. We collected samples from an exceptional group of unvaccinated healthcare workers heavily exposed to SARS-CoV-2 but not infected ('non-susceptible') and subjects who became infected during the follow-up ('susceptible'), including non-hospitalized and hospitalized patients with different disease severity providing samples at early disease stages. Then, we analyzed their plasma metabolomic profiles using mass spectrometry coupled with liquid and gas chromatography. We show specific lipids profiles and metabolites that could explain SARS-CoV-2 susceptibility and COVID-19 severity. More importantly, non-susceptible individuals show a unique lipidomic pattern characterized by the upregulation of most lipids, especially ceramides and sphingomyelin, which could be interpreted as markers of low susceptibility to SARS-CoV-2 infection. This study strengthens the findings of other researchers about the importance of studying lipid profiles as relevant markers of SARS-CoV-2 pathogenesis.
Collapse
Affiliation(s)
- Oihane E Albóniga
- Centro de Metabolómica y Bioanálisis (CEMBIO), Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, 28660, Madrid, Spain
| | - Elena Moreno
- Department of Infectious Diseases, Hospital Universitario Ramón y Cajal, IRYCIS, 28034, Madrid, Spain
- CIBERINFEC, Instituto de Salud Carlos III, Madrid, Spain
| | - Javier Martínez-Sanz
- Department of Infectious Diseases, Hospital Universitario Ramón y Cajal, IRYCIS, 28034, Madrid, Spain
- CIBERINFEC, Instituto de Salud Carlos III, Madrid, Spain
| | - Pilar Vizcarra
- Department of Infectious Diseases, Hospital Universitario Ramón y Cajal, IRYCIS, 28034, Madrid, Spain
- CIBERINFEC, Instituto de Salud Carlos III, Madrid, Spain
| | - Raquel Ron
- Department of Infectious Diseases, Hospital Universitario Ramón y Cajal, IRYCIS, 28034, Madrid, Spain
- CIBERINFEC, Instituto de Salud Carlos III, Madrid, Spain
| | - Jorge Díaz-Álvarez
- Department of Infectious Diseases, Hospital Universitario Ramón y Cajal, IRYCIS, 28034, Madrid, Spain
- CIBERINFEC, Instituto de Salud Carlos III, Madrid, Spain
| | - Marta Rosas Cancio-Suarez
- Department of Infectious Diseases, Hospital Universitario Ramón y Cajal, IRYCIS, 28034, Madrid, Spain
- CIBERINFEC, Instituto de Salud Carlos III, Madrid, Spain
| | - Matilde Sánchez-Conde
- Department of Infectious Diseases, Hospital Universitario Ramón y Cajal, IRYCIS, 28034, Madrid, Spain
- CIBERINFEC, Instituto de Salud Carlos III, Madrid, Spain
| | - Juan Carlos Galán
- Department of Microbiology, Hospital Universitario Ramón y Cajal, IRYCIS, 28034, Madrid, Spain
- CIBERESP, Instituto de Salud Carlos III, Madrid, Spain
| | - Santiago Angulo
- Centro de Metabolómica y Bioanálisis (CEMBIO), Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, 28660, Madrid, Spain
| | - Santiago Moreno
- Department of Infectious Diseases, Hospital Universitario Ramón y Cajal, IRYCIS, 28034, Madrid, Spain
- CIBERINFEC, Instituto de Salud Carlos III, Madrid, Spain
| | - Coral Barbas
- Centro de Metabolómica y Bioanálisis (CEMBIO), Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, 28660, Madrid, Spain
| | - Sergio Serrano-Villar
- Department of Infectious Diseases, Hospital Universitario Ramón y Cajal, IRYCIS, 28034, Madrid, Spain.
- CIBERINFEC, Instituto de Salud Carlos III, Madrid, Spain.
