1
|
Jena R, Choudhury PK. Bifidobacteria in Fermented Dairy Foods: A Health Beneficial Outlook. Probiotics Antimicrob Proteins 2025; 17:1-22. [PMID: 37979040 DOI: 10.1007/s12602-023-10189-w] [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] [Accepted: 11/08/2023] [Indexed: 11/19/2023]
Abstract
Bifidobacteria, frequently present in the human gastrointestinal tract, play a crucial role in preserving gut health and are mostly recognized as beneficial probiotic microorganisms. They are associated with fermenting complex carbohydrates, resulting in the production of short-chain fatty acids, bioactive peptides, exopolysaccharides, and vitamins, which provide energy and contribute to gut homeostasis. In light of these findings, research in food processing technologies has harnessed probiotic bacteria such as lactobacilli and bifidobacteria for the formulation of a wide range of fermented dairy products, ensuring their maximum survival and contributing to the development of distinctive quality characteristics and therapeutic benefits. Despite the increased interest in probiotic dairy products, introducing bifidobacteria into the dairy food chain has proved to be complicated. However, survival of Bifidobacterium species is conditioned by strain of bacteria used, metabolic interactions with lactic acid bacteria (LAB), fermentation parameters, and the temperature of storage and preservation of the dairy products. Furthermore, fortification of dairy foods and whey beverages with bifidobacteria have ability to change physicochemical and rheological properties beyond economic value of dairy products. In summary, this review underscores the significance of bifidobacteria as probiotics in diverse fermented dairy foods and accentuates their positive impact on human health. By enhancing our comprehension of the beneficial repercussions associated with the consumption of bifidobacteria-rich products, we aim to encourage individuals to embrace these probiotics as a means of promoting holistic health.
Collapse
Affiliation(s)
- Rajashree Jena
- Department of Dairy Technology, School of Agricultural and Bioengineering, Centurion University of Technology and Management, Paralakhemundi, Odisha, 761211, India
| | - Prasanta Kumar Choudhury
- Department of Dairy Technology, School of Agricultural and Bioengineering, Centurion University of Technology and Management, Paralakhemundi, Odisha, 761211, India.
| |
Collapse
|
2
|
Uttarwar RG, Mekonnen SA, Van Beeck W, Wang A, Finnegan P, Roberts RF, Merenstein D, Slupsky CM, Marco ML. Effects of Bifidobacterium animalis subsp. lactis BB-12 and yogurt on mice during oral antibiotic administration. Microbiol Res 2024; 286:127794. [PMID: 38852301 DOI: 10.1016/j.micres.2024.127794] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 05/24/2024] [Accepted: 05/29/2024] [Indexed: 06/11/2024]
Abstract
Probiotics have the potential to prevent disruptions to normal gastrointestinal function caused by oral antibiotic use. In this study, we examined the capacity of Bifidobacterium animalis subspecies lactis BB-12 (BB-12) and yogurt, separately and combined, to mitigate the effects of the antibiotic amoxicillin-clavulanate (AMC) on the gut microbiota and metabolomes of C57BL/6 J mice. Male and female mice were administered either BB-12, yogurt, BB-12 in yogurt, or saline for 10 days concurrent with the inclusion of AMC in the drinking water. Male mice exposed to AMC exhibited significant reductions (p<0.05) in body weight over the course of the study compared to sham (no AMC) controls whereas no such effects were observed for female mice. AMC administration resulted in rapid alterations to the intestinal microbiota in both sexes irrespective of BB-12 or yogurt treatment, including significant (p<0.05) losses in bacterial cell numbers and changes in microbial alpha-diversity and beta-diversity in the feces and cecal contents. The effects of AMC on the gut microbiota were observed within one day of administration and the bacterial contents continued to change over time, showing a succession marked by rapid reductions in Muribaculaceae and Lachnospiraceae and temporal increases in proportions of Acholeplasmataceae (day 1) and Streptococcaceae and Leuconostocaceae (day 5). By day 10 of AMC intake, high proportions of Gammaproteobacteria assigned as Erwiniaceae or Enterobacteriaceae (average of 63 %), were contained in the stools and were similarly enriched in the cecum. The cecal contents of mice given AMC harbored significantly reduced concentrations of (branched) short-chain fatty acids (SCFA), aspartate, and other compounds, whereas numerous metabolites, including formate, lactate, and several amino acids and amino acid derivatives were significantly enriched. Despite the extensive impact of AMC, starting at day 7 of the study, the body weights of male mice given yogurt or BB-12 (in saline) with AMC were similar to the healthy controls. BB-12 (in saline) and yogurt intake was associated with increased Streptococcaceae and both yogurt and BB-12 resulted in lower proportions of Erwiniaceae in the fecal and cecal contents. The cecal contents of mice fed BB-12 in yogurt contained levels of formate, glycine, and glutamine that were equivalent to the sham controls. These findings highlight the potential of BB-12 and yogurt to mitigate antibiotic-induced gut dysbiosis.
Collapse
Affiliation(s)
- Ruchita G Uttarwar
- Department of Food Science & Technology, University of California, Davis, USA
| | - Solomon A Mekonnen
- Department of Food Science & Technology, University of California, Davis, USA
| | - Wannes Van Beeck
- Department of Food Science & Technology, University of California, Davis, USA
| | - Aidong Wang
- Department of Food Science & Technology, University of California, Davis, USA; Department of Nutrition, University of California, Davis, CA, USA
| | - Peter Finnegan
- Department of Food Science & Technology, University of California, Davis, USA
| | | | - Daniel Merenstein
- Department of Family Medicine, Georgetown University Medical Center, Washington, DC, USA
| | - Carolyn M Slupsky
- Department of Food Science & Technology, University of California, Davis, USA; Department of Nutrition, University of California, Davis, CA, USA
| | - Maria L Marco
- Department of Food Science & Technology, University of California, Davis, USA.
| |
Collapse
|
3
|
Bermúdez-Humarán LG, Chassaing B, Langella P. Exploring the interaction and impact of probiotic and commensal bacteria on vitamins, minerals and short chain fatty acids metabolism. Microb Cell Fact 2024; 23:172. [PMID: 38867272 PMCID: PMC11167913 DOI: 10.1186/s12934-024-02449-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Accepted: 06/04/2024] [Indexed: 06/14/2024] Open
Abstract
There is increasing evidence that probiotic and commensal bacteria play a role in substrate metabolism, energy harvesting and intestinal homeostasis, and may exert immunomodulatory activities on human health. In addition, recent research suggests that these microorganisms interact with vitamins and minerals, promoting intestinal and metabolic well-being while producing vital microbial metabolites such as short-chain fatty acids (SCFAs). In this regard, there is a flourishing field exploring the intricate dynamics between vitamins, minerals, SCFAs, and commensal/probiotic interactions. In this review, we summarize some of the major hypotheses beyond the mechanisms by which commensals/probiotics impact gut health and their additional effects on the absorption and metabolism of vitamins, minerals, and SCFAs. Our analysis includes comprehensive review of existing evidence from preclinical and clinical studies, with particular focus on the potential interaction between commensals/probiotics and micronutrients. Finally, we highlight knowledge gaps and outline directions for future research in this evolving field.
