1
|
Cai J, Xu J, Tan Y, Xiang Y, Li Z, Zheng J, Li Y. Gut microbiota alteration and its association with immune function in post-COVID-19 patients. Folia Microbiol (Praha) 2024; 69:857-864. [PMID: 38177971 DOI: 10.1007/s12223-023-01118-z] [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: 07/15/2023] [Accepted: 12/02/2023] [Indexed: 01/06/2024]
Abstract
To reveal the variation of gut microbiota and its association with immune function in cured patients with coronavirus 2019 (COVID-19) disease, gut microbiota of patients discharged from hospital for 20 ~ 23 months and healthy volunteers was analyzed by high throughput 16S rRNA sequencing. The diversity and abundance were compared, and the correlation with immunity factors was investigated, and changes in the content of 6 genera microorganisms with proportion higher than 0.1% were revealed in patients with COVID-19 disease: reduced content of Subdoligranulum, Haemophilus, Coprococcus, Eubacterium vertriosum group, and Lachnospiraceae ND3007 group and increased content of Hungatella. NK cells were negatively correlated to Subdoligranulum, while CD8 cells were positively correlated to Subdoligranulum but negative to Hungatella. IL-8 concentration was negatively correlated to Subdoligranulum, Haemophilus, Coprococcus, Eubacterium vertriosum group, and Lachnospiraceae ND3007 group but positively to Hungatella, while IL-1β concentration was negatively correlated to Haemophilus and Eubacterium ventriosum group but positively to Hungatella. The variation of probiotics and potential pathogenic bacteria implies a higher risk in diseases and inflammation, and the modulation of the gut microbiota may help the healing of COVID-19 patients.
Collapse
Affiliation(s)
- Jiaojiao Cai
- College of Public Health, Chongqing Medical University, Chongqing, 400016, China
- Microbiological Laboratory, Chongqing Center for Disease Control and Prevention, Chongqing, 400042, China
| | - Jingru Xu
- Microbiological Laboratory, Chongqing Center for Disease Control and Prevention, Chongqing, 400042, China
| | - Yan Tan
- Microbiological Laboratory, Chongqing Center for Disease Control and Prevention, Chongqing, 400042, China
| | - Yao Xiang
- Microbiological Laboratory, Chongqing Center for Disease Control and Prevention, Chongqing, 400042, China
| | - Zhifeng Li
- Microbiological Laboratory, Chongqing Center for Disease Control and Prevention, Chongqing, 400042, China
| | - Juan Zheng
- Department of Neurology, Chongqing Red Cross Hospital (People's Hospital of Jiangbei District), Chongqing, 400020, China
| | - Yingli Li
- College of Public Health, Chongqing Medical University, Chongqing, 400016, China.
| |
Collapse
|
2
|
Carvajal JJ, García-Castillo V, Cuellar SV, Campillay-Véliz CP, Salazar-Ardiles C, Avellaneda AM, Muñoz CA, Retamal-Díaz A, Bueno SM, González PA, Kalergis AM, Lay MK. New insights into the pathogenesis of SARS-CoV-2 during and after the COVID-19 pandemic. Front Immunol 2024; 15:1363572. [PMID: 38911850 PMCID: PMC11190347 DOI: 10.3389/fimmu.2024.1363572] [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: 12/31/2023] [Accepted: 04/24/2024] [Indexed: 06/25/2024] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is responsible for the respiratory distress condition known as COVID-19. This disease broadly affects several physiological systems, including the gastrointestinal, renal, and central nervous (CNS) systems, significantly influencing the patient's overall quality of life. Additionally, numerous risk factors have been suggested, including gender, body weight, age, metabolic status, renal health, preexisting cardiomyopathies, and inflammatory conditions. Despite advances in understanding the genome and pathophysiological ramifications of COVID-19, its precise origins remain elusive. SARS-CoV-2 interacts with a receptor-binding domain within angiotensin-converting enzyme 2 (ACE2). This receptor is expressed in various organs of different species, including humans, with different abundance. Although COVID-19 has multiorgan manifestations, the main pathologies occur in the lung, including pulmonary fibrosis, respiratory failure, pulmonary embolism, and secondary bacterial pneumonia. In the post-COVID-19 period, different sequelae may occur, which may have various causes, including the direct action of the virus, alteration of the immune response, and metabolic alterations during infection, among others. Recognizing the serious adverse health effects associated with COVID-19, it becomes imperative to comprehensively elucidate and discuss the existing evidence surrounding this viral infection, including those related to the pathophysiological effects of the disease and the subsequent consequences. This review aims to contribute to a comprehensive understanding of the impact of COVID-19 and its long-term effects on human health.
Collapse
Affiliation(s)
- Jonatan J. Carvajal
- Department of Biotechnology, Faculty of Marine Sciences and Biological Resources, University of Antofagasta, Antofagasta, Chile
| | - Valeria García-Castillo
- Department of Biotechnology, Faculty of Marine Sciences and Biological Resources, University of Antofagasta, Antofagasta, Chile
| | - Shelsy V. Cuellar
- Department of Biotechnology, Faculty of Marine Sciences and Biological Resources, University of Antofagasta, Antofagasta, Chile
| | | | - Camila Salazar-Ardiles
- Center for Research in Physiology and Altitude Medicine (FIMEDALT), Biomedical Department, Faculty of Health Sciences, University of Antofagasta, Antofagasta, Chile
| | - Andrea M. Avellaneda
- Department of Biotechnology, Faculty of Marine Sciences and Biological Resources, University of Antofagasta, Antofagasta, Chile
- Department of Basic Sciences, Faculty of Sciences, Universidad Santo Tomás, Antofagasta, Chile
| | - Christian A. Muñoz
- Research Center in Immunology and Biomedical Biotechnology of Antofagasta (CIIBBA), University of Antofagasta, Antofagasta, Chile
- Department of Medical Technology, Faculty of Health Sciences, University of Antofagasta, Antofagasta, Chile
- Millennium Institute on Immunology and Immunotherapy, Department of Biotechnology, Faculty of Marine Sciences and Biological Resources, Department of Medical Technology, Faculty of Health Sciences, University of Antofagasta, Antofagasta, Chile
| | - Angello Retamal-Díaz
- Department of Biotechnology, Faculty of Marine Sciences and Biological Resources, University of Antofagasta, Antofagasta, Chile
- Research Center in Immunology and Biomedical Biotechnology of Antofagasta (CIIBBA), University of Antofagasta, Antofagasta, Chile
- Millennium Institute on Immunology and Immunotherapy, Department of Biotechnology, Faculty of Marine Sciences and Biological Resources, Department of Medical Technology, Faculty of Health Sciences, University of Antofagasta, Antofagasta, Chile
| | - Susan M. Bueno
- Millennium Institute on Immunology and Immunotherapy, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Pablo A. González
- Millennium Institute on Immunology and Immunotherapy, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Alexis M. Kalergis
- Millennium Institute on Immunology and Immunotherapy, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
- Departamento de Endocrinología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Margarita K. Lay
- Department of Biotechnology, Faculty of Marine Sciences and Biological Resources, University of Antofagasta, Antofagasta, Chile
- Research Center in Immunology and Biomedical Biotechnology of Antofagasta (CIIBBA), University of Antofagasta, Antofagasta, Chile
- Millennium Institute on Immunology and Immunotherapy, Department of Biotechnology, Faculty of Marine Sciences and Biological Resources, Department of Medical Technology, Faculty of Health Sciences, University of Antofagasta, Antofagasta, Chile
| |
Collapse
|
3
|
Sun Z, Shi C, Jin L. Mechanisms by Which SARS-CoV-2 Invades and Damages the Central Nervous System: Apart from the Immune Response and Inflammatory Storm, What Else Do We Know? Viruses 2024; 16:663. [PMID: 38793545 PMCID: PMC11125732 DOI: 10.3390/v16050663] [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: 01/30/2024] [Revised: 03/29/2024] [Accepted: 04/23/2024] [Indexed: 05/26/2024] Open
Abstract
Initially reported as pneumonia of unknown origin, COVID-19 is increasingly being recognized for its impact on the nervous system, despite nervous system invasions being extremely rare. As a result, numerous studies have been conducted to elucidate the mechanisms of nervous system damage and propose appropriate coping strategies. This review summarizes the mechanisms by which SARS-CoV-2 invades and damages the central nervous system, with a specific focus on aspects apart from the immune response and inflammatory storm. The latest research findings on these mechanisms are presented, providing new insights for further in-depth research.
Collapse
Affiliation(s)
- Zihan Sun
- Qingdao Medical College, Qingdao University, Qingdao 266071, China
| | - Chunying Shi
- Department of Human Anatomy, Histology and Embryology, School of Basic Medicine, Qingdao University, Qingdao 266071, China
| | - Lixin Jin
- Department of Human Anatomy, Histology and Embryology, School of Basic Medicine, Qingdao University, Qingdao 266071, China
| |
Collapse
|
4
|
Venugopal N, Armstrong PA, Wright TJ, Randolph KM, Batson RD, Yuen KCJ, Masel B, Sheffield-Moore M, Pyles RB, Urban RJ. Is there a role for growth hormone replacement in adults to control acute and post-acute COVID-19? Best Pract Res Clin Endocrinol Metab 2023; 37:101842. [PMID: 37996257 DOI: 10.1016/j.beem.2023.101842] [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] [Indexed: 11/25/2023]
Abstract
The SARS-CoV-2 pandemic created a multitude of medical crossroads requiring real time adaptations of best practice covering preventative and interventional aspects of care. Among the many discoveries borne from efforts to address the myriad clinical presentations across multiple organ systems was a common impact on tissues with cells that express the ACE-2 receptor. The vast majority of acute infections began and often ended in the respiratory tract, but more recent evaluations have confirmed significant extrapulmonary manifestations including symptom clusters that extend beyond the acute phase of infection collectively referred to as "post-acute sequelae SARS-CoV-2 infection" (PASC) or more commonly as "long (-haul) COVID". Both acute SARS-CoV-2 infection and PASC are associated with gut microbiome dysbiosis and alterations in the gut-brain and HPA-axis in a subset of the infected. Mounting evidence suggests these extrapulmonary manifestations may ultimately lead to reduced growth hormone (GH) secretion as demonstrated following stimulation tests. Disrupted GH secretion could cause or exacerbate long lasting neuropsychological symptoms as seen in other similar manifesting conditions. Ongoing clinical research has shown promising improvement in PASC patients with fatigue and cognition complaints can be achieved via GH replacement therapy. GH stimulation testing should be considered in PASC workups and future research should delve deeper into the mechanistic effects of GH on acute COVID and PASC.
Collapse
Affiliation(s)
- Navneet Venugopal
- John Sealy School of Medicine, University of Texas Medical Branch, 301 University Blvd., Galveston, TX 77555, USA.
| | - Peyton A Armstrong
- John Sealy School of Medicine, University of Texas Medical Branch, 301 University Blvd., Galveston, TX 77555, USA.
| | - Traver J Wright
- Department of Internal Medicine, University of Texas Medical Branch, 301 University Blvd., Galveston, TX 77555, USA.
| | - Kathleen M Randolph
- Department of Internal Medicine, University of Texas Medical Branch, 301 University Blvd., Galveston, TX 77555, USA.
| | | | - Kevin C J Yuen
- Department of Neuroendocrinology, Barrow Pituitary Center and Barrow Neuroendocrinology Clinic, St. Joseph's Hospital and Medical Center, Phoenix, AZ 85013, USA.
| | - Brent Masel
- Department of Neurology, University of Texas Medical Branch, 301 University Blvd., Galveston, TX 77555, USA; Centre for Neuro Skills, Bakersfield, CA 93313, USA.
| | - Melinda Sheffield-Moore
- Department of Internal Medicine, University of Texas Medical Branch, 301 University Blvd., Galveston, TX 77555, USA.
| | - Richard B Pyles
- Department of Pediatrics, University of Texas Medical Branch, 301 University Blvd., Galveston, TX 77555, USA.
| | - Randall J Urban
- Department of Internal Medicine, University of Texas Medical Branch, 301 University Blvd., Galveston, TX 77555, USA.
| |
Collapse
|
5
|
Erol I, Kotil SE, Ortakci F, Durdagi S. Exploring the binding capacity of lactic acid bacteria derived bacteriocins against RBD of SARS-CoV-2 Omicron variant by molecular simulations. J Biomol Struct Dyn 2023; 41:10774-10784. [PMID: 36591650 DOI: 10.1080/07391102.2022.2158934] [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: 07/22/2022] [Accepted: 12/10/2022] [Indexed: 01/03/2023]
Abstract
The changes in the SARS-CoV-2 genome have resulted in the emergence of new variants. Some of the variants have been classified as variants of concern (VOC). These strains have higher transmission rate and improved fitness. One of the prevalent were the Omicron variant. Unlike previous VOCs, the Omicron possesses fifteen mutations on the spike protein's receptor binding domain (RBD). The modifications of spike protein's key amino acid residues facilitate the virus' binding capability against ACE2, resulting in an increase in the infectiousness of Omicron variant. Consequently, investigating the prevention and treatment of the Omicron variant is crucial. In the present study, we aim to explore the binding capacity of twenty-two bacteriocins derived from Lactic Acid Bacteria (LAB) against the Omicron variant by using protein-peptidedocking and molecular dynamics (MD) simulations. The Omicron variant RBD was prepared by introducing fifteen mutations using PyMol. The protein-peptide complexes were obtained using HADDOCK v2.4 docking webserver. Top scoring complexes obtained from HADDOCK webserver were retrieved and submitted to the PRODIGY server for the prediction of binding energies. RBD-bacteriocin complexes were subjected to MD simulations. We discovered promising peptide-based therapeutic candidates for the inhibition of Omicron variant for example Salivaricin B, Pediocin PA 1, Plantaricin W, Lactococcin mmfii and Enterocin A. The lead bacteriocins, except Enterocin A, are biosynthesized by food-grade lactic acid bacteria. Our study puts forth a preliminary information regarding potential utilization of food-grade LAB-derived bacteriocins, particularly Salivaricin B and Pediocin PA 1, for Covid-19 treatment and prophylaxis.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Ismail Erol
- Computational Biology and Molecular Simulations Laboratory, Department of Biophysics, School of Medicine, Bahcesehir University, Istanbul, Turkey
| | - Seyfullah Enes Kotil
- Department of Biophysics, School of Medicine, Bahcesehir University, Istanbul, Turkey
| | - Fatih Ortakci
- Bioengineering Department, Faculty of Life and Natural Sciences, Abdullah Gul University, Kayseri, Turkey
| | - Serdar Durdagi
- Computational Biology and Molecular Simulations Laboratory, Department of Biophysics, School of Medicine, Bahcesehir University, Istanbul, Turkey
- School of Pharmacy, Bahcesehir University, Istanbul, Turkey
| |
Collapse
|
6
|
Zeng X, Yue H, Zhang L, Chen G, Zheng Q, Hu Q, Du X, Tian Q, Zhao X, Liang L, Yang Z, Bai H, Liu Y, Zhao M, Fu X. Gut microbiota-derived autoinducer-2 regulates lung inflammation through the gut-lung axis. Int Immunopharmacol 2023; 124:110971. [PMID: 37748222 DOI: 10.1016/j.intimp.2023.110971] [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: 07/25/2023] [Revised: 09/17/2023] [Accepted: 09/18/2023] [Indexed: 09/27/2023]
Abstract
OBJECTIVE This study aimed to determine whether autoinducer-2 (AI-2), a crucial bacterial metabolite and quorum sensing molecule, is involved in lung immunity through the gut-lung axis. METHODS The level of AI-2 and the gut microbiome composition were analysed in the stools from pneumonic patients and the mouse model of acute lung injury. The effect of AI-2 on lung inflammation was further investigated in the mouse model. RESULTS The diversity of the faecal microbiota was reduced in pneumonic patients treated with antibiotics compared with healthy volunteers. The AI-2 level in the stool was positively correlated with inflammatory molecules in the serum of pneumonic patients. Intraperitoneal injection of AI-2 reinforced lung inflammation in the acute lung injury mouse model, characterized by increased secretion of inflammatory molecules, including IL-6, IL-1β, C-C chemokines, and CXCL chemokines, which were alleviated by the AI-2 inhibitor D-ribose. CONCLUSIONS Our results suggested that gut microbiota-derived AI-2 could modulate lung inflammation through the gut-lung axis.