- Department of Infectious Diseases, Hospital Universitario Ramon y Cajal, Facultad de Medicina, Universidad de Alcalá (IRYCIS), Carretera de Colmenar Viejo, Km 9.100, 28034, Madrid, Spain.
| |
Collapse
|
46
|
Liang X, Wang Q, Liu J, Ma J, Zhang Y, Wang M, Yu Y, Wang L. Coinfection of SARS-CoV-2 and influenza A (H3N2) detected in bronchoalveolar lavage fluid of a patient with long COVID using metagenomic next-generation sequencing: a case report. Front Cell Infect Microbiol 2023; 13:1224794. [PMID: 37724290 PMCID: PMC10505437 DOI: 10.3389/fcimb.2023.1224794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Accepted: 08/16/2023] [Indexed: 09/20/2023] Open
Abstract
The growing number of long COVID cases has drawn clinical attention to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), which has been spreading worldwide since winter 2019. Its symptoms are not limited to fatigue and shortness of breath but also affect daily life. We report the use of metagenomic next-generation sequencing (mNGS) to detect coinfection with SARS-CoV-2 and influenza A virus in a patient with long COVID. The patient was admitted with fever, expectoration, fatigue, and shortness of breath. The PCR test was negative due to possible clearance of SARS-Cov-2 in the upper respiratory tract of patients with long COVID. Other routine microbiological tests were also negative, making the clinical diagnosis difficult. Bronchoalveolar lavage fluid (BALF) samples were tested using mNGS. The patient was diagnosed and treated promptly, recovered quickly, and continued taking azvudine after discharge; his condition was stable. This study illustrates that mNGS may be valuable for the timely diagnosis of patients with long COVID and their mixed infections.
Collapse
Affiliation(s)
- Xuefei Liang
- Department of Respiratory and Critical Care Medicine, Sinopharm North Hospital, Baotou, China
| | - Qiushi Wang
- Infection Business Unit, Tianjin Novogene Med LAB Co., Ltd., Tianjin, China
- Infection Business Unit, Novogene Co., Ltd., Beijing, China
| | - Jia Liu
- Infection Business Unit, Tianjin Novogene Med LAB Co., Ltd., Tianjin, China
- Infection Business Unit, Novogene Co., Ltd., Beijing, China
| | - Jing Ma
- Department of Respiratory and Critical Care Medicine, Sinopharm North Hospital, Baotou, China
| | - Yajuan Zhang
- Department of Respiratory and Critical Care Medicine, Sinopharm North Hospital, Baotou, China
| | - Meng Wang
- Infection Business Unit, Tianjin Novogene Med LAB Co., Ltd., Tianjin, China
- Infection Business Unit, Novogene Co., Ltd., Beijing, China
| | - Yang Yu
- Infection Business Unit, Tianjin Novogene Med LAB Co., Ltd., Tianjin, China
- Infection Business Unit, Novogene Co., Ltd., Beijing, China
| | - Linlin Wang
- Department of Respiratory and Critical Care Medicine, Sinopharm North Hospital, Baotou, China
| |
Collapse
|
47
|
Zhong MM, Xie JH, Feng Y, Zhang SH, Xia JN, Tan L, Chen NX, Su XL, Zhang Q, Feng YZ, Guo Y. Causal effects of the gut microbiome on COVID-19 susceptibility and severity: a two-sample Mendelian randomization study. Front Immunol 2023; 14:1173974. [PMID: 37720222 PMCID: PMC10502427 DOI: 10.3389/fimmu.2023.1173974] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 08/10/2023] [Indexed: 09/19/2023] Open
Abstract
Background The coronavirus disease 2019 (COVID-19) caused a global pandemic, with potential severity. We aimed to investigate whether genetically predicted gut microbiome is associated with susceptibility and severity of COVID-19 risk. Methods Mendelian randomization (MR) analysis of two sets with different significance thresholds was carried out to infer the causal relationship between the gut microbiome and COVID-19. SNPs associated with the composition of the gut microbiome (n = 5,717,754) and with COVID-19 susceptibility (n = 14,328,058), COVID-19 severity (n = 11,707,239), and COVID-19 hospitalization (n = 12,018,444) from publicly available genome-wide association studies (GWAS). The random-effect inverse variance weighted (IVW) method was used to determine causality. Three more MR techniques-MR Egger, weighted median, and weighted mode-and a thorough sensitivity analysis were also used to confirm the findings. Results IVW showed that 18 known microbial taxa were causally associated with COVID-19. Among them, six microbial taxa were causally associated with COVID-19 susceptibility; seven microbial taxa were causally associated with COVID-19 severity ; five microbial taxa were causally associated with COVID-19 hospitalization. Sensitivity analyses showed no evidence of pleiotropy or heterogeneity. Then, the predicted 37 species of the gut microbiome deserve further study. Conclusion This study found that some microbial taxa were protective factors or risky factors for COVID-19, which may provide helpful biomarkers for asymptomatic diagnosis and potential therapeutic targets for COVID-19.