Collapse
Affiliation(s)
- Luis G Bermúdez-Humarán
- Laboratory of Commensals and Probiotics-Host Interactions, Université Paris-Saclay, INRAE, Micalis Institute, Jouy-en-Josas, AgroParisTech, 78350, France.
| | - Benoit Chassaing
- Microbiome-Host Interactions, Institut Pasteur, Université Paris Cité, INSERM U1306, Paris, France
- INSERM U1016, team Mucosal microbiota in chronic inflammatory diseases, CNRS UMR 8104, Université de Paris, Paris, France
| | - Philippe Langella
- Laboratory of Commensals and Probiotics-Host Interactions, Université Paris-Saclay, INRAE, Micalis Institute, Jouy-en-Josas, AgroParisTech, 78350, France.
| |
Collapse
|
4
|
Itoh K, Matsueda S. Exploring the Potential of Humoral Immune Response to Commensal Bifidobacterium as a Biomarker for Human Health, including Both Malignant and Non-Malignant Diseases: A Perspective on Detection Strategies and Future Directions. Biomedicines 2024; 12:803. [PMID: 38672158 PMCID: PMC11048515 DOI: 10.3390/biomedicines12040803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 03/24/2024] [Accepted: 03/26/2024] [Indexed: 04/28/2024] Open
Abstract
In this comprehensive review, we explore the pivotal role of commensal Bifidobacterium (c-BIF) as potent non-self-antigens through antigenic mimicry, along with exploring the potential of humoral immune responses for both malignant and non-malignant disease. c-BIF, a predominant component of the human gut microbiome encompassing around 90% of the human genome, has emerged as a pivotal player in human biology. Over recent decades, there has been extensive research elucidating the intricate connections between c-BIF and various facets of human health, with particular emphasis on their groundbreaking impact on anti-cancer effects and the management of non-malignant diseases. The multifaceted role of c-BIF is explored, ranging from enhancing anti-tumor immunity to improving the efficacy of anti-cancer and anti-infectious disease strategies, and serving as predictive biomarkers for various diseases. Recent studies highlight not only c-BIF's promotion of anti-tumor immunity but also their role in enhancing the efficacy of immune checkpoint inhibitors. The review emphasizes the promising avenue of manipulating the gut microbiota, particularly c-BIF, for modulating cancer immunotherapy with targeted effects on tumor cells while minimizing harm to normal tissue. In the context of infectious and inflammatory diseases, the crucial role of c-BIFs in the management of COVID-19 symptoms is examined, emphasizing their impact on the severity of and immune response to COVID-19. Furthermore, c-BIF exhibits preventive and therapeutic effects on Human Papillomaviruses (HPV) and shows promise in improving inflammatory bowel diseases. The potential application of c-BIF as a biomarker for immunotherapy is explored, with a specific emphasis on its predictive and prognostic value in cancer. Suggestions are made regarding the use of humoral immune responses to cytotoxic T lymphocyte (CTL) epitope peptides that share motifs with c-BIF, proposing them as potential markers for predicting overall survival in diverse cancer patients. In conclusion, c-BIF emerges as a crucial and multifaceted determinant of human health, across anti-tumor immunity to infectious and inflammatory disease management. The manipulation of c-BIF and gut microbiota presents a promising avenue for advancing therapeutic strategies, particularly in the realm of cancer immunotherapy. Additionally, this review highlights the significance of c-BIF as potent non-self-antigens via antigenic mimicry, emphasizing the importance of robust humoral immune responses against c-BIF for preventing various diseases, including inflammatory conditions. Elevated levels of circulating antibodies against c-BIF in healthy individuals may serve as potential indicators of lower risks for malignant and non-malignant diseases.
Collapse
Affiliation(s)
| | - Satoko Matsueda
- Cancer Vaccine Center, Kurume University, Kurume 839-0863, Japan
| |
Collapse
|
5
|
Vogel K, Arra A, Lingel H, Bretschneider D, Prätsch F, Schanze D, Zenker M, Balk S, Bruder D, Geffers R, Hachenberg T, Arens C, Brunner-Weinzierl MC. Bifidobacteria shape antimicrobial T-helper cell responses during infancy and adulthood. Nat Commun 2023; 14:5943. [PMID: 37741816 PMCID: PMC10517955 DOI: 10.1038/s41467-023-41630-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Accepted: 09/11/2023] [Indexed: 09/25/2023] Open
Abstract
Microbial infections early in life are challenging for the unexperienced immune system. The SARS-CoV-2 pandemic again has highlighted that neonatal, infant, child, and adult T-helper(Th)-cells respond differently to infections, and requires further understanding. This study investigates anti-bacterial T-cell responses against Staphylococcus aureus aureus, Staphylococcus epidermidis and Bifidobacterium longum infantis in early stages of life and adults and shows age and pathogen-dependent mechanisms. Beside activation-induced clustering, T-cells stimulated with Staphylococci become Th1-type cells; however, this differentiation is mitigated in Bifidobacterium-stimulated T-cells. Strikingly, prestimulation of T-cells with Bifidobacterium suppresses the activation of Staphylococcus-specific T-helper cells in a cell-cell dependent manner by inducing FoxP3+CD4+ T-cells, increasing IL-10 and galectin-1 secretion and showing a CTLA-4-dependent inhibitory capacity. Furthermore Bifidobacterium dampens Th responses of severely ill COVID-19 patients likely contributing to resolution of harmful overreactions of the immune system. Targeted, age-specific interventions may enhance infection defence, and specific immune features may have potential cross-age utilization.
Collapse
Affiliation(s)
- Katrin Vogel
- Department of Experimental Paediatrics, University Hospital, Otto-von-Guericke University, Magdeburg, Germany
| | - Aditya Arra
- Department of Experimental Paediatrics, University Hospital, Otto-von-Guericke University, Magdeburg, Germany
| | - Holger Lingel
- Department of Experimental Paediatrics, University Hospital, Otto-von-Guericke University, Magdeburg, Germany
| | | | - Florian Prätsch
- Department of Anaesthesiology and Intensive Care Medicine, University Hospital, Otto-von-Guericke-University, Magdeburg, Germany
| | - Denny Schanze
- Institute of Human Genetics, University Hospital, Otto-von-Guericke University, Magdeburg, Germany
| | - Martin Zenker
- Institute of Human Genetics, University Hospital, Otto-von-Guericke University, Magdeburg, Germany
| | - Silke Balk
- Department of Experimental Paediatrics, University Hospital, Otto-von-Guericke University, Magdeburg, Germany
| | - Dunja Bruder
- Infection Immunology Group, Institute of Medical Microbiology and Hospital Hygiene, Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke University, Magdeburg, Germany
- Immune Regulation Group, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Robert Geffers
- Genome Analytics, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Thomas Hachenberg
- Department of Anaesthesiology and Intensive Care Medicine, University Hospital, Otto-von-Guericke-University, Magdeburg, Germany
| | - Christoph Arens
- Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital, Otto-von-Guericke University, Magdeburg, Germany
- Justus-Liebig-University Gießen, University Hospital of Gießen and Marburg (UKGM), Gießen Campus, Department of Otorhinolaryngology, Head/Neck Surgery and Plastic Surgery, Gießen, Germany
| | - Monika C Brunner-Weinzierl
- Department of Experimental Paediatrics, University Hospital, Otto-von-Guericke University, Magdeburg, Germany.
| |
Collapse
|
6
|
Ivashkin V, Fomin V, Moiseev S, Brovko M, Maslennikov R, Ulyanin A, Sholomova V, Vasilyeva M, Trush E, Shifrin O, Poluektova E. Efficacy of a Probiotic Consisting of Lacticaseibacillus rhamnosus PDV 1705, Bifidobacterium bifidum PDV 0903, Bifidobacterium longum subsp. infantis PDV 1911, and Bifidobacterium longum subsp. longum PDV 2301 in the Treatment of Hospitalized Patients with COVID-19: a Randomized Controlled Trial. Probiotics Antimicrob Proteins 2023; 15:460-468. [PMID: 34643888 PMCID: PMC8512595 DOI: 10.1007/s12602-021-09858-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/05/2021] [Indexed: 02/07/2023]
Abstract
The treatment of coronavirus disease (COVID-19) and COVID-19-associated diarrhea remains challenging. This study aimed to evaluate the efficacy of a multi-strain probiotic in the treatment of COVID-19. This was a randomized, controlled, single-center, open-label trial (NCT04854941). Inpatients with confirmed COVID-19 and pneumonia were randomly assigned to a group that received a multi-strain probiotic (PRO group) or to the control group (CON group). There were 99 and 101 patients in the PRO and CON groups, respectively. No significant differences in mortality, total duration of disease and hospital stay, incidence of intensive care unit admission, need for mechanical ventilation or oxygen support, liver injury development, and changes in inflammatory biomarker levels were observed between the PRO and CON groups among all included patients as well as among subgroups delineated based on age younger or older than 65 years, and subgroups with chronic cardiovascular diseases and diabetes. Diarrhea on admission was observed in 11.5% of patients; it resolved earlier in the PRO group than in the CON group (2 [1-4] vs. 4 [3-6] days; p = 0.049). Hospital-acquired diarrhea developed less frequently in the PRO group than in the CON group among patients who received a single antibiotic (0% vs. 12.5%; p = 0.023) unlike among those who received > 1 antibiotic (10.5% vs. 13.3%; p = 0.696). The studied probiotic had no significant effect on mortality and changes in most biomarkers in COVID-19. However, it was effective in treating diarrhea associated with COVID-19 and in preventing hospital-acquired diarrhea in patients who received a single antibiotic.