Collapse
Affiliation(s)
- Xianghao Zeng
- Clinical Medical College, North Sichuan Medical College, Nanchong City, Sichuan Province 637000, China; Department of Gastroenterology, Clinical Medical College and the First Affiliated Hospital of Chengdu Medical College, Chengdu City, Sichuan Province 610500, China
| | - Huawen Yue
- Clinical Medical College, North Sichuan Medical College, Nanchong City, Sichuan Province 637000, China
| | - Ling Zhang
- Clinical Medical College, North Sichuan Medical College, Nanchong City, Sichuan Province 637000, China
| | - Guimei Chen
- Clinical Medical College, North Sichuan Medical College, Nanchong City, Sichuan Province 637000, China
| | - Qiao Zheng
- Clinical Medical College, North Sichuan Medical College, Nanchong City, Sichuan Province 637000, China
| | - Qing Hu
- Clinical Medical College, North Sichuan Medical College, Nanchong City, Sichuan Province 637000, China
| | - Xinhao Du
- Department of Gastroenterology, Clinical Medical College and the First Affiliated Hospital of Chengdu Medical College, Chengdu City, Sichuan Province 610500, China
| | - Qian Tian
- Department of Gastroenterology, Clinical Medical College and the First Affiliated Hospital of Chengdu Medical College, Chengdu City, Sichuan Province 610500, China
| | - Xinyu Zhao
- Department of Gastroenterology, Clinical Medical College and the First Affiliated Hospital of Chengdu Medical College, Chengdu City, Sichuan Province 610500, China
| | - Lanfan Liang
- Department of Gastroenterology, Clinical Medical College and the First Affiliated Hospital of Chengdu Medical College, Chengdu City, Sichuan Province 610500, China
| | - Ziyi Yang
- Department of Gastroenterology, Clinical Medical College and the First Affiliated Hospital of Chengdu Medical College, Chengdu City, Sichuan Province 610500, China
| | - Hang Bai
- Department of Gastroenterology, Clinical Medical College and the First Affiliated Hospital of Chengdu Medical College, Chengdu City, Sichuan Province 610500, China
| | - Yanqin Liu
- Department of Gastroenterology, Clinical Medical College and the First Affiliated Hospital of Chengdu Medical College, Chengdu City, Sichuan Province 610500, China
| | - Ming Zhao
- Department of Gastroenterology, Clinical Medical College and the First Affiliated Hospital of Chengdu Medical College, Chengdu City, Sichuan Province 610500, China
| | - Xiangsheng Fu
- Department of Gastroenterology, Clinical Medical College and the First Affiliated Hospital of Chengdu Medical College, Chengdu City, Sichuan Province 610500, China.
| |
Collapse
|
7
|
Haldar S, Jadhav SR, Gulati V, Beale DJ, Balkrishna A, Varshney A, Palombo EA, Karpe AV, Shah RM. Unravelling the gut-lung axis: insights into microbiome interactions and Traditional Indian Medicine's perspective on optimal health. FEMS Microbiol Ecol 2023; 99:fiad103. [PMID: 37656879 PMCID: PMC10508358 DOI: 10.1093/femsec/fiad103] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 07/05/2023] [Accepted: 08/30/2023] [Indexed: 09/03/2023] Open
Abstract
The microbiome of the human gut is a complex assemblage of microorganisms that are in a symbiotic relationship with one another and profoundly influence every aspect of human health. According to converging evidence, the human gut is a nodal point for the physiological performance matrixes of the vital organs on several axes (i.e. gut-brain, gut-lung, etc). As a result of COVID-19, the importance of gut-lung dysbiosis (balance or imbalance) has been realised. In view of this, it is of utmost importance to develop a comprehensive understanding of the microbiome, as well as its dysbiosis. In this review, we provide an overview of the gut-lung axial microbiome and its importance in maintaining optimal health. Human populations have successfully adapted to geophysical conditions through traditional dietary practices from around the world. In this context, a section has been devoted to the traditional Indian system of medicine and its theories and practices regarding the maintenance of optimally customized gut health.
Collapse
Affiliation(s)
- Swati Haldar
- Drug Discovery and Development Division, Patanjali Research Institute, NH-58, Haridwar 249405, Uttarakhand, India
| | - Snehal R Jadhav
- Consumer-Analytical-Safety-Sensory (CASS) Food Research Centre, School of Exercise and Nutrition Sciences, Deakin University, Burwood, VIC 3125, Australia
| | - Vandana Gulati
- Biomedical Science, School of Science and Technology Faculty of Science, Agriculture, Business and Law, University of New England, Armidale, NSW 2351, Australia
| | - David J Beale
- Environment, Commonwealth Scientific and Industrial Research Organisation (CSIRO), Ecosciences Precinct, Dutton Park, QLD 4102, Australia
| | - Acharya Balkrishna
- Drug Discovery and Development Division, Patanjali Research Institute, NH-58, Haridwar 249405, Uttarakhand, India
- Department of Allied and Applied Sciences, University of Patanjali, Patanjali Yog Peeth, Roorkee-Haridwar Road, Haridwar 249405, Uttarakhand, India
| | - Anurag Varshney
- Drug Discovery and Development Division, Patanjali Research Institute, NH-58, Haridwar 249405, Uttarakhand, India
- Department of Allied and Applied Sciences, University of Patanjali, Patanjali Yog Peeth, Roorkee-Haridwar Road, Haridwar 249405, Uttarakhand, India
| | - Enzo A Palombo
- Department of Chemistry and Biotechnology, School of Science, Computing and Engineering Technologies, Swinburne University of Technology, Hawthorn, VIC 3122, Australia
| | - Avinash V Karpe
- Department of Chemistry and Biotechnology, School of Science, Computing and Engineering Technologies, Swinburne University of Technology, Hawthorn, VIC 3122, Australia
- Socio-Eternal Thinking for Unity (SETU), Melbourne, VIC 3805, Australia
- Agriculture and Food, Commonwealth Scientific and Industrial Research Organisation (CSIRO), Acton, ACT 2601, Australia
| | - Rohan M Shah
- Department of Chemistry and Biotechnology, School of Science, Computing and Engineering Technologies, Swinburne University of Technology, Hawthorn, VIC 3122, Australia
- School of Health and Biomedical Sciences, STEM College, RMIT University, Bundoora West, VIC 3083, Australia
| |
Collapse
|
8
|
Jia M, Yi B, Chen X, Xu Y, Xu X, Wu Z, Ji J, Tang J, Yu D, Zheng Y, Zhou Q, Zhao Y. Carbon dots induce pathological damage to the intestine via causing intestinal flora dysbiosis and intestinal inflammation. J Nanobiotechnology 2023; 21:167. [PMID: 37231475 PMCID: PMC10210306 DOI: 10.1186/s12951-023-01931-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 05/16/2023] [Indexed: 05/27/2023] Open
Abstract
BACKGROUND Carbon dots (CDs), as excellent antibacterial nanomaterials, have gained great attention in treating infection-induced diseases such as periodontitis and stomatitis. Given the eventual exposure of CDs to the intestine, elucidating the effect of CDs on intestinal health is required for the safety evaluation of CDs. RESULTS Herein, CDs extracted from ε-poly-L-lysine (PL) were chosen to explore the modulation effect of CDs on probiotic behavior in vitro and intestinal remodeling in vivo. Results verify that PL-CDs negatively regulate Lactobacillus rhamnosus (L. rhamnosus) growth via increasing reactive oxygen species (ROS) production and reducing the antioxidant activity, which subsequently destroys membrane permeability and integrity. PL-CDs are also inclined to inhibit cell viability and accelerate cell apoptosis. In vivo, the gavage of PL-CDs is verified to induce inflammatory infiltration and barrier damage in mice. Moreover, PL-CDs are found to increase the Firmicutes to Bacteroidota (F/B) ratio and the relative abundance of Lachnospiraceae while decreasing that of Muribaculaceae. CONCLUSION Overall, these evidences indicate that PL-CDs may inevitably result in intestinal flora dysbiosis via inhibiting probiotic growth and simultaneously activating intestinal inflammation, thus causing pathological damage to the intestine, which provides an effective and insightful reference for the potential risk of CDs from the perspective of intestinal remodeling.
Collapse
Affiliation(s)
- Mengmeng Jia
- School of Public Health, Qingdao University, Qingdao, 266071 China
| | - Bingcheng Yi
- School of Rehabilitation Sciences and Engineering, University of Health and Rehabilitation Sciences, Qingdao, 266071 China
| | - Xian Chen
- School of Public Health, Qingdao University, Qingdao, 266071 China
| | - Yongzhi Xu
- School of Stomatology, Qingdao University, Qingdao, 266003 China
| | - Xinkai Xu
- School of Stomatology, Qingdao University, Qingdao, 266003 China
| | - Zhaoxu Wu
- School of Public Health, Qingdao University, Qingdao, 266071 China
| | - Jing Ji
- School of Public Health, Qingdao University, Qingdao, 266071 China
| | - Jinglong Tang
- School of Public Health, Qingdao University, Qingdao, 266071 China
| | - Dianke Yu
- School of Public Health, Qingdao University, Qingdao, 266071 China
| | - Yuxin Zheng
- School of Public Health, Qingdao University, Qingdao, 266071 China
| | - Qihui Zhou
- School of Rehabilitation Sciences and Engineering, University of Health and Rehabilitation Sciences, Qingdao, 266071 China
- School of Stomatology, Qingdao University, Qingdao, 266003 China
- Zhejiang Engineering Research Center for Tissue Repair Materials, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325000 Zhejiang China
| | - Yanjie Zhao
- School of Public Health, Qingdao University, Qingdao, 266071 China
| |
Collapse
|
9
|
Niero M, Bartoli G, De Colle P, Scarcella M, Zanetti M. Impact of Dietary Fiber on Inflammation and Insulin Resistance in Older Patients: A Narrative Review. Nutrients 2023; 15:nu15102365. [PMID: 37242248 DOI: 10.3390/nu15102365] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 05/11/2023] [Accepted: 05/16/2023] [Indexed: 05/28/2023] Open
Abstract
The beneficial impact of dietary fiber on the prevention and management of several chronic conditions associated with aging, including diabetes, neurodegenerative, cardiovascular diseases, and cancer, is well-known. High fiber intake has been associated with reduced inflammatory mediators counteracting the low-grade chronic inflammation typical of older age. In addition, dietary fiber improves postprandial glucose response and insulin resistance. In contrast, during acute diseases, its effects on insulin resistance and modulation of immune response are unclear. The aim of this narrative is to summarize the evidence for the potential impact of dietary fiber on inflammation and insulin resistance in older adults, with a particular focus on those acutely ill. Available evidence suggests that dietary fiber has the potential to counteract acute inflammation and to improve metabolic health. In addition, modulation of gut microbiota composition may contribute to improved immune function, particularly in the setting of aging-associated dysbiosis. This phenomenon has relevant implications in those acutely ill, in whom dysbiosis can be exacerbated. Our review leads to the conclusion that dietary interventions based on fiber manipulation could exploit its beneficial effects on inflammation and insulin resistance, if conducted from a precision nutrition perspective. This could also be true for the acutely ill patient, even though strong evidence is lacking.
Collapse
Affiliation(s)
- Michele Niero
- Geriatric Clinic, Maggiore Hospital, Azienda Sanitaria Universitaria Giuliano Isontina, 34148 Trieste, Italy
- Department of Medical Sciences, University of Trieste, 34127 Trieste, Italy
| | - Giulio Bartoli
- Geriatric Clinic, Maggiore Hospital, Azienda Sanitaria Universitaria Giuliano Isontina, 34148 Trieste, Italy
| | - Paolo De Colle
- Geriatric Clinic, Maggiore Hospital, Azienda Sanitaria Universitaria Giuliano Isontina, 34148 Trieste, Italy
| | - Marialaura Scarcella
- Anesthesia, Intensive Care and Nutritional Science, Azienda Ospedaliera "Santa Maria", Via Tristano di Joannuccio, 05100 Terni, Italy
| | - Michela Zanetti
- Geriatric Clinic, Maggiore Hospital, Azienda Sanitaria Universitaria Giuliano Isontina, 34148 Trieste, Italy
- Department of Medical Sciences, University of Trieste, 34127 Trieste, Italy
| |
Collapse
|
10
|
Eilam Y, Khattib H, Pintel N, Avni D. Microalgae-Sustainable Source for Alternative Proteins and Functional Ingredients Promoting Gut and Liver Health. GLOBAL CHALLENGES (HOBOKEN, NJ) 2023; 7:2200177. [PMID: 37205927 PMCID: PMC10190620 DOI: 10.1002/gch2.202200177] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 01/27/2023] [Indexed: 05/21/2023]
Abstract
Dietary proteins derived from animal sources, although containing well-balanced profiles of essential amino acids, have considerable environmental and adverse health effects associated with the intake of some animal protein-based products. Consuming foods based on animal proteins carries a higher risk of developing non-communicable diseases such as cancer, heart disease, non-alcoholic fatty liver disease (NAFLD), and inflammatory bowel disease (IBD). Moreover, dietary protein consumption is increasing due to population growth, posing a supply challenge. There is, therefore, growing interest in discovering novel alternative protein sources. In this context, microalgae have been recognized as strategic crops that can provide a sustainable source of protein. Compared to conventional high-protein crops, using microalgal biomass for protein production presents several advantages in food and feed in terms of productivity, sustainability, and nutritional value. Moreover, microalgae positively impact the environment by not exploiting land or causing water pollution. Many studies have revealed the potential of microalgae as an alternative protein source with the added value of positive effects on human health due to their anti-inflammatory, antioxidant, and anti-cancer properties. The main emphasis of this review is on the potential health-promoting applications of microalgae-based proteins, peptides, and bioactive substances for IBD and NAFLD.