Collapse
Affiliation(s)
- Meng-Mei Zhong
- Department of Stomatology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jia-Hao Xie
- Institute of Artificial Intelligence & Robotics (IAIR), Key Laboratory of Traffic Safety on Track of Ministry of Education, School of Traffic and Transportation Engineering, Central South University, Changsha, Hunan, China
| | - Yao Feng
- Department of Stomatology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Shao-Hui Zhang
- Department of Stomatology, Xiangyang Central Hospital, Xiangyang, Hubei, China
| | - Jiang-Nan Xia
- School of Architecture and Art, Central South University, Changsha, Hunan, China
| | - Li Tan
- Department of Stomatology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Ning-Xin Chen
- Department of Stomatology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xiao-Lin Su
- Department of Stomatology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Qian Zhang
- Department of Stomatology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yun-Zhi Feng
- Department of Stomatology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yue Guo
- Department of Stomatology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
48
|
Larios Serrato V, Meza B, Gonzalez-Torres C, Gaytan-Cervantes J, González Ibarra J, Santacruz Tinoco CE, Anguiano Hernández YM, Martínez Miguel B, Cázarez Cortazar A, Sarquiz Martínez B, Alvarado Yaah JE, Mendoza Pérez AR, Palma Herrera JJ, García Soto LM, Chávez Rojas AI, Bravo Mateos G, Samano Marquez G, Grajales Muñiz C, Torres J. Diversity, composition, and networking of saliva microbiota distinguish the severity of COVID-19 episodes as revealed by an analysis of 16S rRNA variable V1-V3 region sequences. mSystems 2023; 8:e0106222. [PMID: 37310423 PMCID: PMC10470033 DOI: 10.1128/msystems.01062-22] [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: 11/02/2022] [Accepted: 04/17/2023] [Indexed: 06/14/2023] Open
Abstract
Studies on the role of the oral microbiome in SARS-CoV-2 infection and severity of the disease are limited. We aimed to characterize the bacterial communities present in the saliva of patients with varied COVID-19 severity to learn if there are differences in the characteristics of the microbiome among the clinical groups. We included 31 asymptomatic subjects with no previous COVID-19 infection or vaccination; 176 patients with mild respiratory symptoms, positive or negative for SARS-CoV-2 infection; 57 patients that required hospitalization because of severe COVID-19 with oxygen saturation below 92%, and 18 fatal cases of COVID-19. Saliva samples collected before any treatment were tested for SARS-CoV-2 by PCR. Oral microbiota in saliva was studied by amplification and sequencing of the V1-V3 variable regions of 16S gene using an Illumina MiSeq platform. We found significant changes in diversity, composition, and networking in saliva microbiota of patients with COVID-19, as well as patterns associated with severity of disease. The presence or abundance of several commensal species and opportunistic pathogens were associated with each clinical stage. Patterns of networking were also found associated with severity of disease: a highly regulated bacterial community (normonetting) was found in healthy people whereas poorly regulated populations (disnetting) were characteristic of severe cases. Characterization of microbiota in saliva may offer important clues in the pathogenesis of COVID-19 and may also identify potential markers for prognosis in the severity of the disease. IMPORTANCE SARS-CoV-2 infection is the most severe pandemic of humankind in the last hundred years. The outcome of the infection ranges from asymptomatic or mild to severe and even fatal cases, but reasons for this remain unknown. Microbes normally colonizing the respiratory tract form communities that may mitigate the transmission, symptoms, and severity of viral infections, but very little is known on the role of these microbial communities in the severity of COVID-19. We aimed to characterize the bacterial communities in saliva of patients with different severity of COVID-19 disease, from mild to fatal cases. Our results revealed clear differences in the composition and in the nature of interactions (networking) of the bacterial species present in the different clinical groups and show community-patterns associated with disease severity. Characterization of the microbial communities in saliva may offer important clues to learn ways COVID-19 patients may suffer from different disease severities.