Collapse
Affiliation(s)
- Vladimir Ivashkin
- Department of Internal Medicine, Gastroenterology and Hepatology, Sechenov University, Moscow, Russian Federation
- Scientific Community for Human Microbiome Research, Moscow, Russian Federation
| | - Victor Fomin
- Department of Internal Medicine, Gastroenterology and Hepatology, Sechenov University, Moscow, Russian Federation
| | - Sergey Moiseev
- Department of Internal Medicine, Gastroenterology and Hepatology, Sechenov University, Moscow, Russian Federation
| | - Michail Brovko
- Department of Internal Medicine, Gastroenterology and Hepatology, Sechenov University, Moscow, Russian Federation
| | - Roman Maslennikov
- Department of Internal Medicine, Gastroenterology and Hepatology, Sechenov University, Moscow, Russian Federation.
- Scientific Community for Human Microbiome Research, Moscow, Russian Federation.
| | - Anatoly Ulyanin
- Department of Internal Medicine, Gastroenterology and Hepatology, Sechenov University, Moscow, Russian Federation
- Scientific Community for Human Microbiome Research, Moscow, Russian Federation
| | - Victoria Sholomova
- Department of Internal Medicine, Gastroenterology and Hepatology, Sechenov University, Moscow, Russian Federation
| | - Maria Vasilyeva
- Department of Internal Medicine, Gastroenterology and Hepatology, Sechenov University, Moscow, Russian Federation
| | - Elizaveta Trush
- Department of Internal Medicine, Gastroenterology and Hepatology, Sechenov University, Moscow, Russian Federation
| | - Oleg Shifrin
- Department of Internal Medicine, Gastroenterology and Hepatology, Sechenov University, Moscow, Russian Federation
| | - Elena Poluektova
- Department of Internal Medicine, Gastroenterology and Hepatology, Sechenov University, Moscow, Russian Federation
- Scientific Community for Human Microbiome Research, Moscow, Russian Federation
| |
Collapse
|
7
|
Hwang IC, Valeriano VD, Song JH, Pereira M, Oh JK, Han K, Engstrand L, Kang DK. Mucosal immunization with lactiplantibacillus plantarum-displaying recombinant SARS-CoV-2 epitopes on the surface induces humoral and mucosal immune responses in mice. Microb Cell Fact 2023; 22:96. [PMID: 37161468 PMCID: PMC10169176 DOI: 10.1186/s12934-023-02100-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 04/17/2023] [Indexed: 05/11/2023] Open
Abstract
BACKGROUND The use of probiotic lactic acid bacteria as a mucosal vaccine vector is considered a promising alternative compared to the use of other microorganisms because of its "Generally Regarded as Safe" status, its potential adjuvant properties, and its tolerogenicity to the host. Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), which causes coronavirus disease (COVID-19), is highly transmissible and pathogenic. This study aimed to determine the potential of Lactiplantibacillus plantarum expressing SARS-CoV-2 epitopes as a mucosal vaccine against SARS-CoV-2. RESULTS In this study, the possible antigenic determinants of the spike (S1-1, S1-2, S1-3, and S1-4), membrane (ME1 and ME2), and envelope (E) proteins of SARS-CoV-2 were predicted, and recombinant L. plantarum strains surface-displaying these epitopes were constructed. Subsequently, the immune responses induced by these recombinant strains were compared in vitro and in vivo. Most surface-displayed epitopes induced pro-inflammatory cytokines [tumor necrosis factor alpha (TNF-α and interleukin (IL)-6] and anti-inflammatory cytokines (IL-10) in lipopolysaccharide-induced RAW 264.7, with the highest anti-inflammatory to pro-inflammatory cytokine ratio in the S1-1 and S1-2 groups, followed by that in the S1-3 group. When orally administered of recombinant L. plantarum expressing SARS-CoV-2 epitopes in mice, all epitopes most increased the expression of IL-4, along with induced levels of TNF-α, interferon-gamma, and IL-10, specifically in spike protein groups. Thus, the surface expression of epitopes from the spike S1 protein in L. plantarum showed potential immunoregulatory effects, suggesting its ability to potentially circumvent hyperinflammatory states relevant to monocyte/macrophage cell activation. At 35 days post immunization (dpi), serum IgG levels showed a marked increase in the S1-1, S1-2, and S1-3 groups. Fecal IgA levels increased significantly from 21 dpi in all the antigen groups, but the boosting effect after 35 dpi was explicitly observed in the S1-1, S1-2, and S1-3 groups. Thus, the oral administration of SARS-CoV-2 antigens into mice induced significant humoral and mucosal immune responses. CONCLUSION This study suggests that L. plantarum is a potential vector that can effectively deliver SARS-CoV-2 epitopes to intestinal mucosal sites and could serve as a novel approach for SARS-CoV-2 mucosal vaccine development.
Collapse
Affiliation(s)
- In-Chan Hwang
- Department of Animal Resources Science, Dankook University, Cheonan, 31116, Republic of Korea
| | - Valerie Diane Valeriano
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, 17165, Sweden
| | - Ji Hoon Song
- Department of Animal Resources Science, Dankook University, Cheonan, 31116, Republic of Korea
| | - Marcela Pereira
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, 17165, Sweden
| | - Ju Kyoung Oh
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, 17165, Sweden
| | - Kyudong Han
- Department of Microbiology, College of Science and Technology, Dankook University, Cheonan, 31116, Republic of Korea
| | - Lars Engstrand
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, 17165, Sweden
| | - Dae-Kyung Kang
- Department of Animal Resources Science, Dankook University, Cheonan, 31116, Republic of Korea.
| |
Collapse
|
8
|
Fatima S, Altwaijry H, Abulmeaty MMA, Abudawood M, Siddiqi NJ, Alrashoudi RH, Alsobaie S. Combined Supplementation of Clostridium butyricum and Bifidobacterium infantis Diminishes Chronic Unpredictable Mild Stress-Induced Intestinal Alterations via Activation of Nrf-2 Signaling Pathway in Rats. Int J Mol Sci 2023; 24:ijms24098264. [PMID: 37175970 PMCID: PMC10178881 DOI: 10.3390/ijms24098264] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Revised: 04/05/2023] [Accepted: 04/06/2023] [Indexed: 05/15/2023] Open
Abstract
Exposure to long-term chronic unpredictable mild stress (CUMS) can cause redox imbalance and inflammation, which may affect the integrity of the gut barrier. The present study was conducted to investigate the effects of a probiotics bacterium mixture, including Clostridium butyricum (C. butyricum) and Bifidobacterium infantis (B. infantis), on the intestinal homeostasis in rats exposed to multiple low-intensity stressors for 28 days. The mechanism of CUMS-induced altered intestinal homeostasis was evaluated by focusing on the nuclear factor-E2-related factor-2 (Nrf-2) pathway. In contrast to the CUMS group, probiotic mixture supplementation significantly (p < 0.01) reversed the stress-induced elevated corticosterone level, protein and lipid oxidation, and increased enzymatic and non-enzymatic antioxidant levels, as well as upregulated Nrf-2/HO-1 pathway. Probiotics supplementation further significantly (p < 0.01) decreased the CUMS-induced inflammation, altered T-lymphocyte levels, and suppressed the protein expression of nuclear factor kappa B (NF-κB) in rat intestines. Improvement in histological changes and intestinal barrier integrity further validate the beneficial effects of probiotic mixtures on CUMS-induced altered intestinal morphology. In conclusion, our results suggest that the combination of C. butyricum and B. infantis significantly attenuated CUMS-induced oxidative stress, inflammation, and T-lymphocyte modulation by upregulating Nrf-2/HO-1 signaling and inhibiting NF-κB expression in rat intestine.