Collapse
Affiliation(s)
- Yahav Eilam
- Sphingolipids, Active Metabolites, and Immune Modulation LaboratoryMIGAL – Galilee Research InstituteTarshish 2Kiryat ShemonaNorth1101600Israel
- Department of BiotechnologyTel Hai CollegeUpper GalileeNorth1220800Israel
| | - Hamdan Khattib
- Sphingolipids, Active Metabolites, and Immune Modulation LaboratoryMIGAL – Galilee Research InstituteTarshish 2Kiryat ShemonaNorth1101600Israel
| | - Noam Pintel
- Sphingolipids, Active Metabolites, and Immune Modulation LaboratoryMIGAL – Galilee Research InstituteTarshish 2Kiryat ShemonaNorth1101600Israel
| | - Dorit Avni
- Sphingolipids, Active Metabolites, and Immune Modulation LaboratoryMIGAL – Galilee Research InstituteTarshish 2Kiryat ShemonaNorth1101600Israel
- Department of BiotechnologyTel Hai CollegeUpper GalileeNorth1220800Israel
| |
Collapse
|
11
|
Datta AK, Mukherjee A, Biswas A. Gastrointestinal, Respiratory, and Olfactory Neurotropism of Sars-Cov2 as a Possible Trigger of Parkinson's Disease: Is a Multi-Hit Multi-Step Process on the Cards. Ann Indian Acad Neurol 2023; 26:127-136. [PMID: 37179662 PMCID: PMC10171009 DOI: 10.4103/aian.aian_767_22] [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: 09/12/2022] [Revised: 11/20/2022] [Accepted: 12/20/2022] [Indexed: 01/19/2023] Open
Abstract
Since the first emergence of COVID-19 on the global stage, there has been a wealth of evidence to suggest that SARS-Cov2 is not merely a pulmonary pathogen. This virus is unique in its ability to disrupt cellular pathways related to protein homeostasis, mitochondrial function, stress response, and aging. Such effects raise concerns about the long-term fate of survivors of COVID-19 infection, particularly regarding neurodegenerative diseases. The concept of interaction between environmental factors and alpha-synuclein formation in the olfactory bulb and vagal autonomic terminals with subsequent caudo-cranial migration has received much attention in the context of PD pathogenesis. Anosmia and gastrointestinal symptoms are two well-known symptoms of COVID-19, with evidence of an olfactory bulb and vagal infiltration by SARS-CoV2. This raises the possibility of the spread of the viral particles to the brain along multiple cranial nerve routes. Neurotropism, coupled with the ability of the SARS-Cov2 virion to induce abnormal protein folding and stress responses in the central nervous system, in presence of an inflammatory milieu, reinforced by hypoxia, coagulopathy, and endothelial dysfunction, reverberates the intriguing possibility of activation of a neurodegenerative cascade leading to the development of pathological alpha-synuclein aggregates and thus, triggering the development of PD in survivors of COVID19. This review attempts to summarize and critically appraise existing evidence from basic science research and clinical reports of links between COVID-19 and PD and explores the prospect of a multi-hit pathophysiological process, induced by SARS-Cov2 infection, ultimately converging on perturbed cellular protein homeostasis, which although is intriguing, presently lacks robust evidence for confirmation.
Collapse
Affiliation(s)
- Amlan K. Datta
- Department of Neurology, Institute of Post Graduate of Medical Education and Research (IPGME&R) and Bangur Institute of Neurosciences (BIN), Kolkata, West Bengal, India
| | - Adreesh Mukherjee
- Department of Neurology, Institute of Post Graduate of Medical Education and Research (IPGME&R) and Bangur Institute of Neurosciences (BIN), Kolkata, West Bengal, India
| | - Atanu Biswas
- Department of Neurology, Institute of Post Graduate of Medical Education and Research (IPGME&R) and Bangur Institute of Neurosciences (BIN), Kolkata, West Bengal, India
| |
Collapse
|
12
|
Enichen E, Harvey C, Demmig-Adams B. COVID-19 Spotlights Connections between Disease and Multiple Lifestyle Factors. Am J Lifestyle Med 2023; 17:231-257. [PMID: 36883129 PMCID: PMC9445631 DOI: 10.1177/15598276221123005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The SARS-CoV-2 virus (severe acute respiratory syndrome coronavirus 2), and the disease it causes (COVID-19), have had a profound impact on global human society and threaten to continue to have such an impact with newly emerging variants. Because of the widespread effects of SARS-CoV-2, understanding how lifestyle choices impact the severity of disease is imperative. This review summarizes evidence for an involvement of chronic, non-resolving inflammation, gut microbiome disruption (dysbiosis with loss of beneficial microorganisms), and impaired viral defenses, all of which are associated with an imbalanced lifestyle, in severe disease manifestations and post-acute sequelae of SARS-CoV-2 (PASC). Humans' physiological propensity for uncontrolled inflammation and severe COVID-19 are briefly contrasted with bats' low propensity for inflammation and their resistance to viral disease. This insight is used to identify positive lifestyle factors with the potential to act in synergy for restoring balance to the immune response and gut microbiome, and thereby protect individuals against severe COVID-19 and PASC. It is proposed that clinicians should consider recommending lifestyle factors, such as stress management, balanced nutrition and physical activity, as preventative measures against severe viral disease and PASC.
Collapse
Affiliation(s)
- Elizabeth Enichen
- Department of Ecology and Evolutionary Biology, University of Colorado, Boulder, CO, USA (EE, CH, BDA)
| | - Caitlyn Harvey
- Department of Ecology and Evolutionary Biology, University of Colorado, Boulder, CO, USA (EE, CH, BDA)
| | - Barbara Demmig-Adams
- Department of Ecology and Evolutionary Biology, University of Colorado, Boulder, CO, USA (EE, CH, BDA)
| |
Collapse
|
13
|
Ahmed LA, Al-Massri KF. Gut Microbiota Modulation for Therapeutic Management of Various Diseases: A New Perspective Using Stem Cell Therapy. Curr Mol Pharmacol 2023; 16:43-59. [PMID: 35196976 DOI: 10.2174/1874467215666220222105004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 11/08/2021] [Accepted: 12/16/2021] [Indexed: 11/22/2022]
Abstract
Dysbiosis has been linked to various diseases ranging from cardiovascular, neurologic, gastrointestinal, respiratory, and metabolic illnesses to cancer. Restoring of gut microbiota balance represents an outstanding clinical target for the management of various multidrug-resistant diseases. Preservation of gut microbial diversity and composition could also improve stem cell therapy which now has diverse clinical applications in the field of regenerative medicine. Gut microbiota modulation and stem cell therapy may be considered a highly promising field that could add up towards the improvement of different diseases, increasing the outcome and efficacy of each other through mutual interplay or interaction between both therapies. Importantly, more investigations are required to reveal the cross-talk between microbiota modulation and stem cell therapy to pave the way for the development of new therapies with enhanced therapeutic outcomes. This review provides an overview of dysbiosis in various diseases and their management. It also discusses microbiota modulation via antibiotics, probiotics, prebiotics, and fecal microbiota transplant to introduce the concept of dysbiosis correction for the management of various diseases. Furthermore, we demonstrate the beneficial interactions between microbiota modulation and stem cell therapy as a way for the development of new therapies in addition to limitations and future challenges regarding the applications of these therapies.
Collapse
Affiliation(s)
- Lamiaa A Ahmed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Khaled F Al-Massri
- Department of Pharmacy and Biotechnology, Faculty of Medicine and Health Sciences, University of Palestine, Gaza, Palestine
| |
Collapse
|
14
|
Mozota M, Castro I, Gómez-Torres N, Arroyo R, Gutiérrez-Díaz I, Delgado S, Rodríguez JM, Alba C. Administration of Ligilactobacillus salivarius CECT 30632 to elderly during the COVID-19 pandemic: Nasal and fecal metataxonomic analysis and fatty acid profiling. Front Microbiol 2022; 13:1052675. [PMID: 36590434 PMCID: PMC9800801 DOI: 10.3389/fmicb.2022.1052675] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2022] [Accepted: 11/30/2022] [Indexed: 12/23/2022] Open
Abstract
Elderly was the most affected population during the first COVID-19 and those living in nursing homes represented the most vulnerable group, with high mortality rates, until vaccines became available. In a previous article, we presented an open-label trial showing the beneficial effect of the strain Ligilactobacillus salivarius CECT 30632 (previously known as L. salivarius MP101) on the functional and nutritional status, and on the nasal and fecal inflammatory profiles of elderly residing in a nursing home highly affected by the pandemic. The objective of this post-hoc analysis was to elucidate if there were changes in the nasal and fecal bacteriomes of a subset of these patients as a result of the administration of the strain for 4 months and, also, its impact on their fecal fatty acids profiles. Culture-based methods showed that, while L. salivarius (species level) could not be detected in any of the fecal samples at day 0, L. salivarius CECT 30632 (strain level) was present in all the recruited people at day 120. Paradoxically, the increase in the L. salivarius counts was not reflected in changes in the metataxonomic analysis of the nasal and fecal samples or in changes in the fatty acid profiles in the fecal samples of the recruited people. Overall, our results indicate that L. salivarius CECT 30632 colonized, at least temporarily, the intestinal tract of the recruited elderly and may have contributed to improvements in their functional, nutritional, and immunological status, without changing the general structure of their nasal and fecal bacteriomes when assessed at the genus level. They also suggest the ability of low abundance bacteria to train immunity.
Collapse
Affiliation(s)
- Marta Mozota
- Department of Nutrition and Food Science, Complutense University of Madrid, Madrid, Spain
| | - Irma Castro
- Department of Nutrition and Food Science, Complutense University of Madrid, Madrid, Spain
| | - Natalia Gómez-Torres
- Department of Nutrition and Food Science, Complutense University of Madrid, Madrid, Spain
| | - Rebeca Arroyo
- Department of Nutrition and Food Science, Complutense University of Madrid, Madrid, Spain
| | - Isabel Gutiérrez-Díaz
- Department of Microbiology and Biochemistry, Dairy Research Institute of Asturias (IPLA-CSIC), Villaviciosa, Spain
| | - Susana Delgado
- Department of Microbiology and Biochemistry, Dairy Research Institute of Asturias (IPLA-CSIC), Villaviciosa, Spain
| | - Juan Miguel Rodríguez
- Department of Nutrition and Food Science, Complutense University of Madrid, Madrid, Spain
| | - Claudio Alba
- Department of Nutrition and Food Science, Complutense University of Madrid, Madrid, Spain
| |
Collapse
|
15
|
Saikarthik J, Saraswathi I, Alarifi A, Al-Atram AA, Mickeymaray S, Paramasivam A, Shaikh S, Jeraud M, Alothaim AS. Role of neuroinflammation mediated potential alterations in adult neurogenesis as a factor for neuropsychiatric symptoms in Post-Acute COVID-19 syndrome-A narrative review. PeerJ 2022; 10:e14227. [PMID: 36353605 PMCID: PMC9639419 DOI: 10.7717/peerj.14227] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 09/22/2022] [Indexed: 11/06/2022] Open
Abstract
Persistence of symptoms beyond the initial 3 to 4 weeks after infection is defined as post-acute COVID-19 syndrome (PACS). A wide range of neuropsychiatric symptoms like anxiety, depression, post-traumatic stress disorder, sleep disorders and cognitive disturbances have been observed in PACS. The review was conducted based on PRISMA-S guidelines for literature search strategy for systematic reviews. A cytokine storm in COVID-19 may cause a breach in the blood brain barrier leading to cytokine and SARS-CoV-2 entry into the brain. This triggers an immune response in the brain by activating microglia, astrocytes, and other immune cells leading to neuroinflammation. Various inflammatory biomarkers like inflammatory cytokines, chemokines, acute phase proteins and adhesion molecules have been implicated in psychiatric disorders and play a major role in the precipitation of neuropsychiatric symptoms. Impaired adult neurogenesis has been linked with a variety of disorders like depression, anxiety, cognitive decline, and dementia. Persistence of neuroinflammation was observed in COVID-19 survivors 3 months after recovery. Chronic neuroinflammation alters adult neurogenesis with pro-inflammatory cytokines supressing anti-inflammatory cytokines and chemokines favouring adult neurogenesis. Based on the prevalence of neuropsychiatric symptoms/disorders in PACS, there is more possibility for a potential impairment in adult neurogenesis in COVID-19 survivors. This narrative review aims to discuss the various neuroinflammatory processes during PACS and its effect on adult neurogenesis.
Collapse
Affiliation(s)
- Jayakumar Saikarthik
- Department of Basic Medical Sciences, College of Dentistry, Al Zulfi, Majmaah University, Al-Majmaah, Riyadh, Kingdom of Saudi Arabia,Department of Medical Education, College of Dentistry, Al Zulfi, Majmaah University, Al Majmaah, Riyadh, Kingdom of Saudi Arabia
| | - Ilango Saraswathi
- Department of Physiology, Madha Medical College and Research Institute, Chennai, Tamil Nadu, India
| | - Abdulaziz Alarifi
- Department of Basic Sciences, College of Science and Health Professions, King Saud Bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia,King Abdullah International Medical Research Centre, Riyadh, Saudi Arabia
| | - Abdulrahman A. Al-Atram
- Department of Psychiatry, College of Medicine, Majmaah University, Al Majmaah, Riyadh, Kingdom of Saudi Arabia
| | - Suresh Mickeymaray
- Department of Biology, College of Science, Al Zulfi, Majmaah University, Al Majmaah, Riyadh, Kingdom of Saudi Arabia
| | - Anand Paramasivam
- Department of Physiology, RVS Dental College and Hospital, Kumaran Kottam Campus, Kannampalayan, Coimbatore, Tamilnadu, India
| | - Saleem Shaikh
- Department of Medical Education, College of Dentistry, Al Zulfi, Majmaah University, Al Majmaah, Riyadh, Kingdom of Saudi Arabia,Department of Maxillofacial Surgery and Diagnostic Sciences, College of Dentistry, Al Zulfi, Majmaah University, Al Majmaah, Riyadh, Kingdom of Saudi Arabia
| | - Mathew Jeraud
- Department of Physiology, Ibn Sina National College for Medical Studies, Jeddah, Saudi Arabia
| | - Abdulaziz S. Alothaim
- Department of Biology, College of Science, Al Zulfi, Majmaah University, Al Majmaah, Riyadh, Kingdom of Saudi Arabia
| |
Collapse
|
16
|
Nayebi A, Navashenaq JG, Soleimani D, Nachvak SM. Probiotic supplementation: A prospective approach in the treatment of COVID-19. Nutr Health 2022; 28:163-175. [PMID: 34747257 PMCID: PMC9160438 DOI: 10.1177/02601060211049631] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Background: Despite strategies based on social distancing, the coronavirus disease 2019 (COVID-19) expands globally, and so far, many attempts have been made to achieve effective treatment for patients with COVID-19. This disease infects the lower respiratory tract and may lead to severe acute respiratory syndrome coronavirus (SARS-CoV). COVID-19 also can cause gastrointestinal infections. Therefore, COVID-19 patients with gastrointestinal symptoms are more likely to be complicated by SARS-CoV. In this disease, acquired immune responses are impaired, and uncontrolled inflammatory responses result in cytokine storms, leading to acute lung injury and thrombus formation. Probiotics are living microorganisms that contribute to the health of the host if administered in appropriate doses. Aim: This study aimed to provide evidence to show the importance of gut dysbiosis in viral disease, especially COVID-19. Therefore, we have focused on the impact of probiotics consumption on preventing severe symptoms of the disease. Methods: We have entirely searched SCOPUS, PubMed, and Google Scholar databases to collect evidence regarding the relationship between probiotics and viral infections to expand this relationship to the COVID-19. Results: It has been shown that probiotics directly counteract SARS-CoV in the gastrointestinal and respiratory tracts. Moreover, probiotics suppress severe immune responses and prevent cytokine storms to inhibit pathologic inflammatory conditions in the body via modulation of immune responses. Conclusion: According to available evidence based on their antiviral and respiratory activities, using probiotics might be an adjuvant therapy to reduce the burden and severity of this disease.