Collapse
Affiliation(s)
- Violeta Larios Serrato
- Departamento de Bioquímica, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, México, Mexico
| | - Beatriz Meza
- Universidad Autónoma de Baja California Sur, La Paz, Baja California Sur, Mexico
- Centro de Investigaciones Biológicas del Noroeste SC, La Paz, Baja California Sur, Mexico
- Unidad de Investigación Médica en Enfermedades Infecciosas, UMAE Pediatría, Centro Médico Nacional SXXI, IMSS, Torreón, Mexico
| | | | - Javier Gaytan-Cervantes
- Laboratorio de Secuenciación, División de Desarrollo de la Investigación, IMSS, Torreón, Mexico
| | - Joaquín González Ibarra
- División de Desarrollo de la Investigación en Salud, Coordinación de Investigación en Salud, IMSS, Torreón, Mexico
| | - Clara Esperanza Santacruz Tinoco
- División de Laboratorios Especializados, Coordinación de Calidad de Insumos y Laboratorios Especializados, IMSS, Torreón, Mexico
| | - Yu-Mei Anguiano Hernández
- División de Laboratorios Especializados, Coordinación de Calidad de Insumos y Laboratorios Especializados, IMSS, Torreón, Mexico
| | - Bernardo Martínez Miguel
- División de Laboratorios Especializados, Coordinación de Calidad de Insumos y Laboratorios Especializados, IMSS, Torreón, Mexico
| | - Allison Cázarez Cortazar
- División de Laboratorios Especializados, Coordinación de Calidad de Insumos y Laboratorios Especializados, IMSS, Torreón, Mexico
| | - Brenda Sarquiz Martínez
- División de Laboratorios Especializados, Coordinación de Calidad de Insumos y Laboratorios Especializados, IMSS, Torreón, Mexico
| | - Julio Elias Alvarado Yaah
- División de Laboratorios Especializados, Coordinación de Calidad de Insumos y Laboratorios Especializados, IMSS, Torreón, Mexico
| | | | | | | | | | | | | | | | - Javier Torres
- Unidad de Investigación Médica en Enfermedades Infecciosas, UMAE Pediatría, Centro Médico Nacional SXXI, IMSS, Torreón, Mexico
| |
Collapse
|
49
|
Reuben RC, Beugnon R, Jurburg SD. COVID-19 alters human microbiomes: a meta-analysis. Front Cell Infect Microbiol 2023; 13:1211348. [PMID: 37600938 PMCID: PMC10433767 DOI: 10.3389/fcimb.2023.1211348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 06/23/2023] [Indexed: 08/22/2023] Open
Abstract
Introduction Severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) has infected a substantial portion of the world's population, and novel consequences of COVID-19 on the human body are continuously being uncovered. The human microbiome plays an essential role in host health and well-being, and multiple studies targeting specific populations have reported altered microbiomes in patients infected with SARS-CoV-2. Given the global scale and massive incidence of COVID on the global population, determining whether the effects of COVID-19 on the human microbiome are consistent and generalizable across populations is essential. Methods We performed a synthesis of human microbiome responses to COVID-19. We collected 16S rRNA gene amplicon sequence data from 11 studies sampling the oral and nasopharyngeal or gut microbiome of COVID-19-infected and uninfected subjects. Our synthesis included 1,159 respiratory (oral and nasopharyngeal) microbiome samples and 267 gut microbiome samples from patients in 11 cities across four countries. Results Our reanalyses revealed communitywide alterations in the respiratory and gut microbiomes across human populations. We found significant overall reductions in the gut microbial diversity of COVID-19-infected patients, but not in the respiratory microbiome. Furthermore, we found more consistent community shifts in the gut microbiomes of infected patients than in the respiratory microbiomes, although the microbiomes in both sites exhibited higher host-to-host variation in infected patients. In respiratory microbiomes, COVID-19 infection resulted in an increase in the relative abundance of potentially pathogenic bacteria, including Mycoplasma. Discussion Our findings shed light on the impact of COVID-19 on the human-associated microbiome across populations, and highlight the need for further research into the relationship between long-term effects of COVID-19 and altered microbiota.