Collapse
Affiliation(s)
- Sabiha Fatima
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Saud University, Riyadh 11433, Saudi Arabia
| | - Haifa Altwaijry
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Saud University, Riyadh 11433, Saudi Arabia
| | - Mahmoud M A Abulmeaty
- Department of Community Health Sciences, College of Applied Medical Sciences, King Saud University, Riyadh 11362, Saudi Arabia
| | - Manal Abudawood
- Chair of Medical and Molecular Genetics Research, Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Saud University, Riyadh 11433, Saudi Arabia
| | - Nikhat J Siddiqi
- Department of Biochemistry, College of Science, King Saud University, Riyadh 11495, Saudi Arabia
| | - Reem Hamoud Alrashoudi
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Saud University, Riyadh 11433, Saudi Arabia
| | - Sarah Alsobaie
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Saud University, Riyadh 11433, Saudi Arabia
| |
Collapse
|
9
|
Ray M, Manjunath A, Halami PM. Effect of probiotics as an immune modulator for the management of COVID-19. Arch Microbiol 2023; 205:182. [PMID: 37031431 PMCID: PMC10098245 DOI: 10.1007/s00203-023-03504-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 03/17/2023] [Accepted: 03/22/2023] [Indexed: 04/10/2023]
Abstract
COVID-19, an acute respiratory viral infection conveyed by pneumonia caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has affected millions of individuals globally, and is a public health emergency of international concern. Till now, there are no highly effective therapies for this infection without vaccination. As they can evolve quickly and cross the strain level easily, these viruses are causing epidemics or pandemics that are allied with more severe clinical diseases. A new approach is needed to improve immunity to confirm the protection against emerging viral infections. Probiotics can modify gut microbial dysbiosis, improve the host immune system, and stimulate immune signaling, increasing systemic immunity. Several probiotic bacterial therapies have been proven to decrease the period of bacterial or viral infections. Superinduction of inflammation, termed cytokine storm, has been directly linked with pneumonia and severe complications of viral respiratory infections. In this case, probiotics as potential immunomodulatory agents can be an appropriate candidate to improve the host's response to respiratory viral infections. During this COVID-19 pandemic, any approach that can induce mucosal and systemic immunity could be helpful. Here, we summarize contexts regarding the effectiveness of various probiotics for preventing virus-induced respiratory infectious diseases, especially those that could be employed for COVID-19 patients. In addition, the effects of probiotics, their mechanisms on different aspects of immune responses against respiratory viral infection, and their antiviral properties in clinical findings have been described in detail.
Collapse
Affiliation(s)
- Mousumi Ray
- Department of Microbiology and Fermentation Technology, CSIR-Central Food Technological Research Institute, Mysuru, 570020, India
| | - Ashwini Manjunath
- Department of Developmental Biology and Genetics, Indian Institute of Science, Bangalore, 560012, India
| | - Prakash M Halami
- Department of Microbiology and Fermentation Technology, CSIR-Central Food Technological Research Institute, Mysuru, 570020, India.
| |
Collapse
|
10
|
Gualtieri P, Trombetta D, Smeriglio A, Frank G, Alibrandi A, Leggeri G, Marchetti M, Zingale I, Fanelli S, Stocchi A, Di Renzo L. Effectiveness of Nutritional Supplements for Attenuating the Side Effects of SARS-CoV-2 Vaccines. Nutrients 2023; 15:nu15081807. [PMID: 37111026 PMCID: PMC10141698 DOI: 10.3390/nu15081807] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 04/04/2023] [Accepted: 04/05/2023] [Indexed: 04/29/2023] Open
Abstract
Supplementation is known to enhance the immune response and reduce infection. Therefore, the association between immune nutrients and vaccine side effects needs to be investigated. Our aim was to analyze the relationship between vaccination side effects and supplement intake among the Italian population. The study included a questionnaire asking for personal data, anthropometric information, COVID-19 infection and immunity response, and COVID-19 vaccination and supplementation. The survey was conducted from 8 February to 15 June 2022. In the study, 776 respondents were included, aged between 18 and 86 (71.3% females). We observed a statistically significant correlation between supplement consumption and side effects at the end of the vaccination cycle (p = 0.000), which was also confirmed by logistic regression (p = 0.02). Significant associations were observed between supplement intake and side effects of diarrhea and nausea at the end of the vaccination cycle (p = 0.001; p = 0.04, respectively). Significant associations were observed between side effects and omega-3 and mineral supplementation at the start of the vaccination cycle (p = 0.02; p = 0.001, respectively), and between side effects and vitamin supplementation at the end of the vaccination cycle (p = 0.005). In conclusion, our study shows a positive impact of supplementation on vaccination response, increasing host immune defenses, and reducing side effects.
Collapse
Affiliation(s)
- Paola Gualtieri
- Section of Clinical Nutrition and Nutrigenomic, Department of Biomedicine and Prevention, University of Tor Vergata, Via Montpellier 1, 00133 Rome, Italy
| | - Domenico Trombetta
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno d'Alcontres 31, 98166 Messina, Italy
| | - Antonella Smeriglio
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno d'Alcontres 31, 98166 Messina, Italy
| | - Giulia Frank
- Ph.D. School of Applied Medical-Surgical Sciences, University of Tor Vergata, Via Montpellier 1, 00133 Rome, Italy
| | - Angela Alibrandi
- Department of Economy, University of Messina, Via dei Verdi 75, 98122 Messina, Italy
| | - Giulia Leggeri
- Section of Clinical Nutrition and Nutrigenomic, Department of Biomedicine and Prevention, University of Tor Vergata, Via Montpellier 1, 00133 Rome, Italy
| | - Marco Marchetti
- School of Specialization in Food Science, University of Tor Vergata, Via Montpellier 1, 00133 Rome, Italy
| | - Ilaria Zingale
- School of Specialization in Food Science, University of Tor Vergata, Via Montpellier 1, 00133 Rome, Italy
| | - Silvia Fanelli
- School of Specialization in Food Science, University of Tor Vergata, Via Montpellier 1, 00133 Rome, Italy
| | - Arianna Stocchi
- School of Specialization in Food Science, University of Tor Vergata, Via Montpellier 1, 00133 Rome, Italy
| | - Laura Di Renzo
- Section of Clinical Nutrition and Nutrigenomic, Department of Biomedicine and Prevention, University of Tor Vergata, Via Montpellier 1, 00133 Rome, Italy
| |
Collapse
|
11
|
Chioma Mgbodile F, Nwagu TNT. Probiotic therapy, African fermented foods and food-derived bioactive peptides in the management of SARS-CoV-2 cases and other viral infections. BIOTECHNOLOGY REPORTS 2023; 38:e00795. [PMID: 37041970 PMCID: PMC10066861 DOI: 10.1016/j.btre.2023.e00795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 03/20/2023] [Accepted: 03/27/2023] [Indexed: 04/04/2023]
Abstract
The current paper focuses on the impact of probiotics, African fermented foods and bioactive peptides on severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection severity and related viral infections. Using probiotics or bioactive peptides as therapeutic adjuncts appears superior to standard care alone. Probiotics play critical roles in innate and adaptive immune modulation by balancing the gut microbiota to combat viral infections, secondary bacterial infections and microbial dysbiosis. African fermented foods contain abundant potential probiotic microorganisms such as the lactic acid bacteria (LAB), Saccharomyces, and Bacillus. More so, fermented food-derived bioactive peptides play vital roles in preventing cardiovascular diseases, hypertension, lung injury, diabetes, and other COVID-19 comorbidities. Regularly incorporating potential probiotics and bioactive peptides into diets should enable a build-up of the benefits in the body system that may result in a better prognosis, especially in COVID-19 patients with underlying complexities. Despite the reported therapeutic potentials of probiotics and fermented foods, numerous setbacks exist regarding their application in disease management. These shortfalls underscore an evident need for more studies to evaluate the specific potentials of probiotics and traditional fermented foods in ameliorating SARS-CoV-2 and other viral infections.