Collapse
Affiliation(s)
- Atiyeh Nayebi
- Student Research Committee, Nutritional Sciences Department, School of Nutrition Sciences and Food Technology, Kermanshah University of Medical Sciences, Kermanshah, Iran
- Nutritional Sciences Department, School of Nutrition Sciences and Food Technology, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | | | - Davood Soleimani
- Student Research Committee, Nutritional Sciences Department, School of Nutrition Sciences and Food Technology, Kermanshah University of Medical Sciences, Kermanshah, Iran
- Research Center of Oils and Fats, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Seyyed Mostafa Nachvak
- Student Research Committee, Nutritional Sciences Department, School of Nutrition Sciences and Food Technology, Kermanshah University of Medical Sciences, Kermanshah, Iran
- Nutritional Sciences Department, School of Nutrition Sciences and Food Technology, Kermanshah University of Medical Sciences, Kermanshah, Iran
| |
Collapse
|
17
|
Hajipour A, Afsharfar M, Jonoush M, Ahmadzadeh M, Gholamalizadeh M, Hassanpour Ardekanizadeh N, Doaei S, Mohammadi‐Nasrabadi F. The effects of dietary fiber on common complications in critically ill patients; with a special focus on viral infections; a systematic reveiw. Immun Inflamm Dis 2022; 10:e613. [PMID: 35478440 PMCID: PMC9017620 DOI: 10.1002/iid3.613] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 02/16/2022] [Accepted: 03/14/2022] [Indexed: 12/17/2022] Open
Abstract
Background Viral infections are mostly highly contagious and may cause widespread health problems. Some studies reported that the dietary fiber (DF) may be effective in reducing the complications of viral infections in intensive care unit (ICU) patients. The present review study aimed to investigate the effect of DF on common complications in critically ill patients with viral infections. Methods A literature review was conducted for the published papers in English from January 2001 to July 2021 using related keywords. Studies with clinical trial or case‐control design described the effects of fiber intake on the complications of viral infections in patients admitted to the ICU were collected. Results DF may reduce the mortality rate of viral infections through modulating inflammatory processes. A higher intake of DF intake may improve hyperglycemia and impaired glucose tolerance in patients with viral infections. A high‐fiber formula in enteral nutrition was reported to reduce the risk of diarrhea in patients with viral infections. Conclusion DF may reduce the complications of viral infections such as inflammation, diarrhea, hyperglycemia, and mortality in critically ill patients. Future longitudinal studies on the amount and type of DF are warranted.
Collapse
Affiliation(s)
- Azadeh Hajipour
- Department of Nutrition, School of Health Qazvin University of Medical Sciences Qazvin Iran
| | - Maryam Afsharfar
- Department of Nutrition, School of Medicine Zahedan University of Medical Sciences Zahedan Iran
| | - Mona Jonoush
- Department of Nutrition, School of Medicine Mashhad University of Medical Sciences Mashhad Iran
| | - Mina Ahmadzadeh
- Department of Clinical Nutrition and Dietetics, Faculty of Nutrition and Food Technology, National Nutrition and Food Technology Research Institute Shahid Beheshti University of Medical Sciences Tehran Iran
| | - Maryam Gholamalizadeh
- School of Medicine, Cancer Research Center Shahid Beheshti University of Medical Sciences Tehran Iran
| | - Naeemeh Hassanpour Ardekanizadeh
- Department of Clinical Nutrition and Dietetics, Faculty of Nutrition and Food Technology, National Nutrition and Food Technology Research Institute Shahid Beheshti University of Medical Sciences Tehran Iran
| | - Saeid Doaei
- Reproductive Health Research Center, Department of Obstetrics and Gynecology, School of Medicine, Al‐Zahra Hospital Guilan University of Medical Sciences Rasht Iran
| | - Fatemeh Mohammadi‐Nasrabadi
- Department of Food and Nutrition Policy and Planning Research, Faculty of Nutrition Sciences and Food Technology, National Nutrition and Food Technology Research Institute Shahid Beheshti University of Medical Sciences Tehran Iran
| |
Collapse
|
18
|
Proteomics analysis of the hypothalamus of high-fat diet fed mice after Lactiplantibacillus plantarum Y44 administration. FOOD BIOSCI 2022. [DOI: 10.1016/j.fbio.2022.101762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
19
|
García-Carnero LC, Mora-Montes HM. Mucormycosis and COVID-19-Associated Mucormycosis: Insights of a Deadly but Neglected Mycosis. J Fungi (Basel) 2022; 8:445. [PMID: 35628701 PMCID: PMC9144279 DOI: 10.3390/jof8050445] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 04/22/2022] [Accepted: 04/22/2022] [Indexed: 02/04/2023] Open
Abstract
The ongoing COVID-19 pandemic has quickly become a health threat worldwide, with high mortality and morbidity among patients with comorbidities. This viral infection promotes the perfect setting in patients for the development of opportunistic infections, such as those caused by fungi. Mucormycosis, a rare but deadly fungal infection, has recently increased its incidence, especially in endemic areas, since the onset of the pandemic. COVID-19-associated mucormycosis is an important complication of the pandemic because it is a mycosis hard to diagnose and treat, causing concern among COVID-19-infected patients and even in the already recovered population. The risk factors for the development of mucormycosis in these patients are related to the damage caused by the SARS-CoV-2 itself, the patient's overstimulated immune response, and the therapy used to treat COVID-19, causing alterations such as hyperglycemia, acidosis, endothelial and lung damage, and immunosuppression. In this review, the molecular aspects of mucormycosis and the main risk factors for the development of COVID-19-associated mucormycosis are explained to understand this virus-fungi-host interaction and highlight the importance of this neglected mycosis.
Collapse
Affiliation(s)
- Laura C. García-Carnero
- Departamento de Biología, División de Ciencias Naturales y Exactas, Campus Guanajuato, Universidad de Guanajuato, Noria Alta s/n, col. Noria Alta, C.P., Guanajuato 36050, Mexico
| | - Héctor M. Mora-Montes
- Departamento de Biología, División de Ciencias Naturales y Exactas, Campus Guanajuato, Universidad de Guanajuato, Noria Alta s/n, col. Noria Alta, C.P., Guanajuato 36050, Mexico
| |
Collapse
|
20
|
Xu L, Ho CT, Liu Y, Wu Z, Zhang X. Potential Application of Tea Polyphenols to the Prevention of COVID-19 Infection: Based on the Gut-Lung Axis. Front Nutr 2022; 9:899842. [PMID: 35495940 PMCID: PMC9046984 DOI: 10.3389/fnut.2022.899842] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Accepted: 03/25/2022] [Indexed: 12/14/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19) disrupts the intestinal micro-ecological balance, and patients often develop the intestinal disease. The gut is the largest immune organ in the human body; intestinal microbes can affect the immune function of the lungs through the gut-lung axis. It has been reported that tea polyphenols (TPs) have antiviral and prebiotic activity. In this review, we discussed TPs reduced lung-related diseases through gut-lung axis by inhibiting dysbiosis. In addition, we also highlighted the preventive and therapeutic effects of TPs on COVID-19 complications, further demonstrating the importance of research on TPs for the prevention and treatment of COVID-19 in humans. Based on this understanding, we recommend using TPs to regulate the gut microbiota to prevent or alleviate COVID-19 through the gut-lung axis.
Collapse
Affiliation(s)
- Lei Xu
- Department of Food Science and Engineering, Ningbo University, Ningbo, China
| | - Chi-Tang Ho
- Department of Food Science, Rutgers University, New Brunswick, NJ, United States
- *Correspondence: Chi-Tang Ho
| | - Yanan Liu
- Department of Food Science and Engineering, Ningbo University, Ningbo, China
| | - Zufang Wu
- Department of Food Science and Engineering, Ningbo University, Ningbo, China
| | - Xin Zhang
- Department of Food Science and Engineering, Ningbo University, Ningbo, China
- Xin Zhang
| |
Collapse
|
21
|
Zhang L, Yao L, Guo Y, Li X, Ma L, Sun R, Han X, Liu J, Huang J. Oral SARS-CoV-2 Spike Protein Recombinant Yeast Candidate Prompts Specific Antibody and Gut Microbiota Reconstruction in Mice. Front Microbiol 2022; 13:792532. [PMID: 35464985 PMCID: PMC9022078 DOI: 10.3389/fmicb.2022.792532] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Accepted: 03/07/2022] [Indexed: 11/13/2022] Open
Abstract
A recent study showed that patients with coronavirus disease 2019 (COVID-19) have gastrointestinal symptoms and intestinal flora dysbiosis. Yeast probiotics shape the gut microbiome and improve immune homeostasis. In this study, an oral candidate of yeast-derived spike protein receptor-binding domain (RBD) and fusion peptide displayed on the surface of the yeast cell wall was generated. The toxicity and immune efficacy of oral administration were further performed in Institute of Cancer Research (ICR) mice. No significant difference in body weights, viscera index, and other side effects were detected in the oral-treated group. The detectable RBD-specific immunoglobulin G (IgG) and immunoglobulin A (IgA) of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and more complex microbiota were detected from oral administration mice compared with those of the control group. Interestingly, the recombinant yeast was identified in female fetal of the high-dose group. These results revealed that the displaying yeast could fulfill the agent-driven immunoregulation and gut microbiome reconstitution. The findings will shed light on new dimensions against SARS-CoV-2 infection with the synergistic oral agents as promising non-invasive immunization and restoring gut flora.
Collapse
Affiliation(s)
- Lilin Zhang
- School of Life Sciences, Tianjin University, Tianjin, China
| | - Lan Yao
- School of Life Sciences, Tianjin University, Tianjin, China
| | - Yanyu Guo
- School of Life Sciences, Tianjin University, Tianjin, China
| | - Xiaoyang Li
- School of Life Sciences, Tianjin University, Tianjin, China
| | - Li Ma
- Tianjin Institute of Pharmaceutical Research Co., Ltd., Tianjin, China
| | - Ruiqi Sun
- School of Life Sciences, Tianjin University, Tianjin, China
| | - Xueqing Han
- Chinese Academy of Inspection and Quarantine, Beijing, China
| | - Jing Liu
- Tianjin Institute of Pharmaceutical Research Co., Ltd., Tianjin, China
- Jing Liu,
| | - Jinhai Huang
- School of Life Sciences, Tianjin University, Tianjin, China
- *Correspondence: Jinhai Huang,
| |
Collapse
|
22
|
Pérez-Cobas AE, Baquero F, de Pablo R, Soriano MC, Coque TM. Altered Ecology of the Respiratory Tract Microbiome and Nosocomial Pneumonia. Front Microbiol 2022; 12:709421. [PMID: 35222291 PMCID: PMC8866767 DOI: 10.3389/fmicb.2021.709421] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Accepted: 12/21/2021] [Indexed: 12/23/2022] Open
Abstract
Nosocomial pneumonia is one of the most frequent infections in critical patients. It is primarily associated with mechanical ventilation leading to severe illness, high mortality, and prolonged hospitalization. The risk of mortality has increased over time due to the rise in multidrug-resistant (MDR) bacterial infections, which represent a global public health threat. Respiratory tract microbiome (RTM) research is growing, and recent studies suggest that a healthy RTM positively stimulates the immune system and, like the gut microbiome, can protect against pathogen infection through colonization resistance (CR). Physiological conditions of critical patients and interventions as antibiotics administration and mechanical ventilation dramatically alter the RTM, leading to dysbiosis. The dysbiosis of the RTM of ICU patients favors the colonization by opportunistic and resistant pathogens that can be part of the microbiota or acquired from the hospital environments (biotic or built ones). Despite recent evidence demonstrating the significance of RTM in nosocomial infections, most of the host-RTM interactions remain unknown. In this context, we present our perspective regarding research in RTM altered ecology in the clinical environment, particularly as a risk for acquisition of nosocomial pneumonia. We also reflect on the gaps in the field and suggest future research directions. Moreover, expected microbiome-based interventions together with the tools to study the RTM highlighting the "omics" approaches are discussed.
Collapse
Affiliation(s)
- Ana Elena Pérez-Cobas
- Department of Microbiology, Ramón y Cajal Institute for Health Research (IRYCIS), Ramón y Cajal University Hospital, Madrid, Spain
| | - Fernando Baquero
- Department of Microbiology, Ramón y Cajal Institute for Health Research (IRYCIS), Ramón y Cajal University Hospital, Madrid, Spain.,CIBER in Epidemiology and Public Health (CIBERESP), Madrid, Spain
| | - Raúl de Pablo
- Intensive Care Department, Ramón y Cajal University Hospital, Madrid, Spain
| | - María Cruz Soriano
- Intensive Care Department, Ramón y Cajal University Hospital, Madrid, Spain
| | - Teresa M Coque
- Department of Microbiology, Ramón y Cajal Institute for Health Research (IRYCIS), Ramón y Cajal University Hospital, Madrid, Spain.,CIBER in Infectious Diseases (CIBERINFEC), Madrid, Spain
| |
Collapse
|
23
|
Foolchand A, Ghazi T, Chuturgoon AA. Malnutrition and Dietary Habits Alter the Immune System Which May Consequently Influence SARS-CoV-2 Virulence: A Review. Int J Mol Sci 2022; 23:2654. [PMID: 35269795 PMCID: PMC8910702 DOI: 10.3390/ijms23052654] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 02/22/2022] [Accepted: 02/23/2022] [Indexed: 02/06/2023] Open
Abstract
COVID-19, resulting from the SARS-CoV-2 virus, is a major pandemic that the world is fighting. SARS-CoV-2 primarily causes lung infection by attaching to the ACE2 receptor on the alveolar epithelial cells. However, the ACE2 receptor is also present in intestinal epithelial cells, suggesting a link between nutrition, virulence and clinical outcomes of COVID-19. Respiratory viral infections perturb the gut microbiota. The gut microbiota is shaped by our diet; therefore, a healthy gut is important for optimal metabolism, immunology and protection of the host. Malnutrition causes diverse changes in the immune system by repressing immune responses and enhancing viral vulnerability. Thus, improving gut health with a high-quality, nutrient-filled diet will improve immunity against infections and diseases. This review emphasizes the significance of dietary choices and its subsequent effects on the immune system, which may potentially impact SARS-CoV-2 vulnerability.
Collapse
Affiliation(s)
| | | | - Anil A. Chuturgoon
- Discipline of Medical Biochemistry, School of Laboratory Medicine and Medical Science, College of Health Sciences, Howard College Campus, University of Kwa-Zulu Natal, Durban 4041, South Africa; (A.F.); (T.G.)
| |
Collapse
|
24
|
Algal Metabolites Can Be an Immune Booster against COVID-19 Pandemic. Antioxidants (Basel) 2022; 11:antiox11030452. [PMID: 35326102 PMCID: PMC8944855 DOI: 10.3390/antiox11030452] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Revised: 02/22/2022] [Accepted: 02/23/2022] [Indexed: 02/05/2023] Open
Abstract
The world has faced the challenges of Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) for the last two years, first diagnosed at the end of 2019 in Wuhan and widely distributed worldwide. As a result, the WHO has proclaimed the illness brought on by this virus to be a global pandemic. To combat COVID-19, researcher communities continuously develop and implement rapid diagnoses, safe and effective vaccinations and other alternative therapeutic procedures. However, synthetic drug-related side effects and high costs have piqued scientists’ interest in natural product-based therapies and medicines. In this regard, antiviral substances derived from natural resources and some medicines have seen a boom in popularity. For instance, algae are a rich source of compounds such as lectins and sulfated polysaccharides, which have potent antiviral and immunity-boosting properties. Moreover, Algae-derived compounds or metabolites can be used as antibodies and vaccine raw materials against COVID-19. Furthermore, some algal species can boost immunity, reduce viral activity in humans and be recommended for usage as a COVID-19 preventative measure. However, this field of study is still in its early stages of development. Therefore, this review addresses critical characteristics of algal metabolites, their antioxidant potential and therapeutic potential in COVID-19.