Collapse
Affiliation(s)
- Rine Christopher Reuben
- German Centre of Integrative Biodiversity Research (iDiv) Halle-Jena-Leipzig, Leipzig, Germany
- Institute of Biology, Leipzig University, Leipzig, Germany
| | - Rémy Beugnon
- German Centre of Integrative Biodiversity Research (iDiv) Halle-Jena-Leipzig, Leipzig, Germany
- Leipzig Institute for Meteorology, Universität Leipzig, Leipzig, Germany
- CEFE, Université de Montpellier, CNRS, EPHE, IRD, Montpellier, France
| | - Stephanie D. Jurburg
- German Centre of Integrative Biodiversity Research (iDiv) Halle-Jena-Leipzig, Leipzig, Germany
- Department of Environmental Microbiology, Helmholtz Centre for Environmental Research - UFZ, Leipzig, Germany
| |
Collapse
|
50
|
Armstrong AJS, Horton DB, Andrews T, Greenberg P, Roy J, Gennaro ML, Carson JL, Panettieri RA, Barrett ES, Blaser MJ. Saliva microbiome in relation to SARS-CoV-2 infection in a prospective cohort of healthy US adults. EBioMedicine 2023; 94:104731. [PMID: 37487417 PMCID: PMC10382861 DOI: 10.1016/j.ebiom.2023.104731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 06/08/2023] [Accepted: 07/13/2023] [Indexed: 07/26/2023] Open
Abstract
BACKGROUND The clinical outcomes of SARS-CoV-2 infection vary in severity, potentially influenced by the resident human microbiota. There is limited consensus on conserved microbiome changes in response to SARS-CoV-2 infection, with many studies focusing on severely ill individuals. This study aimed to assess the variation in the upper respiratory tract microbiome using saliva specimens in a cohort of individuals with primarily mild to moderate disease. METHODS In early 2020, a cohort of 831 adults without known SARS-CoV-2 infection was followed over a six-month period to assess the occurrence and natural history of SARS-CoV-2 infection. From this cohort, 81 participants with a SARS-CoV-2 infection, along with 57 unexposed counterparts were selected with a total of 748 serial saliva samples were collected for analysis. Total bacterial abundance, composition, population structure, and gene function of the salivary microbiome were measured using 16S rRNA gene and shotgun metagenomic sequencing. FINDINGS The salivary microbiome remained stable in unexposed individuals over the six-month study period, as evidenced by all measured metrics. Similarly, participants with mild to moderate SARS-CoV-2 infection showed microbiome stability throughout and after their infection. However, there were significant reductions in microbiome diversity among SARS-CoV-2-positive participants with severe symptoms early after infection. Over time, the microbiome diversity in these participants showed signs of recovery. INTERPRETATION These findings demonstrate the resilience of the salivary microbiome in relation to SARS-CoV-2 infection. Mild to moderate infections did not significantly disrupt the stability of the salivary microbiome, suggesting its ability to maintain its composition and function. However, severe SARS-CoV-2 infection was associated with temporary reductions in microbiome diversity, indicating the limits of microbiome resilience in the face of severe infection. FUNDING This project was supported in part by Danone North America and grants from the National Institutes of Health, United States.
Collapse
Affiliation(s)
- Abigail J S Armstrong
- Center for Advanced Biotechnology and Medicine, Rutgers University, Piscataway, New Jersey, USA
| | - Daniel B Horton
- Department of Pediatrics, Rutgers Robert Wood Johnson Medical School, New Brunswick, New Jersey, USA; Rutgers Center for Pharmacoepidemiology and Treatment Science, Institute for Health, Health Care Policy, and Aging Research, New Brunswick, New Jersey, USA; Department of Biostatistics and Epidemiology, Rutgers School of Public Health, Piscataway, New Jersey, USA
| | - Tracy Andrews
- Department of Biostatistics and Epidemiology, Rutgers School of Public Health, Piscataway, New Jersey, USA
| | - Patricia Greenberg
- Department of Biostatistics and Epidemiology, Rutgers School of Public Health, Piscataway, New Jersey, USA
| | - Jason Roy
- Department of Biostatistics and Epidemiology, Rutgers School of Public Health, Piscataway, New Jersey, USA
| | - Maria Laura Gennaro
- Department of Medicine, Public Health Research Institute, New Jersey Medical School, Rutgers University, Newark, New Jersey, USA
| | - Jeffrey L Carson
- Department of Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, New Jersey, USA
| | - Reynold A Panettieri
- Department of Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, New Jersey, USA
| | - Emily S Barrett
- Department of Biostatistics and Epidemiology, Rutgers School of Public Health, Piscataway, New Jersey, USA; Environmental and Occupational Health Sciences Institute, Rutgers University, Piscataway, New Jersey, USA
| | - Martin J Blaser
- Center for Advanced Biotechnology and Medicine, Rutgers University, Piscataway, New Jersey, USA.
| |
Collapse
|