Collapse
|
12
|
The promise of probiotics in honeybee health and disease management. Arch Microbiol 2023; 205:73. [PMID: 36705763 DOI: 10.1007/s00203-023-03416-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Revised: 12/27/2022] [Accepted: 01/11/2023] [Indexed: 01/28/2023]
Abstract
Over the last decades, losses of bee populations have been observed worldwide. A panoply of biotic and abiotic factors, as well as the interplay among them, has been suggested to be responsible for bee declines, but definitive causes have not yet been identified. Among pollinators, the honeybee Apis mellifera is threatened by various diseases and environmental stresses, which have been shown to impact the insect gut microbiota that is known to be fundamental for host metabolism, development and immunity. Aimed at preserving the gut homeostasis, many researches are currently focusing on improving the honeybee health through the administration of probiotics e.g., by boosting the innate immune response against microbial infections. Here, we review the knowledge available on the characterization of the microbial diversity associated to honeybees and the use of probiotic symbionts as a promising approach to maintain honeybee fitness, sustaining a healthy gut microbiota and enhancing its crucial relationship with the host immune system.
Collapse
|
13
|
Mishra M, Singh SK, Kaushalendra, Kumar A. Natural prebiotics and probiotics in use as an alternative to antiviral drugs against the pandemic COVID-19. MICROBIAL BIOMOLECULES 2023:489-501. [DOI: 10.1016/b978-0-323-99476-7.00003-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
14
|
Allen J, Evans CA, Datta S. Probiotics for preventing or treating COVID-19; a systematic review of research evidence and meta-analyses of efficacy for preventing death, severe disease, or disease progression. Wellcome Open Res 2022; 7:292. [PMID: 39364259 PMCID: PMC11447443 DOI: 10.12688/wellcomeopenres.18526.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/23/2022] [Indexed: 10/05/2024] Open
Abstract
Background: COVID-19 variants threaten health globally. Despite improving vaccines and treatments, there is an urgent need for alternative strategies to prevent or reduce the severity of COVID-19. Potential strategies include probiotics, which are safe, inexpensive, globally available and have been studied previously in relation to respiratory infections. Methods: We performed a systematic review and meta-analyses of experimental, trial or observational research evidence evaluating probiotics compared with control groups for preventing or treating COVID-19. We searched PubMed, ProQuest, Google Scholar and Web of Science bibliographic databases for studies published until December 6, 2021. We then performed meta-analyses for outcomes reported consistently across studies. Outcomes reported inconsistently or not amenable to meta-analysis were compared descriptively. Results: We identified six eligible studies, which were all published in 2020 and 2021: one randomized controlled trial and five retrospective cohort studies. The only randomized controlled trial reported that groups that ingested probiotics compared with control groups that did not ingest probiotics did not differ significantly with respect to death, severe disease requiring admission to an intensive care unit or disease progression (all p>0.5). The five retrospective cohort studies reported various apparently beneficial and harmful COVID-19 outcome associations with probiotic ingestion. Meta-analyses revealed no significant associations between probiotic use and death, severe disease, or disease progression caused by COVID-19. Descriptive data revealed that probiotic ingestion was associated with a trend towards worsened duration of hospital stay, improvements in measures of respiratory condition and worsened disease duration. The evidence for these contradictory associations was weak because all studies were prone to bias and none were considered to be of high quality. Conclusions: Current evidence does not suggest that probiotics affect COVID-19 severity or mortality. However, additional higher quality studies need to be conducted to definitively determine if probiotics would be a useful adjunctive treatment for COVID-19.
Collapse
Affiliation(s)
- Jawara Allen
- School of Medicine, Johns Hopkins University, Baltimore, Maryland, 21205, USA
| | - Carlton A Evans
- IFHAD: Innovation For Health and Development, Laboratory of Research and Development, Universidad Peruana Cayetano Heredia, Lima, 15102, Peru
- IFHAD: Innovation For Health and Development, Infectious Diseases and Immunity, Imperial College London and Wellcome Trust Imperial College Centre for Global Health Research, London, W12 0NN, UK
- Innovacion Por la Salud Y el Desarrollo (IPSYD), Asociación Benéfica Prisma, Lima, 15088, Peru
| | - Sumona Datta
- IFHAD: Innovation For Health and Development, Laboratory of Research and Development, Universidad Peruana Cayetano Heredia, Lima, 15102, Peru
- IFHAD: Innovation For Health and Development, Infectious Diseases and Immunity, Imperial College London and Wellcome Trust Imperial College Centre for Global Health Research, London, W12 0NN, UK
- Innovacion Por la Salud Y el Desarrollo (IPSYD), Asociación Benéfica Prisma, Lima, 15088, Peru
- Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, L3 5QA, UK
| |
Collapse
|
15
|
Veterini AS, Prakoeswa CRS, Tinduh D, Satuman S. Simple neutralization test report: Do probiotics contribute to COVID-19 therapy? Biochem Biophys Rep 2022; 32:101348. [PMID: 36120494 PMCID: PMC9468316 DOI: 10.1016/j.bbrep.2022.101348] [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: 03/12/2022] [Revised: 06/07/2022] [Accepted: 09/12/2022] [Indexed: 01/08/2023] Open
Abstract
Background There is an urgent need to identify effective therapy to treat coronavirus diseases 2019 (COVID-19). Supplement consumption is becoming popular in this pandemic era. An example of this is probiotic consumption to improve the host's immune system. Objective This study aimed to prove whether antibodies from people taking probiotics could influence lactate dehydrogenase (LDH), adenosine triphosphate (ATP) values, and cell viability in vitro in peripheral blood mononuclear cells (PBMCs) inoculated with the SARS-CoV-2 spike protein as COVID-19 cells models. Methods This was an experimental study with control and intervention groups, totally in 12 groups divided based on antibody levels, probiotic intervention, probiotic non-intervention group, SARS-CoV-2 infection group, and non-SARS-CoV-2 infection group. In vitro assays were carried out on PBMC cell cultures inoculated with S1 SARS-CoV-2 recombinant as a COVID-19 cell model. The COVID-19 cell model was given antibodies divided into three antibody level groups: sRBD levels of <3, 325.76 and 646.18. The cytotoxicity assessment examined increased levels of LDH, cytopathic activity by measuring ATP levels, and cell viability by XTT (2,3-Bis-(2-Methoxy-4-Nitro-5-Sulfophenyl)-2H-Tetrazolium-5-Carboxanilide) assay. Data were analyzed with SPSS 21 for Windows. Results This study showed a significant difference in the LDH value (p < 0.001) between each group. The difference in ATP values between groups was significant (p < 0.001). Meanwhile, the cell viability examination found that there was a tendency of decreased XTT (cell viability in %) when there was an increase of LDH and ATP. Conclusion The change of LDH values occurred most in the antibody group that did not consume probiotics. The highest cytopathic activity based on the ATP values occurred in the infected cell culture group with antibody levels of 325.76 and consuming probiotics. In addition, LDH and ATP activities provided evidence of a significant influence on cell viability. Antibody from subject who consumed probiotic perform higher values in cell viability assay (XTT). Higher antibody titer (IgG) didn't determine better performance in neutralisation test. There is a tendency to decrease XTT if the ATP and LDH values are increased.