Collapse
|
25
|
Tea Polyphenols Prevent and Intervene in COVID-19 through Intestinal Microbiota. Foods 2022; 11:foods11040506. [PMID: 35205982 PMCID: PMC8871045 DOI: 10.3390/foods11040506] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 01/22/2022] [Accepted: 02/05/2022] [Indexed: 12/13/2022] Open
Abstract
Although all countries have taken corresponding measures, the coronavirus disease 2019 (COVID-19) is still ravaging the world. To consolidate the existing anti-epidemic results and further strengthen the prevention and control measures against the new coronavirus, we are now actively pioneering a novel research idea of regulating the intestinal microbiota through tea polyphenols for reference. Although studies have long revealed the regulatory effect of tea polyphenols on the intestinal microbiota to various gastrointestinal inflammations, little is known about the prevention and intervention of COVID-19. This review summarizes the possible mechanism of the influence of tea polyphenols on COVID-19 mediated by the intestinal microbiota. In this review, the latest studies of tea polyphenols exhibiting their own antibacterial and anti-inflammatory activities and protective effects on the intestinal mucosal barrier are combed through and summarized. Among them, (−)-epigallocatechin-3-gallate (EGCG), one of the main monomers of catechins, may be activated as nuclear factor erythroid 2 p45-related factor 2 (Nrf2). The agent inhibits the expression of ACE2 (a cellular receptor for SARS-CoV-2) and TMPRSS2 to inhibit SARS-CoV-2 infection, inhibiting the life cycle of SARS-CoV-2. Thus, preliminary reasoning and judgments have been made about the possible mechanism of the effect of tea polyphenols on the COVID-19 control and prevention mediated by the microbiota. These results may be of great significance to the future exploration of specialized research in this field.
Collapse
|
26
|
Alibeik N, Pishgar E, Bozorgmehr R, Aghaaliakbari F, Rahimian N. Potential role of gut microbiota in patients with COVID-19, its relationship with lung axis, central nervous system (CNS) axis, and improvement with probiotic therapy. IRANIAN JOURNAL OF MICROBIOLOGY 2022; 14:1-9. [PMID: 35611351 PMCID: PMC9085538 DOI: 10.18502/ijm.v14i1.8794] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Coronavirus Disease 2019 (COVID-19) is a pandemic disease caused by a new corona virus. COVID-19 affects different people in different ways. COVID-19 could affect the gastrointestinal system via gut microbiota impairment. Gut microbiota could affect lung health through a relationship between gut and lung microbiota, which is named gut-lung axis. Gut microbiota impairment plays a role in pathogenesis of various pulmonary disease states, so GI diseases were found to be associated with respiratory diseases. Moreover, most infected people will develop mild to moderate gastrointestinal (GI) symptoms such as diarrhea, vomiting, and stomachache, which is caused by impairment in gut microbiota. Therefore, the current study aimed to review potential role of gut microbiota in patients with COVID-19, its relation with lung axis, Central Nervous System (CNS) axis and improvement with probiotic therapy. Also, this review can be a guide for potential role of gut microbiota in patients with COVID-19.
Collapse
Affiliation(s)
- Nazanin Alibeik
- Department of Internal Medicine, School of Medicine, Firoozgar Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Elham Pishgar
- Gastrointestinal and Liver Diseases Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Ramin Bozorgmehr
- Department of Surgery, School of Medicine, Shahid Madani Hospital, Alborz University of Medical Sciences, Karaj, Iran
| | - Farshad Aghaaliakbari
- Department of Internal Medicine, School of Medicine, Firoozgar Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Neda Rahimian
- Department of Internal Medicine, School of Medicine, Firoozgar Hospital, Iran University of Medical Sciences, Tehran, Iran
- Endocrine Research Center, Institute of Endocrinology and Metabolism, Iran University of Medical Sciences (IUMS), Tehran, Iran
| |
Collapse
|
27
|
Samson R, Dharne M. COVID-19 associated mucormycosis: evolving technologies for early and rapid diagnosis. 3 Biotech 2022; 12:6. [PMID: 34900512 PMCID: PMC8647065 DOI: 10.1007/s13205-021-03080-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 11/26/2021] [Indexed: 12/12/2022] Open
Abstract
The post-coronavirus disease (COVID-19) mucormycosis is a deadly addition to the pandemic spectrum. Although it’s a rare, aggressive, and opportunistic disease, the associated morbidity and mortality are significant. The complex interplay of factors aggravating CAM is uncontrolled diabetes, irrational and excessive use of antibiotics, steroids, and an impaired immune system. Recently, India has been witnessing a rapid surge in the cases of coronavirus disease-associated mucormycosis (CAM), since the second wave of COVID-19. The devastating and lethal implications of CAM had now become a matter of global attention. A delayed diagnosis is often associated with a poor prognosis. Therefore, the rapid and early diagnosis of infection would be life-saving. Prevention and effective management of mucormycosis depend upon its early and accurate diagnosis followed by a multimodal therapeutic approach. The current review summarizes an array of detection methods and highlights certain evolving technologies for early and rapid diagnosis of CAM. Furthermore, several potential management strategies have also been discussed, which would aid in tackling the neglected yet fatal crisis of mucormycosis associated with COVID-19.
Collapse
|
28
|
Tosta E. The seven constitutive respiratory defense barriers against SARS-CoV-2 infection. Rev Soc Bras Med Trop 2021; 54:e04612021. [PMID: 34932765 PMCID: PMC8687496 DOI: 10.1590/0037-8682-0461-2021] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 09/17/2021] [Indexed: 12/18/2022] Open
Abstract
Before eliciting an adaptive immune response, SARS-CoV-2 must overcome seven constitutive respiratory defense barriers. The first is the mucus covering the respiratory tract's luminal surface, which entraps inhaled particles, including infectious agents, and eliminates them by mucociliary clearance. The second barrier comprises various components present in the airway lining fluid, the surfactants. Besides providing low surface tension that allows efficient gas exchange at the alveoli, surfactants inhibit the invasion of epithelial cells by respiratory viruses, enhance pathogen uptake by phagocytes, and regulate immune cells' functions. The respiratory tract microbiota constitutes the third defense barrier against SARS-CoV-2. It activates the innate and adaptive immune cells and elicits anti-infectious molecules such as secretory IgA antibodies, defensins, and interferons. The fourth defense barrier comprises the antimicrobial peptides defensins, and lactoferrin. They show direct antiviral activity, inhibit viral fusion, and modulate the innate and adaptive immune responses. Secretory IgA antibodies, the fifth defense barrier, besides protecting the local microbiota against noxious agents, also inhibit SARS-CoV-2 cell invasion. If the virus overcomes this barrier, it reaches its target, the respiratory epithelial cells. However, these cells also act as a defense barrier, the sixth one, since they hinder the virus' access to receptors and produce antiviral and immunomodulatory molecules such as interferons, lactoferrin, and defensins. Finally, the sensing of the virus by the cells of innate immunity, the last constitutive defense barrier, elicits a cascade of signals that activate adaptive immune cells and may inhibit the development of productive infection. The subject of the present essay is discussing these mechanisms.
Collapse
Affiliation(s)
- Eduardo Tosta
- Professor Emeritus, Faculdade de Medicina, Universidade de Brasília, Brasília, DF , Brasil
| |
Collapse
|
29
|
Deschasaux-Tanguy M, Srour B, Bourhis L, Arnault N, Druesne-Pecollo N, Esseddik Y, de Edelenyi FS, Allègre J, Allès B, Andreeva VA, Baudry J, Fezeu LK, Galan P, Julia C, Kesse-Guyot E, Péneau S, Hercberg S, Bajos N, Severi G, Zins M, de Lamballerie X, Carrat F, Touvier M. Nutritional risk factors for SARS-CoV-2 infection: a prospective study within the NutriNet-Santé cohort. BMC Med 2021; 19:290. [PMID: 34844606 PMCID: PMC8629697 DOI: 10.1186/s12916-021-02168-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 10/21/2021] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Nutritional factors are essential for the functioning of the immune system and could therefore play a role in COVID-19 but evidence is needed. Our objective was to study the associations between diet and the risk of SARS-CoV-2 infection in a large population-based sample. METHODS Our analyses were conducted in the French prospective NutriNet-Santé cohort study (2009-2020). Seroprevalence of anti-SARS-CoV-2 antibodies was assessed by ELISA on dried blood spots. Dietary intakes were derived from repeated 24 h dietary records (at least 6) in the two years preceding the start of the COVID-19 pandemic in France (February 2020). Multi-adjusted logistic regression models were computed. RESULTS A total of 7766 adults (70.3% women, mean age: 60.3 years) were included, among which 311 were positive for anti-SARS-CoV-2 antibodies. Dietary intakes of vitamin C (OR for 1 SD=0.86 (0.75-0.98), P=0.02), vitamin B9 (OR=0.84 (0.72-0.98), P=0.02), vitamin K (OR=0.86 (0.74-0.99), P=0.04), fibers (OR=0.84 (0.72-0.98), P=0.02), and fruit and vegetables (OR=0.85 (0.74-0.97), P=0.02) were associated to a decreased probability of SARS-CoV-2 infection while dietary intakes of calcium (OR=1.16 (1.01-1.35), P=0.04) and dairy products (OR=1.19 (1.06-1.33), P=0.002) associated to increased odds. No association was detected with other food groups or nutrients or with the overall diet quality. CONCLUSIONS Higher dietary intakes of fruit and vegetables and, consistently, of vitamin C, folate, vitamin K and fibers were associated with a lower susceptibility to SARS-CoV-2 infection. Beyond its established role in the prevention of non-communicable diseases, diet could therefore also contribute to prevent some infectious diseases such as COVID-19.
Collapse
Affiliation(s)
- Mélanie Deschasaux-Tanguy
- Sorbonne Paris Nord University, Inserm U1153, Inrae U1125, Cnam, Nutritional Epidemiology Research Team (EREN), Epidemiology and Statistics Research Center - University of Paris (CRESS), Bobigny, France
| | - Bernard Srour
- Sorbonne Paris Nord University, Inserm U1153, Inrae U1125, Cnam, Nutritional Epidemiology Research Team (EREN), Epidemiology and Statistics Research Center - University of Paris (CRESS), Bobigny, France.
| | - Laurent Bourhis
- Sorbonne Paris Nord University, Inserm U1153, Inrae U1125, Cnam, Nutritional Epidemiology Research Team (EREN), Epidemiology and Statistics Research Center - University of Paris (CRESS), Bobigny, France
| | - Nathalie Arnault
- Sorbonne Paris Nord University, Inserm U1153, Inrae U1125, Cnam, Nutritional Epidemiology Research Team (EREN), Epidemiology and Statistics Research Center - University of Paris (CRESS), Bobigny, France
| | - Nathalie Druesne-Pecollo
- Sorbonne Paris Nord University, Inserm U1153, Inrae U1125, Cnam, Nutritional Epidemiology Research Team (EREN), Epidemiology and Statistics Research Center - University of Paris (CRESS), Bobigny, France
| | - Younes Esseddik
- Sorbonne Paris Nord University, Inserm U1153, Inrae U1125, Cnam, Nutritional Epidemiology Research Team (EREN), Epidemiology and Statistics Research Center - University of Paris (CRESS), Bobigny, France
| | - Fabien Szabo de Edelenyi
- Sorbonne Paris Nord University, Inserm U1153, Inrae U1125, Cnam, Nutritional Epidemiology Research Team (EREN), Epidemiology and Statistics Research Center - University of Paris (CRESS), Bobigny, France
| | - Julien Allègre
- Sorbonne Paris Nord University, Inserm U1153, Inrae U1125, Cnam, Nutritional Epidemiology Research Team (EREN), Epidemiology and Statistics Research Center - University of Paris (CRESS), Bobigny, France
| | - Benjamin Allès
- Sorbonne Paris Nord University, Inserm U1153, Inrae U1125, Cnam, Nutritional Epidemiology Research Team (EREN), Epidemiology and Statistics Research Center - University of Paris (CRESS), Bobigny, France
| | - Valentina A Andreeva
- Sorbonne Paris Nord University, Inserm U1153, Inrae U1125, Cnam, Nutritional Epidemiology Research Team (EREN), Epidemiology and Statistics Research Center - University of Paris (CRESS), Bobigny, France
| | - Julia Baudry
- Sorbonne Paris Nord University, Inserm U1153, Inrae U1125, Cnam, Nutritional Epidemiology Research Team (EREN), Epidemiology and Statistics Research Center - University of Paris (CRESS), Bobigny, France
| | - Leopold K Fezeu
- Sorbonne Paris Nord University, Inserm U1153, Inrae U1125, Cnam, Nutritional Epidemiology Research Team (EREN), Epidemiology and Statistics Research Center - University of Paris (CRESS), Bobigny, France
| | - Pilar Galan
- Sorbonne Paris Nord University, Inserm U1153, Inrae U1125, Cnam, Nutritional Epidemiology Research Team (EREN), Epidemiology and Statistics Research Center - University of Paris (CRESS), Bobigny, France
| | - Chantal Julia
- Sorbonne Paris Nord University, Inserm U1153, Inrae U1125, Cnam, Nutritional Epidemiology Research Team (EREN), Epidemiology and Statistics Research Center - University of Paris (CRESS), Bobigny, France
| | - Emmanuelle Kesse-Guyot
- Sorbonne Paris Nord University, Inserm U1153, Inrae U1125, Cnam, Nutritional Epidemiology Research Team (EREN), Epidemiology and Statistics Research Center - University of Paris (CRESS), Bobigny, France
| | - Sandrine Péneau
- Sorbonne Paris Nord University, Inserm U1153, Inrae U1125, Cnam, Nutritional Epidemiology Research Team (EREN), Epidemiology and Statistics Research Center - University of Paris (CRESS), Bobigny, France
| | - Serge Hercberg
- Sorbonne Paris Nord University, Inserm U1153, Inrae U1125, Cnam, Nutritional Epidemiology Research Team (EREN), Epidemiology and Statistics Research Center - University of Paris (CRESS), Bobigny, France
| | - Nathalie Bajos
- IRIS, UMR CNRS 8156, EHESS, Inserm U997, Aubervilliers, France
| | - Gianluca Severi
- Paris-Saclay University, UVSQ, Inserm, Gustave Roussy, "Exposome and Heredity" team, CESP UMR1018, Villejuif, France
- Department of Statistics, Computer Science and Applications "G. Parenti", University of Florence, Florence, Italy
| | - Marie Zins
- Paris University, Paris, France
- Inserm UMS 11, Paris Saclay University, Villejuif, France
| | - Xavier de Lamballerie
- Unité des Virus Emergents (UVE), Aix Marseille Univ, IRD 190, INSERM 1207, IHU Méditerranée Infection, Marseille, France
| | - Fabrice Carrat
- Sorbonne Université, Inserm, Institut Pierre-Louis d'Epidémiologie et de Santé Publique, Paris, France
- Département de Santé Publique, APHP, Sorbonne Université, Paris, France
| | - Mathilde Touvier
- Sorbonne Paris Nord University, Inserm U1153, Inrae U1125, Cnam, Nutritional Epidemiology Research Team (EREN), Epidemiology and Statistics Research Center - University of Paris (CRESS), Bobigny, France
| |
Collapse
|
30
|
Al Bataineh MT, Henschel A, Mousa M, Daou M, Waasia F, Kannout H, Khalili M, Kayasseh MA, Alkhajeh A, Uddin M, Alkaabi N, Tay GK, Feng SF, Yousef AF, Alsafar HS. Gut Microbiota Interplay With COVID-19 Reveals Links to Host Lipid Metabolism Among Middle Eastern Populations. Front Microbiol 2021; 12:761067. [PMID: 34803986 PMCID: PMC8603808 DOI: 10.3389/fmicb.2021.761067] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 09/30/2021] [Indexed: 12/15/2022] Open
Abstract
The interplay between the compositional changes in the gastrointestinal microbiome, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) susceptibility and severity, and host functions is complex and yet to be fully understood. This study performed 16S rRNA gene-based microbial profiling of 143 subjects. We observed structural and compositional alterations in the gut microbiota of the SARS-CoV-2-infected group in comparison to non-infected controls. The gut microbiota composition of the SARS-CoV-2-infected individuals showed an increase in anti-inflammatory bacteria such as Faecalibacterium (p-value = 1.72 × 10-6) and Bacteroides (p-value = 5.67 × 10-8). We also revealed a higher relative abundance of the highly beneficial butyrate producers such as Anaerostipes (p-value = 1.75 × 10-230), Lachnospiraceae (p-value = 7.14 × 10-65), and Blautia (p-value = 9.22 × 10-18) in the SARS-CoV-2-infected group in comparison to the control group. Moreover, phylogenetic investigation of communities by reconstructing unobserved state (PICRUSt) functional prediction analysis of the 16S rRNA gene abundance data showed substantial differences in the enrichment of metabolic pathways such as lipid, amino acid, carbohydrate, and xenobiotic metabolism, in comparison between both groups. We discovered an enrichment of linoleic acid, ether lipid, glycerolipid, and glycerophospholipid metabolism in the SARS-CoV-2-infected group, suggesting a link to SARS-CoV-2 entry and replication in host cells. We estimate the major contributing genera to the four pathways to be Parabacteroides, Streptococcus, Dorea, and Blautia, respectively. The identified differences provide a new insight to enrich our understanding of SARS-CoV-2-related changes in gut microbiota, their metabolic capabilities, and potential screening biomarkers linked to COVID-19 disease severity.