Collapse
|
16
|
Zeinali T, Faraji N, Joukar F, Khan Mirzaei M, Kafshdar Jalali H, Shenagari M, Mansour-Ghanaei F. Gut bacteria, bacteriophages, and probiotics: Tripartite mutualism to quench the SARS-CoV2 storm. Microb Pathog 2022; 170:105704. [PMID: 35948266 PMCID: PMC9357283 DOI: 10.1016/j.micpath.2022.105704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Revised: 07/19/2022] [Accepted: 08/01/2022] [Indexed: 11/30/2022]
Abstract
Patients with SARS-CoV-2 infection, exhibit various clinical manifestations and severity including respiratory and enteric involvements. One of the main reasons for death among covid-19 patients is excessive immune responses directed toward cytokine storm with a low chance of recovery. Since the balanced gut microbiota could prepare health benefits by protecting against pathogens and regulating immune homeostasis, dysbiosis or disruption of gut microbiota could promote severe complications including autoimmune disorders; we surveyed the association between the imbalanced gut bacteria and the development of cytokine storm among COVID-19 patients, also the impact of probiotics and bacteriophages on the gut bacteria community to alleviate cytokine storm in COVID-19 patients. In present review, we will scrutinize the mechanism of immunological signaling pathways which may trigger a cytokine storm in SARS-CoV2 infections. Moreover, we are explaining in detail the possible immunological signaling pathway-directing by the gut bacterial community. Consequently, the specific manipulation of gut bacteria by using probiotics and bacteriophages for alleviation of the cytokine storm will be investigated. The tripartite mutualistic cooperation of gut bacteria, probiotics, and phages as a candidate prophylactic or therapeutic approach in SARS-CoV-2 cytokine storm episodes will be discussed at last.
Collapse
Affiliation(s)
- Tahereh Zeinali
- Gastrointestinal and Liver Diseases Research Center, Guilan University of Medical Sciences, Rasht, Iran
| | - Niloofar Faraji
- Gastrointestinal and Liver Diseases Research Center, Guilan University of Medical Sciences, Rasht, Iran
| | - Farahnaz Joukar
- Gastrointestinal and Liver Diseases Research Center, Guilan University of Medical Sciences, Rasht, Iran
| | - Mohammadali Khan Mirzaei
- Institute of Virology, Helmholtz Center Munich and Technical University of Munich, 85764, Neuherberg, Germany
| | - Hossnieh Kafshdar Jalali
- Department of Microbiology, Faculty of Science, Lahijan Branch, Islamic Azad University, Lahijan, Iran
| | - Mohammad Shenagari
- Gastrointestinal and Liver Diseases Research Center, Guilan University of Medical Sciences, Rasht, Iran; Department of Microbiology, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran.
| | - Fariborz Mansour-Ghanaei
- Gastrointestinal and Liver Diseases Research Center, Guilan University of Medical Sciences, Rasht, Iran; Caspian Digestive Diseases Research Center, Guilan University of Medical Sciences, Rasht, Iran.
| |
Collapse
|
17
|
Psychobiotics: the Influence of Gut Microbiota on the Gut-Brain Axis in Neurological Disorders. J Mol Neurosci 2022; 72:1952-1964. [PMID: 35849305 PMCID: PMC9289355 DOI: 10.1007/s12031-022-02053-3] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Accepted: 07/12/2022] [Indexed: 12/01/2022]
Abstract
Nervous system disorders are one of the common problems that affect many people around the world every year. Regarding the beneficial effects of the probiotics on the gut and the gut-brain axis, their application along with current medications has been the subject of intense interest. Psychobiotics are a probiotic strain capable to affect the gut-brain axis. The effective role of Psychobiotics in several neurological disorders is documented. Consumption of the Psychobiotics containing nutrients has positive effects on the improvement of microbiota as well as alleviation of some symptoms of central nervous system (CNS) disorders. In the present study, the effects of probiotic strains on some CNS disorders in terms of controlling the disease symptoms were reviewed. Finding suggests that Psychobiotics can efficiently alleviate the symptoms of several CNS disorders such as autism spectrum disorders, Parkinson’s disease, multiple sclerosis, insomnia, depression, diabetic neuropathy, and anorexia nervosa. It can be concluded that functional foods containing psychotropic strains can help to improve mental health.
Collapse
|
18
|
Hazan S. Microbiome-Based Hypothesis on Ivermectin's Mechanism in COVID-19: Ivermectin Feeds Bifidobacteria to Boost Immunity. Front Microbiol 2022; 13:952321. [PMID: 35898916 PMCID: PMC9309549 DOI: 10.3389/fmicb.2022.952321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 06/10/2022] [Indexed: 01/24/2023] Open
Abstract
Ivermectin is an anti-parasitic agent that has gained attention as a potential COVID-19 therapeutic. It is a compound of the type Avermectin, which is a fermented by-product of Streptomyces avermitilis. Bifidobacterium is a member of the same phylum as Streptomyces spp., suggesting it may have a symbiotic relation with Streptomyces. Decreased Bifidobacterium levels are observed in COVID-19 susceptibility states, including old age, autoimmune disorder, and obesity. We hypothesize that Ivermectin, as a by-product of Streptomyces fermentation, is capable of feeding Bifidobacterium, thereby possibly preventing against COVID-19 susceptibilities. Moreover, Bifidobacterium may be capable of boosting natural immunity, offering more direct COVID-19 protection. These data concord with our study, as well as others, that show Ivermectin protects against COVID-19.
Collapse
|
19
|
Loo WK, Hasikin K, Suhaimi A, Yee PL, Teo K, Xia K, Qian P, Jiang Y, Zhang Y, Dhanalakshmi S, Azizan MM, Lai KW. Systematic Review on COVID-19 Readmission and Risk Factors: Future of Machine Learning in COVID-19 Readmission Studies. Front Public Health 2022; 10:898254. [PMID: 35677770 PMCID: PMC9168237 DOI: 10.3389/fpubh.2022.898254] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 04/20/2022] [Indexed: 01/19/2023] Open
Abstract
In this review, current studies on hospital readmission due to infection of COVID-19 were discussed, compared, and further evaluated in order to understand the current trends and progress in mitigation of hospital readmissions due to COVID-19. Boolean expression of (“COVID-19” OR “covid19” OR “covid” OR “coronavirus” OR “Sars-CoV-2”) AND (“readmission” OR “re-admission” OR “rehospitalization” OR “rehospitalization”) were used in five databases, namely Web of Science, Medline, Science Direct, Google Scholar and Scopus. From the search, a total of 253 articles were screened down to 26 articles. In overall, most of the research focus on readmission rates than mortality rate. On the readmission rate, the lowest is 4.2% by Ramos-Martínez et al. from Spain, and the highest is 19.9% by Donnelly et al. from the United States. Most of the research (n = 13) uses an inferential statistical approach in their studies, while only one uses a machine learning approach. The data size ranges from 79 to 126,137. However, there is no specific guide to set the most suitable data size for one research, and all results cannot be compared in terms of accuracy, as all research is regional studies and do not involve data from the multi region. The logistic regression is prevalent in the research on risk factors of readmission post-COVID-19 admission, despite each of the research coming out with different outcomes. From the word cloud, age is the most dominant risk factor of readmission, followed by diabetes, high length of stay, COPD, CKD, liver disease, metastatic disease, and CAD. A few future research directions has been proposed, including the utilization of machine learning in statistical analysis, investigation on dominant risk factors, experimental design on interventions to curb dominant risk factors and increase the scale of data collection from single centered to multi centered.
Collapse
Affiliation(s)
- Wei Kit Loo
- Department of Biomedical Engineering, Faculty of Engineering, Universiti Malaya, Kuala Lumpur, Malaysia
| | - Khairunnisa Hasikin
- Department of Biomedical Engineering, Faculty of Engineering, Universiti Malaya, Kuala Lumpur, Malaysia
| | - Anwar Suhaimi
- Department of Rehabilitation Medicine, Faculty of Medicine, Universiti Malaya, Kuala Lumpur, Malaysia
| | - Por Lip Yee
- Department of Computer System and Technology, Faculty of Computer Science and Information Technology, Universiti Malaya, Kuala Lumpur, Malaysia
| | - Kareen Teo
- Department of Biomedical Engineering, Faculty of Engineering, Universiti Malaya, Kuala Lumpur, Malaysia
| | - Kaijian Xia
- Department of Biomedical Engineering, Faculty of Engineering, Universiti Malaya, Kuala Lumpur, Malaysia
| | - Pengjiang Qian
- School of Artificial Intelligence and Computer Science, Jiangnan University, Wuxi, China
| | - Yizhang Jiang
- School of Artificial Intelligence and Computer Science, Jiangnan University, Wuxi, China
| | - Yuanpeng Zhang
- Department of Medical Informatics of Medical (Nursing) School, Nantong University, Nantong, China
| | - Samiappan Dhanalakshmi
- Department of ECE, Faculty of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur, India
- Samiappan Dhanalakshmi
| | - Muhammad Mokhzaini Azizan
- Department of Electrical and Electronic Engineering, Faculty of Engineering and Built Environment, Universiti Sains Islam Malaysia, Nilai, Malaysia
- Muhammad Mokhzaini Azizan
| | - Khin Wee Lai
- Department of Biomedical Engineering, Faculty of Engineering, Universiti Malaya, Kuala Lumpur, Malaysia
- *Correspondence: Khin Wee Lai
| |
Collapse
|
20
|
Denktaş C, Yilmaz Baysoy D, Bozdoğan A, Bozkurt HS, Bozkurt K, Özdemir O, Yilmaz M. Development and characterization of sodium alginate/ bifidobacterium probiotic biohybrid material used in tissue engineering. J Appl Polym Sci 2022; 139. [DOI: 10.1002/app.52086] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 12/15/2021] [Indexed: 04/17/2025]
Abstract
Abstract“Mechanical properties” are crucial for biodegradable and/or non‐biodegradable materials used in tissue engineering applications. In this study, bio‐hybrid films were produced by using both Bifidobacterium animalis subsp. lactis BB‐12 probiotic strain and Bifidobacterium infantis in combination with sodium alginate (SA), which demonstrates biocompatibility and facilitated gelation properties. Bio‐hybrid films were characterized by using different methods. Based on the spectroscopic and mechanical analysis, it was found that mechanical strength increased in films produced by adding Bifidobacterium infantis in SA while this increase was relatively lower as compared to those containing Bifidobacterium animalis subsp. lactis BB‐12 as cross‐linking ratio increases. Besides, bacteria contained in bio‐hybrid films increased the percentage of amorphous zone of SA in SA/bacteria films, which reduced the crystallinity ratio. This indicated that crystalline chains contained in the structure of SA are degraded by bacteria.
Collapse
Affiliation(s)
- Cenk Denktaş
- Faculty of Arts and Sciences, Physics Department Yildiz Technical University Istanbul Turkey
| | | | - Altan Bozdoğan
- Faculty of Arts and Sciences, Physics Department Yildiz Technical University Istanbul Turkey
| | - Hüseyin Sancar Bozkurt
- Maltepe University Medicine Faculty Internal Medicine, Clinic of Gastroenterology Istanbul Turkey
| | - Kutsal Bozkurt
- Faculty of Arts and Sciences, Physics Department Yildiz Technical University Istanbul Turkey
| | - Orhan Özdemir
- Faculty of Arts and Sciences, Physics Department Yildiz Technical University Istanbul Turkey
| | - Mehmet Yilmaz
- Faculty of Arts and Sciences, Physics Department Yildiz Technical University Istanbul Turkey
| |
Collapse
|
21
|
Plant Polysaccharides Modulate Immune Function via the Gut Microbiome and May Have Potential in COVID-19 Therapy. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27092773. [PMID: 35566123 PMCID: PMC9101721 DOI: 10.3390/molecules27092773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 04/17/2022] [Accepted: 04/18/2022] [Indexed: 11/24/2022]
Abstract
Plant polysaccharides can increase the number and variety of beneficial bacteria in the gut and produce a variety of active substances, including short-chain fatty acids (SCFAs). Gut microbes and their specific metabolites have the effects of promoting anti-inflammatory activity, enhancing the intestinal barrier, and activating and regulating immune cells, which are beneficial for improving immunity. A strong immune system reduces inflammation caused by external viruses and other pathogens. Coronavirus disease 2019 (COVID-19) is still spreading globally, and patients with COVID-19 often have intestinal disease and weakened immune systems. This article mainly evaluates how polysaccharides in plants can improve the immune system barrier by improving the intestinal microecological balance, which may have potential in the prevention and treatment of COVID-19.
Collapse
|
22
|
Zhang YL, Chen Q, Zheng L, Zhang ZW, Chen YJ, Dai YC, Tang ZP. Jianpi Qingchang Bushen decoction improves inflammatory response and metabolic bone disorder in inflammatory bowel disease-induced bone loss. World J Gastroenterol 2022; 28:1315-1328. [PMID: 35645540 PMCID: PMC9099185 DOI: 10.3748/wjg.v28.i13.1315] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 01/17/2022] [Accepted: 02/27/2022] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Bone loss and osteoporosis are commonly described as extra-intestinal manifestations of inflammatory bowel disease (IBD). Jianpi Qingchang Bushen decoction (JQBD) is a prescription used in clinical practice. However, further studies are needed to determine whether JQBD regulates the receptor activator of nuclear factor kappa B (NF-κB) (RANK)/receptor activator of NF-κB ligand (RANKL)/ osteoprotegerin (OPG) pathways and could play a role in treating IBD-induced bone loss. AIM To evaluate the therapeutic effect of JQBD in IBD-induced bone loss and explore the underlying mechanisms. METHODS An IBD-induced bone loss model was constructed by feeding 12 6-to-8-wk-old interleukin-10 (IL-10)-knockout mice with piroxicam for 10 d. The mice were randomly divided into model and JQBD groups. We used wild-type mice as a control. The JQBD group was administered the JQBD suspension for 2 wk by gavage, while the control and model groups were given normal saline at the corresponding time points. All mice were killed after the intervention. The effect of JQBD on body weight, disease activity index (DAI), and colon length was analyzed. Histopathological examination, colon ultrastructure observation, and micro-computed tomographic scanning of the lumbar vertebrae were performed. The gene expression of NF-κB, tumor necrosis factor-α (TNF-α), IL-1β, IL-6, and IL-8 in the colon was evaluated by real-time polymerase chain reaction. Colon samples were assessed by Western blot for the expression of RANKL, OPG, RANK, and NF-κB proteins. RESULTS The model group lost body weight, had a shorter colon, and showed a dramatic increase in DAI score, whereas JQBD had protective and therapeutic effects. Treatment with JQBD significantly improved inflammatory cell infiltration and reduced crypt abscess and ulcer formation. Three-dimensional imaging of the vertebral centrum in the model group revealed a lower bone mass, loose trabeculae, and "rod-shaped" changes in the structure compared to the control group and JQBD groups. The bone volume/total volume ratio and bone mineral density were significantly lower in the model group than in the control group. JQBD intervention downregulated the NF-κB, TNF-α, IL-1β, IL-6, and IL-8 mRNA expression levels. The RANKL and OPG protein levels were also improved. CONCLUSION JQBD reduces inflammation of the colonic mucosa and inhibits activation of the RANK/ RANKL/OPG signaling pathway, thereby reducing osteoclast activation and bone resorption and improving bone metabolism.