Collapse
Affiliation(s)
- Mohammad Tahseen Al Bataineh
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah, United Arab Emirates.,Department of Clinical Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates.,Center for Biotechnology, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates.,Department of Genetics and Molecular Biology, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Andreas Henschel
- Center for Biotechnology, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates.,Department of Electrical Engineering and Computer Science, Khalifa University, Abu Dhabi, United Arab Emirates
| | - Mira Mousa
- Center for Biotechnology, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates.,Nuffield Department of Women's and Reproduction Health, Oxford University, Oxford, United Kingdom
| | - Marianne Daou
- Department of Chemistry, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Fathimathuz Waasia
- Center for Biotechnology, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Hussein Kannout
- Center for Biotechnology, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Mariam Khalili
- Center for Biotechnology, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Mohd Azzam Kayasseh
- Emirates Specialty Hospital, Dubai Healthcare City, Dubai, United Arab Emirates
| | - Abdulmajeed Alkhajeh
- Medical Education and Research Department, Dubai Health Authority, Dubai, United Arab Emirates
| | - Maimunah Uddin
- Department of Pediatric Infectious Disease, Sheikh Khalifa Medical City, Abu Dhabi, United Arab Emirates
| | - Nawal Alkaabi
- Department of Pediatric Infectious Disease, Sheikh Khalifa Medical City, Abu Dhabi, United Arab Emirates
| | - Guan K Tay
- Division of Psychiatry, Faculty of Health and Medical Sciences, The University of Western Australia, Crawley, WA, Australia.,School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia
| | - Samuel F Feng
- Center for Biotechnology, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates.,Department of Mathematics, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Ahmed F Yousef
- Department of Chemistry, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Habiba S Alsafar
- Center for Biotechnology, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates.,Department of Mathematics, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates.,Department of Biomedical Engineering, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| |
Collapse
|
31
|
He LH, Yao DH, Wang LY, Zhang L, Bai XL. Gut Microbiome-Mediated Alteration of Immunity, Inflammation, and Metabolism Involved in the Regulation of Non-alcoholic Fatty Liver Disease. Front Microbiol 2021; 12:761836. [PMID: 34795655 PMCID: PMC8593644 DOI: 10.3389/fmicb.2021.761836] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Accepted: 10/05/2021] [Indexed: 12/12/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is one of the leading causes of end-stage liver disease, leading to a rapidly growing global public health burden. The term “gut microbiome (GM)” refers to the approximately 100 trillion microbial cells that inhabit the host’s gastrointestinal tract. There is increasing evidence that GM is involved in the pathogenesis of NAFLD and may be a potential target for intervention. To explore GM-based strategies for precise diagnosis and treatment of NAFLD, great efforts have been made to develop a comprehensive and in-depth understanding of the host–microbe interaction. This review evaluates this interaction critically, mainly considering the intricate regulation of the metabolism, immunity, and inflammatory status during the evolution of the disease pathogenesis, revealing roles for the GM in NAFLD by examining advances in potential mechanisms, diagnostics, and modulation strategies. Synopsis: Considering the intricate metabolic and immune/inflammatory homeostasis regulation, we evaluate the latest understanding of the host–microbe interaction and reveal roles for the gastrointestinal microbiome in NAFLD. Strategies targeting the gastrointestinal microbiome for the diagnosis and treatment of NAFLD are proposed.
Collapse
Affiliation(s)
- Li-Hong He
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,The First Clinical Medical College, Lanzhou University, Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, China
| | - Dun-Han Yao
- The First Clinical Medical College, Lanzhou University, Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, China
| | - Ling-Yun Wang
- The First Clinical Medical College, Lanzhou University, Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, China
| | - Lei Zhang
- The First Clinical Medical College, Lanzhou University, Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, China
| | - Xue-Li Bai
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
32
|
Utay NS, Asmuth DM, Gharakhanian S, Contreras M, Warner CD, Detzel CJ. Potential use of serum-derived bovine immunoglobulin/protein isolate for the management of COVID-19. Drug Dev Res 2021; 82:873-879. [PMID: 34110032 PMCID: PMC9293029 DOI: 10.1002/ddr.21841] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 05/13/2021] [Accepted: 06/01/2021] [Indexed: 11/23/2022]
Abstract
COVID-19 manifests as a mild disease in most people but can progress to severe disease in nearly 20% of individuals. Disease progression is likely driven by a cytokine storm, either directly stimulated by SARS-CoV-2 or by increased systemic inflammation in which the gut might play an integral role. SARS-CoV-2 replication in the gut may cause increased intestinal permeability, alterations to the fecal microbiome, and increased inflammatory cytokines. Each effect may lead to increased systemic inflammation and the transport of cytokines and inflammatory antigens from the gut to the lung. Few interventions are being studied to treat people with mild disease and prevent the cytokine storm. Serumderived bovine immunoglobulin/protein isolate (SBI) may prevent progression by (1) binding and neutralizing inflammatory antigens, (2) decreasing gut permeability, (3) interfering with ACE2 binding by viral proteins, and (4) improving the fecal microbiome. SBI is therefore a promising intervention to prevent disease progression in COVID-19 patients.
Collapse
Affiliation(s)
- Netanya S. Utay
- Department of Internal MedicineMcGovern Medical School, University of Texas Health Science Center at HoustonHoustonTexasUSA
| | - David M. Asmuth
- Department of Internal MedicineUniversity of California DavisSacramentoCaliforniaUSA
| | - Shahin Gharakhanian
- Pharmaceutical Medicine & Infectious DiseasesShahin Gharakhanian MD Consulting LLC, Cambridge Innovation Center (CIC)CambridgeMassachusettsUSA
| | - Moises Contreras
- Department of Research and DevelopmentEntera Health, LLCAnkenyIowaUSA
| | | | | |
Collapse
|
33
|
Rahban M, Stanek A, Hooshmand A, Khamineh Y, Ahi S, Kazim SN, Ahmad F, Muronetz V, Samy Abousenna M, Zolghadri S, Saboury AA. Infection of Human Cells by SARS-CoV-2 and Molecular Overview of Gastrointestinal, Neurological, and Hepatic Problems in COVID-19 Patients. J Clin Med 2021; 10:4802. [PMID: 34768321 PMCID: PMC8584649 DOI: 10.3390/jcm10214802] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 10/12/2021] [Accepted: 10/16/2021] [Indexed: 02/07/2023] Open
Abstract
The gastrointestinal tract is the body's largest interface between the host and the external environment. People infected with SARS-CoV-2 are at higher risk of microbiome alterations and severe diseases. Recent evidence has suggested that the pathophysiological and molecular mechanisms associated with gastrointestinal complicity in SARS-CoV-2 infection could be explained by the role of angiotensin-converting enzyme-2 (ACE2) cell receptors. These receptors are overexpressed in the gut lining, leading to a high intestinal permeability to foreign pathogens. It is believed that SARS-CoV-2 has a lesser likelihood of causing liver infection because of the diminished expression of ACE2 in liver cells. Interestingly, an interconnection between the lungs, brain, and gastrointestinal tract during severe COVID-19 has been mentioned. We hope that this review on the molecular mechanisms related to the gastrointestinal disorders as well as neurological and hepatic manifestations experienced by COVID-19 patients will help scientists to find a convenient solution for this and other pandemic events.
Collapse
Affiliation(s)
- Mahdie Rahban
- Institute of Biochemistry and Biophysics, University of Tehran, Tehran 1417614335, Iran;
| | - Agata Stanek
- Department of Internal Medicine, Angiology and Physical Medicine, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, Batorego 15 St., 41-902 Bytom, Poland;
| | - Amirreza Hooshmand
- Young Researchers and Elite Club, Jahrom Branch, Islamic Azad University, Jahrom 7414785318, Iran; (A.H.); (Y.K.)
| | - Yasaman Khamineh
- Young Researchers and Elite Club, Jahrom Branch, Islamic Azad University, Jahrom 7414785318, Iran; (A.H.); (Y.K.)
| | - Salma Ahi
- Research Center for Noncommunicable Diseases, Jahrom University of Medical Sciences, Jahrom 7414846199, Iran;
| | - Syed Naqui Kazim
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi 110025, India; (S.N.K.); (F.A.)
| | - Faizan Ahmad
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi 110025, India; (S.N.K.); (F.A.)
| | - Vladimir Muronetz
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119234 Moscow, Russia;
| | - Mohamed Samy Abousenna
- Central Laboratory for Evaluation of Veterinary Biologics, Agriculture Research Center, Cairo 11517, Egypt;
| | - Samaneh Zolghadri
- Department of Biology, Jahrom Branch, Islamic Azad University, Jahrom 7414785318, Iran
| | - Ali A. Saboury
- Institute of Biochemistry and Biophysics, University of Tehran, Tehran 1417614335, Iran;
| |
Collapse
|
34
|
Cruz CS, Ricci MF, Vieira AT. Gut Microbiota Modulation as a Potential Target for the Treatment of Lung Infections. Front Pharmacol 2021; 12:724033. [PMID: 34557097 PMCID: PMC8453009 DOI: 10.3389/fphar.2021.724033] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Accepted: 08/20/2021] [Indexed: 12/12/2022] Open
Abstract
The gastrointestinal and respiratory systems are colonized by a complex ecosystem of microorganisms called the microbiota. These microorganisms co-evolved over millions of years with the host, creating a symbiotic relationship that is fundamental for promoting host homeostasis by producing bioactive metabolites and antimicrobial molecules, and regulating the immune and inflammatory responses. Imbalance in the abundance, diversity, and function of the gut microbiota (known as dysbiosis) have been shown to increase host susceptibility to infections in the lungs, suggesting crosstalk between these organs. This crosstalk is now referred to as the gut-lung axis. Hence, the use of probiotics, prebiotics, and synbiotics for modulation of gut microbiota has been studied based on their effectiveness in reducing the duration and severity of respiratory tract infections, mainly owing to their effects on preventing pathogen colonization and modulating the immune system. This review discusses the role and responses of probiotics, prebiotics, and synbiotics in the gut-lung axis in the face of lung infections.
Collapse
Affiliation(s)
- Clênio Silva Cruz
- Laboratory of Microbiota and Immunomodulation (LMI), Department of Biochemistry and Immunology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Mayra Fernanda Ricci
- Laboratory of Microbiota and Immunomodulation (LMI), Department of Biochemistry and Immunology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Angélica Thomaz Vieira
- Laboratory of Microbiota and Immunomodulation (LMI), Department of Biochemistry and Immunology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil
| |
Collapse
|
35
|
Bo W, Xi Y, Tian Z. The role of exercise in rehabilitation of discharged COVID-19 patients. SPORTS MEDICINE AND HEALTH SCIENCE 2021; 3:194-201. [PMID: 34541561 PMCID: PMC8438861 DOI: 10.1016/j.smhs.2021.09.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Revised: 09/04/2021] [Accepted: 09/05/2021] [Indexed: 01/08/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) mainly caused pneumonia and pulmonary fibrosis through upper respiratory tract infection, which resulted in acute respiratory distress syndrome (ARDS) and multiorgan damage of cardiovascular, nervous, digestive, and genitourinary systems. Although the virus test turned negative after the patient recovered, the damage to multiorgan caused by SARS-CoV-2 may irreversible. Therefore, the health status of the recovered patients has gradually become the focus of people's attention. Whether coronavirus disease 2019 (COVID-19) patients can receive exercise rehabilitation training after discharge? and what's the basis? We try to analyze and answer these questions, will provide some ideas about the patients to develop a reasonable and effective exercise rehabilitation program.
Collapse
Key Words
- ACE2, angiotensin-converting enzyme 2
- ACS, acute coronary syndrome
- AKI, acute kidney injury
- ARDS, acute respiratory distress syndrome
- Ang II, accumulation of angiotensin Ⅱ
- BDNF, brain-derived neurotrophic factor
- CHI3L1, chitinase-3-like protein 1
- CNS, central nervous system
- COPD, chronic obstructive pulmonary disease
- COVID-19
- COVID-19, coronavirus disease 2019
- CRS, cytokine release syndrome
- EcSOD, extracellular superoxide dismutase
- Exercise rehabilitation
- Exercise response factor
- FGF21, fibroblast growth factor 21
- FSTL1, follistatin-related protein 1
- GDF-15, growth/differentiation factor-15
- ICU, intensive care unit
- Inflammation cytokine storm
- LIF, leukemia inhibitory factor
- NK, natural killer
- PBMCs, peripheral blood mononuclear cells
- RAAS, renin-angiotensin-aldosterone system
- ROS, reactive oxygen species
- SARS-CoV-2
- SARS-CoV-2, severe acute respiratory syndrome coronavirus 2
- TMPRSS2, transmembrane protease serine 2
- TNF, tumor necrosis factor
- WBV, whole-body vibration
Collapse
Affiliation(s)
- Wenyan Bo
- Institute of Sports and Exercise Biology, School of Physical Education, Shaanxi Normal University, Xi'an, 710119, China
| | - Yue Xi
- Institute of Sports and Exercise Biology, School of Physical Education, Shaanxi Normal University, Xi'an, 710119, China
| | - Zhenjun Tian
- Institute of Sports and Exercise Biology, School of Physical Education, Shaanxi Normal University, Xi'an, 710119, China
| |
Collapse
|
36
|
Mozota M, Castro I, Gómez-Torres N, Arroyo R, Lailla Y, Somada M, Alba C, Rodríguez JM. Administration of Ligilactobacillus salivarius MP101 in an Elderly Nursing Home during the COVID-19 Pandemic: Immunological and Nutritional Impact. Foods 2021; 10:2149. [PMID: 34574259 PMCID: PMC8470390 DOI: 10.3390/foods10092149] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 09/05/2021] [Accepted: 09/09/2021] [Indexed: 12/26/2022] Open
Abstract
The elderly population living in nursing homes is particularly vulnerable to COVID-19 although individual susceptibility to SARS-CoV-2 infection may be related to the host microbiota. The objective of this work was to investigate the effect of Ligilactobacillus salivarius MP101 on the functional (Barthel index), cognitive (GDS/FAST), and nutritional (MNA) status as well as on the nasal and fecal inflammatory profiles of elderly residents living in a nursing home that is highly affected by COVID-19. A total of 25 residents participated in the trial, which involved the daily ingestion of a dairy product (L. salivarius MP101: 9.3 log10 CFU per unit) for 4 months. Nasal and fecal samples were analyzed for 37 immune factors at recruitment and at the end of the study. After the trial, no change in the GDS/FAST scores were found but, in contrast, the values for the Barthel index and the MNA score improved significantly. The concentrations of some immune factors changed significantly after the trial, including a decrease in the concentrations of BAFF/TNFSF13B, APRIL/TNFSF13, IL8, IL31, osteopontin, sTNF-R1, and sTNF-R2, and an increase in chitinase 3-like 1, IL19, IL35, and pentraxin 3 was also observed. In conclusion, L. salivarius MP101 seems to be a promising strain for improving or maintaining health in this highly vulnerable population.
Collapse
Affiliation(s)
- Marta Mozota
- Department of Nutrition and Food Science, Complutense University of Madrid, 28040 Madrid, Spain; (M.M.); (I.C.); (N.G.-T.); (R.A.); (C.A.)
| | - Irma Castro
- Department of Nutrition and Food Science, Complutense University of Madrid, 28040 Madrid, Spain; (M.M.); (I.C.); (N.G.-T.); (R.A.); (C.A.)
| | - Natalia Gómez-Torres
- Department of Nutrition and Food Science, Complutense University of Madrid, 28040 Madrid, Spain; (M.M.); (I.C.); (N.G.-T.); (R.A.); (C.A.)
| | - Rebeca Arroyo
- Department of Nutrition and Food Science, Complutense University of Madrid, 28040 Madrid, Spain; (M.M.); (I.C.); (N.G.-T.); (R.A.); (C.A.)
| | - Yolanda Lailla
- Villa Villera, 22142 Sieso de Huesca, Spain; (Y.L.); (M.S.)
| | - Mario Somada
- Villa Villera, 22142 Sieso de Huesca, Spain; (Y.L.); (M.S.)
| | - Claudio Alba
- Department of Nutrition and Food Science, Complutense University of Madrid, 28040 Madrid, Spain; (M.M.); (I.C.); (N.G.-T.); (R.A.); (C.A.)
| | - Juan Miguel Rodríguez
- Department of Nutrition and Food Science, Complutense University of Madrid, 28040 Madrid, Spain; (M.M.); (I.C.); (N.G.-T.); (R.A.); (C.A.)
| |
Collapse
|
37
|
Zhou D, Wang Q, Liu H. Coronavirus disease-19 and the gut-lung axis. Int J Infect Dis 2021; 113:300-307. [PMID: 34517046 PMCID: PMC8431834 DOI: 10.1016/j.ijid.2021.09.013] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Revised: 08/19/2021] [Accepted: 09/06/2021] [Indexed: 01/08/2023] Open
Abstract
Gastrointestinal and respiratory tract diseases often occur together. There are many overlapping pathologies, leading to the concept of the ‘gut–lung axis’ in which stimulation on one side triggers a response on the other side. This axis appears to be implicated in infections involving severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), which has triggered the global coronavirus disease 2019 (COVID-19) pandemic, in which respiratory symptoms of fever, cough and dyspnoea often occur together with gastrointestinal symptoms such as nausea, vomiting, abdominal pain and diarrhoea. Besides the gut–lung axis, it should be noted that the gut participates in numerous axes which may affect lung function, and consequently the severity of COVID-19, through several pathways. This article focuses on the latest evidence and the mechanisms that drive the operation of the gut–lung axis, and discusses the interaction between the gut–lung axis and its possible involvement in COVID-19 from the perspective of microbiota, microbiota metabolites, microbial dysbiosis, common mucosal immunity and angiotensin-converting enzyme II, raising hypotheses and providing methods to guide future research on this new disease and its treatments.
Collapse
Affiliation(s)
- Dan Zhou
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China; Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education
| | - Qiu Wang
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education; Department of Rehabilitation Medicine, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Hanmin Liu
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China; Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education.
| |
Collapse
|
38
|
Chen J, Hall S, Vitetta L. Altered gut microbial metabolites could mediate the effects of risk factors in Covid-19. Rev Med Virol 2021; 31:1-13. [PMID: 34546607 PMCID: PMC7995004 DOI: 10.1002/rmv.2211] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 12/08/2020] [Accepted: 12/09/2020] [Indexed: 01/08/2023]
Abstract
Coronavirus disease 2019 (Covid-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, is now pandemic. While most Covid-19 patients will experience mild symptoms, a small proportion will develop severe disease, which could be fatal. Clinically, Covid-19 patients manifest fever with dry cough, fatigue and dyspnoea, and in severe cases develop into acute respiratory distress syndrome (ARDS), sepsis and multi-organ failure. These severe patients are characterized by hyperinflammation with highly increased pro-inflammatory cytokines including IL-6, IL-17 and TNF-alpha as well as C-reactive protein, which are accompanied by decreased lymphocyte counts. Clinical evidence supports that gut microbiota dysregulation is common in Covid-19 and plays a key role in the pathogenesis of Covid-19. In this narrative review, we summarize the roles of intestinal dysbiosis in Covid-19 pathogenesis and posit that the associated mechanisms are being mediated by gut bacterial metabolites. Based on this premise, we propose possible clinical implications. Various risk factors could be causal for severe Covid-19, and these include advanced age, concomitant chronic disease, SARS-CoV-2 infection of enterocytes, use of antibiotics and psychological distress. Gut dysbiosis is associated with risk factors and severe Covid-19 due to decreased commensal microbial metabolites, which cause reduced anti-inflammatory mechanisms and chronic low-grade inflammation. The preconditioned immune dysregulation enables SARS-CoV-2 infection to progress to an uncontrolled hyperinflammatory response. Thus, a pre-existing gut microbiota that is diverse and abundant could be beneficial for the prevention of severe Covid-19, and supplementation with commensal microbial metabolites may facilitate and augment the treatment of severe Covid-19.
Collapse
Affiliation(s)
| | - Sean Hall
- Research DepartmentMedlab ClinicalSydneyAustralia
| | - Luis Vitetta
- Research DepartmentMedlab ClinicalSydneyAustralia
- Faculty of Medicine and HealthThe University of SydneySydneyAustralia
| |
Collapse
|
39
|
Abstract
BACKGROUND There is a bidirectional interaction between the intestines and lungs, the so-called lung-intestinal axis. METHOD The review article reports on studies that deal with a possible influence of the intestinal microbiota on the immune response to a SARS-CoV-2 infection. RESULTS AND CONCLUSIONS Studies have shown that COVID-19 is accompanied by dysbiosis that persists even after successful virus conversion (negative PCR). One study found that the severity of COVID-19 is associated with the intestinal microbiota. A dysbiosis could thus favor the so-called cytokine storm. There is indication that pre- and probiotics could boost the immune response in both the guts and lungs.
Collapse
Affiliation(s)
- Peter C Konturek
- Klinik für Innere Med. II/Saalfeld, Thüringen-Kliniken "Georgius Agricola" GmbH, Rainweg 68, 07318, Saalfeld, Germany.
| |
Collapse
|
40
|
Parosmia, Dysgeusia, and Tongue Features Changes in a Patient with Post-Acute COVID-19 Syndrome. Case Rep Dent 2021; 2021:3788727. [PMID: 34457365 PMCID: PMC8397571 DOI: 10.1155/2021/3788727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Accepted: 08/10/2021] [Indexed: 12/15/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19) is the pandemic of major global concern that has been causing several tragic events ever since December 2019. Infection with COVID-19 varies from being asymptomatic or mild with general unwellness to severe with difficulty in breathing and even mortality. Recently, several reports signified the persistence of symptoms or the development of new ones for weeks or months after the virus has gone, the so-called "post-acute COVID-19 syndrome." This article presents a 31-year-old female with post-acute COVID-19 syndrome showing parosmia, dysgeusia, and tongue feature changes as simultaneous newly developed manifestations after the viral clearance. Follow-up revealed complete remission of manifestations after one month. Post-acute COVID-19 syndrome can show oral manifestations. Despite the lack of evidence regarding the etiology, risk factors, and consequences of post-COVID-19 syndrome, it is important to monitor COVID-19 survivors to avoid complications during their recovery period.
Collapse
|
41
|
Tong T, Niu X, Li Q, Ling Y, Li Z, Liu J, Zhang M, Bai Z, Xia R, Wu Z, Liu X. The Effect of Lactobacillus plantarum BW2013 on The Gut Microbiota in Mice Analyzed by 16S rRNA Amplicon Sequencing. Pol J Microbiol 2021; 70:235-243. [PMID: 34349813 PMCID: PMC8326986 DOI: 10.33073/pjm-2021-022] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Revised: 05/16/2021] [Accepted: 05/17/2021] [Indexed: 12/17/2022] Open
Abstract
Lactobacillus plantarum BW2013 was isolated from the fermented Chinese cabbage. This study aimed to test the effect of this strain on the gut microbiota in BALB/c mice by 16S rRNA amplicon sequencing. The mice were randomly allocated to the control group and three treatment groups of L. plantarum BW2013 (a low-dose group of 108 CFU/ml, a medium-dose group of 109 CFU/ml, and a high-dose group of 1010 CFU/ml). The weight of mice was recorded once a week, and the fecal samples were collected for 16S rRNA amplicon sequencing after 28 days of continuous treatment. Compared with the control group, the body weight gain in the treatment groups was not significant. The 16S rRNA amplicon sequencing analysis showed that both the Chao1 and ACE indexes increased slightly in the medium-dose group compared to the control group, but the difference was not significant. Based on PCoA results, there was no significant difference in β diversity between the treatment groups. Compared to the control group, the abundance of Bacteroidetes increased in the low-dose group. The abundance of Firmicutes increased in the medium-dose group. At the genus level, the abundance of Alloprevotella increased in the low-dose group compared to the control group. The increased abundance of Ruminococcaceae and decreased abundance of Candidatus_Saccharimonas was observed in the medium-dose group. Additionally, the abundance of Bacteroides increased, and Alistipes and Candidatus_Saccharimonas decreased in the high-dose group. These results indicated that L. plantarum BW2013 could ameliorate gut microbiota composition, but its effects vary with the dose.
Collapse
Affiliation(s)
- Tong Tong
- Beijing Key Laboratory of Bioactive Substances and Functional Foods and Department of Food Science, College of Biochemical Engineering, Beijing Union University, Beijing, China
| | - Xiaohui Niu
- Beijing Key Laboratory of Bioactive Substances and Functional Foods and Department of Food Science, College of Biochemical Engineering, Beijing Union University, Beijing, China
| | - Qian Li
- Beijing Key Laboratory of Bioactive Substances and Functional Foods and Department of Food Science, College of Biochemical Engineering, Beijing Union University, Beijing, China
| | - Yuxi Ling
- Beijing Key Laboratory of Bioactive Substances and Functional Foods and Department of Food Science, College of Biochemical Engineering, Beijing Union University, Beijing, China
| | - Zuming Li
- Beijing Key Laboratory of Bioactive Substances and Functional Foods and Department of Food Science, College of Biochemical Engineering, Beijing Union University, Beijing, China
| | - Jia Liu
- Internal Trade Food Science and Technology (Beijing) Co., Ltd, Beijing, China
| | - Michael Zhang
- Department of Physics and Astronomy, University of Manitoba, Winnipeg, Canada
| | - Zhihui Bai
- Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, China
| | - Ran Xia
- Department of Physics and Astronomy, University of Manitoba, Winnipeg, Canada
| | - Zhichao Wu
- Department of Physics and Astronomy, University of Manitoba, Winnipeg, Canada
| | - Xiu Liu
- China National Research Institute of Food and Fermentation Industies Co., Ltd, Beijing, China
| |
Collapse
|
42
|
Gu S, Chen Y, Lv L, Li L. Reply to Marcialis et al. Clin Infect Dis 2021; 72:2246-2247. [PMID: 32645719 DOI: 10.1093/cid/ciaa966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Silan Gu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Yanfei Chen
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Longxian Lv
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Lanjuan Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
43
|
Katz-Agranov N, Zandman-Goddard G. Autoimmunity and COVID-19 - The microbiotal connection. Autoimmun Rev 2021; 20:102865. [PMID: 34118455 PMCID: PMC8189735 DOI: 10.1016/j.autrev.2021.102865] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2021] [Accepted: 03/28/2021] [Indexed: 02/07/2023]
Abstract
Background and aims The novel SARS-CoV-2 has been rattling the world since its outbreak in December 2019, leading to the COVID-19 pandemic. The learning curve of this new virus has been steep, with a global scientific community desperate to learn how the virus is transmitted, how it replicates, why it causes such a wide spectrum of disease manifestations, resulting in none or few symptoms in some. Others are burdened by an intense immune response that resembles the cytokine storm syndrome (CSS), which leads to severe disease manifestations, often complicated by fatal acute respiratory distress syndrome and death. Research efforts have been focusing on finding effective cures and vaccinations for this virus. The presence of SARS-CoV-2 in the gastrointestinal (GI) tract, represented by several GI manifestations, has led to its investigation as a target for the virus and as an indicator of disease severity. The response of the microbiome (which is heavily linked to immunity) to the novel SARS-CoV-2 virus, and its role in igniting the exaggerated immune response has therefore become a focus of interest. The objective of our study was to gather the data connecting between the microbiome, the GI tract and COVID-19 and to investigate whether these reported alterations in the gut microbiome bear any resemblance to those seen in lupus, the prototypical autoimmune disease. Confirming such changes may become the steppingstone to potential therapies that may prevent transmission, progression and immune related manifestations of COVID-19, via manipulation of the gut microbiota. Methods We performed an extensive literature review, utilizing the Pubmed search engine and Google Scholar for studies evaluating the microbiome in COVID-19 patients and compared results with studies evaluating the microbiome in lupus. We searched for the terms: microbiome, dysbiosis, COVID-19, SARS-CoV-2, gastrointestinal as well as lupus and autoimmune. While there were hundreds of articles which referred to gastrointestinal manifestations in COVID-19, to date only 4 studies investigated the gastrointestinal microbiome in this setting. We compared the similarities between microbiome of COVID-19 patients and lupus patients. Results We found that there are several similar processes of immune dysregulation in patients with COVID-19 and in those with lupus, with several other alterations seen in other pathological states. Some of these similarities include loss of microbiota biodiversity, increased representation of pathobionts, which are microbes associated with inflammation and disease (i.e Proteobacteria) and a relative decrease of symbionts, which are protective microbes, associated with anti-inflammatory properties (i.e Lactobacillus). Compromise to the intestinal barrier has also been reported in both. Conclusions We conclude that the gastrointestinal tract contributes to the disease manifestations in COVID-19. Whether gastrointestinal dysbiosis is the cause or effect of gastrointestinal manifestations and several severe systemic manifestations, which may be the response to an increased pro-inflammatory environment, is still debatable and warrants further investigation. Given the resemblance of the microbiome in COVID-19 patients to that seen in lupus patients, it becomes clearer why several therapies used in autoimmune conditions are currently under investigation for the treatment of COVID-19 patients. Moreover, these findings should promote further investigating the utility of manipulation of the microbiome, via nutritional supplementation or even fecal transplantations, interventions that may alter the course of the disease, and potentially prevent disease transmission at low cost and low risk.
Collapse
Affiliation(s)
- Nurit Katz-Agranov
- Department of Medicine, Saint Elizabeth's Medical Center, Boston, MA, USA; Tufts University School of Medicine, Boston, MA, USA.
| | - Gisele Zandman-Goddard
- Department of Medicine C, Wolfson Medical Center, Holon, Israel; Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
44
|
Grinevich VB, Kravchuk YA, Ped VI, Sas EI, Salikova SP, Gubonina IV, Tkachenko EI, Sitkin SI, Lazebnik LB, Golovanova EV, Belousova EA, Makarchuk PA, Eremina EY, Sarsenbaeva AS, Abdulganieva DI, Tarasova LV, Gromova OA, Ratnikov VA, Kozlov KV, Ratnikova AK. Management of patients with digestive diseases during the COVID-19 pandemic. Clinical Practice Guidelines by the Russian scientific medical society of internal medicine (RSMSIM) and the Gastroenterological Scientific Society of Russia (2nd edition). EXPERIMENTAL AND CLINICAL GASTROENTEROLOGY 2021:5-82. [DOI: 10.31146/1682-8658-ecg-187-3-5-82] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/26/2023]
Abstract
The presented clinical practice guidelines of the Gastroenterological Scientific Society of Russia (GSSR), diagnostic, and therapeutic approaches for patients with digestive diseases during the COVID-19 pandemic. The guidelines were approved by the XXIII Congress of the GSSR and the 22nd International Slavonic-Baltic Scientifi c Forum “St. Petersburg - Gastro-2020 ON-LINE” (St. Petersburg, June 11, 2020). The presented clinical practice guidelines of the Russian Scientific Medical Society of Internal Medicine (RSMSIM) and the Gastroenterological Scientific Society of Russia (GSSR), diagnostic, and therapeutic approaches for patients with digestive diseases during the COVID-19 pandemic. The recommendations were approved at the XV National Congress of Internal Medicine, XXIII Congress of NOGR on the basis of the 1st edition, adopted at the 22nd International Slavic- Baltic Scientific Forum “St. Petersburg - Gastro-2020 ON-LINE”.
Collapse
Affiliation(s)
| | | | - V. I. Ped
- Military Medical Academy named after S. M. Kirov
| | - E. I. Sas
- Military Medical Academy named after S. M. Kirov
| | | | | | | | - S. I. Sitkin
- State Research Institute of Highly Pure Biopreparations of FMBA of Russia; Almazov National Medical Research Centre; North-Western state medical University named after I. I. Mechnikov, Ministry of health of the Russian Federation
| | - L. B. Lazebnik
- Moscow state University of Medicine a. Densitry named after A. I. Yevdokimov of the Ministry of Health of Russia
| | - E. V. Golovanova
- Moscow state University of Medicine a. Densitry named after A. I. Yevdokimov of the Ministry of Health of Russia
| | - E. A. Belousova
- State Budgetary Institution of Moscow Region “Moscow Regional Research Clinical Institute n.a. M. F. Vladimirsky”
| | - P. A. Makarchuk
- State Budgetary Institution of Moscow Region “Moscow Regional Research Clinical Institute n.a. M. F. Vladimirsky”
| | - E. Yu. Eremina
- Federal State Budgetary Educational Institution of Higher Education “National Research Ogarev Mordovia State University”
| | - A. S. Sarsenbaeva
- FSBEI HE SUSMU MOH Russia, st. Vorovskogo, 64, Ural Federal District
| | | | - L. V. Tarasova
- FSBEI of HE “The Chuvash State University n.a. I. N. Ulyanov”; BI of HE “The Surgut State University”
| | - O. A. Gromova
- Federal Research Center “Informatics and Management” of the Russian Academy of Sciences; Federal State Educational Institution of Higher Education Lomonosov Moscow State University
| | - V. A. Ratnikov
- Federal state budgetary institution “North-West District Scientific and Clinical Center named after L. G. Sokolov Federal Medical and Biological Agency“
| | - K. V. Kozlov
- Military Medical Academy named after S. M. Kirov
| | - A. K. Ratnikova
- Military Medical Academy named after S. M. Kirov; Federal state budgetary institution “North-West District Scientific and Clinical Center named after L. G. Sokolov Federal Medical and Biological Agency“
| |
Collapse
|
45
|
Gunduz A, Ozturk E, Cagasar O. The Impact of COVID-19 on Conjunctival Flora. Ocul Immunol Inflamm 2021; 29:709-714. [PMID: 33983868 DOI: 10.1080/09273948.2021.1927118] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/09/2022]
Abstract
Purpose: This study aims to evaluate the effect of coronavirus disease 2019 (COVID-19) on conjunctival flora in patients hospitalized with COVID-19.Methods: This prospective, controlled study was conducted between June 2020 and December 2020. The study group consisted of 45 confirmed COVID-19 patients and 43 control subjects. The collected samples were inoculated into the Thioglycollate broth media without delay. The samples with growth were then passed on eosin methylene blue agar, sabouraud dextrose agar, chocolate agar, and 5% sheep blood agar solid media.Results: The mean age of the COVID-19 patients was 64.24 ± 15.4 years, and the control subjects were 59.72 ± 11.4 years. The culture positivity of conjunctiva samples in COVID-19 patients (95.6%) was statistically significantly higher than control subjects (76.7%) (p = .024). Coagulase-negative staphylococcus and Staphylococcus aureus' positivity was significantly higher in COVID-19 patients than control subjects (p < .05).Conclusion: Patients with COVID-19 demonstrate significantly higher culture positivity on conjunctival flora than the control subjects.
Collapse
Affiliation(s)
- Ayten Gunduz
- Faculty of Medicine, Department of Medical Microbiology, Malatya Turgut Ozal University, Malatya, Turkey
| | - Emrah Ozturk
- Department of Ophthalmology, Malatya Training and Research Hospital, Malatya, Turkey
| | - Ozlem Cagasar
- Department of Infectious Diseases and Clinical Microbiology, Malatya Training and Research Hospital, Malatya, Turkey
| |
Collapse
|
46
|
Petrillo M, Brogna C, Cristoni S, Querci M, Piazza O, Van den Eede G. Increase of SARS-CoV-2 RNA load in faecal samples prompts for rethinking of SARS-CoV-2 biology and COVID-19 epidemiology. F1000Res 2021; 10:370. [PMID: 34336189 PMCID: PMC8283343 DOI: 10.12688/f1000research.52540.3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/25/2021] [Indexed: 01/08/2023] Open
Abstract
Background Scientific evidence for the involvement of human microbiota in the development of COVID-19 disease has been reported recently. SARS-CoV-2 RNA presence in human faecal samples and SARS-CoV-2 activity in faeces from COVID-19 patients have been observed. Methods Starting from these observations, an experimental design was developed to cultivate in vitro faecal microbiota from infected individuals, to monitor the presence of SARS-CoV-2, and to collect data on the relationship between faecal bacteria and the virus. Results Our results indicate that SARS-CoV-2 replicates in vitro in bacterial growth medium, that the viral replication follows bacterial growth and it is influenced by the administration of specific antibiotics. SARS-CoV-2-related peptides have been detected in 30-day bacterial cultures and characterised. Discussion Our observations are compatible with a 'bacteriophage-like' behaviour of SARS-CoV-2, which, to our knowledge has not been observed or described before. These results are unexpected and hint towards a novel hypothesis on the biology of SARS-CoV-2 and on the COVID-19 epidemiology. The discovery of possible new modes of action of SARS-CoV-2 has far-reaching implications for the prevention and the treatment of the disease.
Collapse
Affiliation(s)
- Mauro Petrillo
- European Commission, Joint Research Centre (JRC), Ispra, Italy
| | | | | | | | - Ornella Piazza
- Department of Medicine and Surgery, University of Salerno, Baronissi, Italy
| | - Guy Van den Eede
- European Commission, Joint Research Centre (JRC), Ispra, Italy
- European Commission, Joint Research Centre (JRC), Geel, Belgium
| |
Collapse
|
47
|
Petrillo M, Brogna C, Cristoni S, Querci M, Piazza O, Van den Eede G. Increase of SARS-CoV-2 RNA load in faecal samples prompts for rethinking of SARS-CoV-2 biology and COVID-19 epidemiology. F1000Res 2021; 10:370. [PMID: 34336189 PMCID: PMC8283343 DOI: 10.12688/f1000research.52540.1] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/05/2021] [Indexed: 08/11/2023] Open
Abstract
Background Scientific evidence for the involvement of human microbiota in the development of COVID-19 disease has been reported recently. SARS-CoV-2 RNA presence in human faecal samples and SARS-CoV-2 activity in faeces from COVID-19 patients have been observed. Methods Starting from these observations, an experimental design was developed to cultivate in vitro faecal microbiota from infected individuals, to monitor the presence of SARS-CoV-2, and to collect data on the relationship between faecal bacteria and the virus. Results Our results indicate that SARS-CoV-2 replicates in vitro in bacterial growth medium, that the viral replication follows bacterial growth and it is influenced by the administration of specific antibiotics. SARS-CoV-2-related peptides have been detected in 30-day bacterial cultures and characterised. Discussion Our observations are compatible with a 'bacteriophage-like' behaviour of SARS-CoV-2, which, to our knowledge has not been observed or described before. These results are unexpected and hint towards a novel hypothesis on the biology of SARS-CoV-2 and on the COVID-19 epidemiology. The discovery of possible new modes of action of SARS-CoV-2 has far-reaching implications for the prevention and the treatment of the disease.
Collapse
Affiliation(s)
- Mauro Petrillo
- European Commission, Joint Research Centre (JRC), Ispra, Italy
| | | | | | | | - Ornella Piazza
- Department of Medicine and Surgery, University of Salerno, Baronissi, Italy
| | - Guy Van den Eede
- European Commission, Joint Research Centre (JRC), Ispra, Italy
- European Commission, Joint Research Centre (JRC), Geel, Belgium
| |
Collapse
|
48
|
Petrillo M, Brogna C, Cristoni S, Querci M, Piazza O, Van den Eede G. Increase of SARS-CoV-2 RNA load in faecal samples prompts for rethinking of SARS-CoV-2 biology and COVID-19 epidemiology. F1000Res 2021; 10:370. [PMID: 34336189 PMCID: PMC8283343 DOI: 10.12688/f1000research.52540.2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/09/2021] [Indexed: 04/04/2024] Open
Abstract
Background Scientific evidence for the involvement of human microbiota in the development of COVID-19 disease has been reported recently. SARS-CoV-2 RNA presence in human faecal samples and SARS-CoV-2 activity in faeces from COVID-19 patients have been observed. Methods Starting from these observations, an experimental design was developed to cultivate in vitro faecal microbiota from infected individuals, to monitor the presence of SARS-CoV-2, and to collect data on the relationship between faecal bacteria and the virus. Results Our results indicate that SARS-CoV-2 replicates in vitro in bacterial growth medium, that the viral replication follows bacterial growth and it is influenced by the administration of specific antibiotics. SARS-CoV-2-related peptides have been detected in 30-day bacterial cultures and characterised. Discussion Our observations are compatible with a 'bacteriophage-like' behaviour of SARS-CoV-2, which, to our knowledge has not been observed or described before. These results are unexpected and hint towards a novel hypothesis on the biology of SARS-CoV-2 and on the COVID-19 epidemiology. The discovery of possible new modes of action of SARS-CoV-2 has far-reaching implications for the prevention and the treatment of the disease.
Collapse
Affiliation(s)
- Mauro Petrillo
- European Commission, Joint Research Centre (JRC), Ispra, Italy
| | | | | | | | - Ornella Piazza
- Department of Medicine and Surgery, University of Salerno, Baronissi, Italy
| | - Guy Van den Eede
- European Commission, Joint Research Centre (JRC), Ispra, Italy
- European Commission, Joint Research Centre (JRC), Geel, Belgium
| |
Collapse
|
49
|
Shi Y, Li Z, Yang C, Liu C. The role of gut-brain axis in SARA-CoV-2 neuroinvasion: Culprit or innocent bystander? Brain Behav Immun 2021; 94:476-477. [PMID: 33600935 PMCID: PMC7883713 DOI: 10.1016/j.bbi.2021.01.024] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 01/20/2021] [Indexed: 12/16/2022] Open
Affiliation(s)
| | | | - Chun Yang
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China.
| | - Cunming Liu
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China.
| |
Collapse
|
50
|
Alam MA, Parra-Saldivar R, Bilal M, Afroze CA, Ahmed MN, Iqbal HM, Xu J. Algae-Derived Bioactive Molecules for the Potential Treatment of SARS-CoV-2. Molecules 2021; 26:2134. [PMID: 33917694 PMCID: PMC8068085 DOI: 10.3390/molecules26082134] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 03/24/2021] [Accepted: 03/24/2021] [Indexed: 02/06/2023] Open
Abstract
The recently emerged COVID-19 disease caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has adversely affected the whole world. As a significant public health threat, it has spread worldwide. Scientists and global health experts are collaborating to find and execute speedy diagnostics, robust and highly effective vaccines, and therapeutic techniques to tackle COVID-19. The ocean is an immense source of biologically active molecules and/or compounds with antiviral-associated biopharmaceutical and immunostimulatory attributes. Some specific algae-derived molecules can be used to produce antibodies and vaccines to treat the COVID-19 disease. Algae have successfully synthesized several metabolites as natural defense compounds that enable them to survive under extreme environments. Several algae-derived bioactive molecules and/or compounds can be used against many diseases, including microbial and viral infections. Moreover, some algae species can also improve immunity and suppress human viral activity. Therefore, they may be recommended for use as a preventive remedy against COVID-19. Considering the above critiques and unique attributes, herein, we aimed to systematically assess algae-derived, biologically active molecules that could be used against this disease by looking at their natural sources, mechanisms of action, and prior pharmacological uses. This review also serves as a starting point for this research area to accelerate the establishment of anti-SARS-CoV-2 bioproducts.
Collapse
Affiliation(s)
- Md. Asraful Alam
- School of Chemical Engineering, Zhengzhou University, Zhengzhou 450001, China;
| | | | - Muhammad Bilal
- School of Life Science and Food Engineering, Huaiyin Institute of Technology, Huaian 223003, China;
| | - Chowdhury Alfi Afroze
- Department of Biotechnology & Genetic Engineering, University of Development Alternative, Dhaka 1209, Bangladesh;
| | - Md. Nasir Ahmed
- Biotechnology & Natural Medicine Division, TechB Nutrigenomics, Dhaka 1209, Bangladesh;
| | - Hafiz M.N. Iqbal
- Tecnologico de Monterrey, School of Engineering and Sciences, Monterrey 64849, Mexico;
| | - Jingliang Xu
- School of Chemical Engineering, Zhengzhou University, Zhengzhou 450001, China;
| |
Collapse
|