Collapse
Affiliation(s)
- Ya-Li Zhang
- Institute of Digestive Diseases, LongHua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Qian Chen
- Institute of Digestive Diseases, LongHua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Lie Zheng
- Department of Gastroenterology, Traditional Chinese Medicine Hospital of Shaanxi Province, Xi’an 710003, Shaanxi Province, China
| | - Zi-Wei Zhang
- Institute of Digestive Diseases, LongHua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Yu-Jun Chen
- Institute of Digestive Diseases, LongHua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Yan-Cheng Dai
- Department of Gastroenterology, Shanghai Traditional Chinese Medicine-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200082, China
| | - Zhi-Peng Tang
- Institute of Digestive Diseases, LongHua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| |
Collapse
|
23
|
Hazan S, Stollman N, Bozkurt HS, Dave S, Papoutsis AJ, Daniels J, Barrows BD, Quigley EM, Borody TJ. Lost microbes of COVID-19: Bifidobacterium, Faecalibacterium depletion and decreased microbiome diversity associated with SARS-CoV-2 infection severity. BMJ Open Gastroenterol 2022; 9:e000871. [PMID: 35483736 PMCID: PMC9051551 DOI: 10.1136/bmjgast-2022-000871] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Accepted: 03/28/2022] [Indexed: 12/11/2022] Open
Abstract
OBJECTIVE The study objective was to compare gut microbiome diversity and composition in SARS-CoV-2 PCR-positive patients whose symptoms ranged from asymptomatic to severe versus PCR-negative exposed controls. DESIGN Using a cross-sectional design, we performed shotgun next-generation sequencing on stool samples to evaluate gut microbiome composition and diversity in both patients with SARS-CoV-2 PCR-confirmed infections, which had presented to Ventura Clinical Trials for care from March 2020 through October 2021 and SARS-CoV-2 PCR-negative exposed controls. Patients were classified as being asymptomatic or having mild, moderate or severe symptoms based on National Institute of Health criteria. Exposed controls were individuals with prolonged or repeated close contact with patients with SARS-CoV-2 infection or their samples, for example, household members of patients or frontline healthcare workers. Microbiome diversity and composition were compared between patients and exposed controls at all taxonomic levels. RESULTS Compared with controls (n=20), severely symptomatic SARS-CoV-2-infected patients (n=28) had significantly less bacterial diversity (Shannon Index, p=0.0499; Simpson Index, p=0.0581), and positive patients overall had lower relative abundances of Bifidobacterium (p<0.0001), Faecalibacterium (p=0.0077) and Roseburium (p=0.0327), while having increased Bacteroides (p=0.0075). Interestingly, there was an inverse association between disease severity and abundance of the same bacteria. CONCLUSION We hypothesise that low bacterial diversity and depletion of Bifidobacterium genera either before or after infection led to reduced proimmune function, thereby allowing SARS-CoV-2 infection to become symptomatic. This particular dysbiosis pattern may be a susceptibility marker for symptomatic severity from SARS-CoV-2 infection and may be amenable to preinfection, intrainfection or postinfection intervention. TRIAL REGISTRATION NUMBER NCT04031469 (PCR-) and 04359836 (PCR+).
Collapse
Affiliation(s)
| | - Neil Stollman
- Division of Gastroenterology, Alta Bates Summit Medical Center, Berkeley, California, USA
| | - Huseyin S Bozkurt
- Clinic of Gastroenterology, Istanbul Maltepe University, Istanbul, Turkey
| | - Sonya Dave
- N/A, Microbiome Research, Inc, Ventura, California, USA
- Medical Writing and Biostatistics, North End Advisory, Smyrna, Georgia, USA
| | | | | | | | - Eamonn Mm Quigley
- Division of Gastroenterology and Hepatology, The Methodist Hospital, Weill Cornell Medical College, Houston, Texas, USA
| | - Thomas J Borody
- N/A, Centre for Digestive Diseases, Five Dock, New South Wales, Australia
| |
Collapse
|
24
|
Farsi Y, Tahvildari A, Arbabi M, Vazife F, Sechi LA, Shahidi Bonjar AH, Jamshidi P, Nasiri MJ, Mirsaeidi M. Diagnostic, Prognostic, and Therapeutic Roles of Gut Microbiota in COVID-19: A Comprehensive Systematic Review. Front Cell Infect Microbiol 2022; 12:804644. [PMID: 35310853 PMCID: PMC8930898 DOI: 10.3389/fcimb.2022.804644] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 02/07/2022] [Indexed: 12/14/2022] Open
Abstract
Introduction The Coronavirus Disease 2019 (COVID-19) pandemic caused by Severe Acute Respiratory Coronavirus 2 (SARS-CoV-2) emerged in late December 2019. Considering the important role of gut microbiota in maturation, regulation, and induction of the immune system and subsequent inflammatory processes, it seems that evaluating the composition of gut microbiota in COVID-19 patients compared with healthy individuals may have potential value as a diagnostic and/or prognostic biomarker for the disease. Also, therapeutic interventions affecting gut microbial flora may open new horizons in the treatment of COVID-19 patients and accelerating their recovery. Methods A systematic search was conducted for relevant studies published from December 2019 to December 2021 using Pubmed/Medline, Embase, and Scopus. Articles containing the following keywords in titles or abstracts were selected: "SARS-CoV-2" or "COVID-19" or "Coronavirus Disease 19" and "gastrointestinal microbes" or "dysbiosis" or "gut microbiota" or "gut bacteria" or "gut microbes" or "gastrointestinal microbiota". Results Out of 1,668 studies, 22 articles fulfilled the inclusion criteria and a total of 1,255 confirmed COVID-19 patients were examined. All included studies showed a significant association between COVID-19 and gut microbiota dysbiosis. The most alteration in bacterial composition of COVID-19 patients was depletion in genera Ruminococcus, Alistipes, Eubacterium, Bifidobacterium, Faecalibacterium, Roseburia, Fusicathenibacter, and Blautia and enrichment of Eggerthella, Bacteroides, Actinomyces, Clostridium, Streptococcus, Rothia, and Collinsella. Also, some gut microbiome alterations were associated with COVID-19 severity and poor prognosis including the increment of Bacteroides, Parabacteroides, Clostridium, Bifidobacterium, Ruminococcus, Campylobacter, Rothia, Corynebacterium, Megasphaera, Enterococcus, and Aspergillus spp. and the decrement of Roseburia, Eubacterium, Lachnospira, Faecalibacterium, and the Firmicutes/Bacteroidetes ratio. Conclusion Our study showed a significant change of gut microbiome composition in COVID-19 patients compared with healthy individuals. This great extent of impact has proposed the gut microbiota as a potential diagnostic, prognostic, and therapeutic strategy for COVID-19. There is much evidence about this issue, and it is expected to be increased in near future.
Collapse
Affiliation(s)
- Yeganeh Farsi
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Azin Tahvildari
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mahta Arbabi
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fateme Vazife
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Leonardo A. Sechi
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
- Struttura Complessa (SC), Microbiologia e Virologia, Azienda Ospedaliera Universitaria, Sassari, Italy
| | - Amir Hashem Shahidi Bonjar
- Clinician Scientist of Dental Materials and Restorative Dentistry, School of Dentistry, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Parnian Jamshidi
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Javad Nasiri
- Department of Microbiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mehdi Mirsaeidi
- Division of Pulmonary and Critical Care, College of Medicine-Jacksonville, University of Florida, Jacksonville, FL, United States
| |
Collapse
|
25
|
Zhao S, Feng P, Meng W, Jin W, Li X, Li X. Modulated Gut Microbiota for Potential COVID-19 Prevention and Treatment. Front Med (Lausanne) 2022; 9:811176. [PMID: 35308540 PMCID: PMC8927624 DOI: 10.3389/fmed.2022.811176] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 02/08/2022] [Indexed: 12/12/2022] Open
Abstract
COVID-19, caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has gained global attention. SARS-CoV-2 identifies and invades human cells via angiotensin-converting enzyme 2 receptors, which is highly expressed both in lung tissues and intestinal epithelial cells. The existence of the gut-lung axis in disease could be profoundly important for both disease etiology and treatment. Furthermore, several studies reported that infected patients suffer from gastrointestinal symptoms. The gut microbiota has a noteworthy effect on the intestinal barrier and affects many aspects of human health, including immunity, metabolism, and the prevention of several diseases. This review highlights the function of the gut microbiota in the host's immune response, providing a novel potential strategy through the use of probiotics, gut microbiota metabolites, and dietary products to enhance the gut microbiota as a target for COVID-19 prevention and treatment.
Collapse
Affiliation(s)
- Shuai Zhao
- Intersection Laboratory of Life Medicine, School of Life Sciences, Lanzhou University, Lanzhou, China
| | - Pengya Feng
- Intersection Laboratory of Life Medicine, School of Life Sciences, Lanzhou University, Lanzhou, China
| | - Wenbo Meng
- Medical Frontier Innovation Research Center, Institute of Cancer Neuroscience, The First Hospital of Lanzhou University, The First Clinical Medical College of Lanzhou University, Lanzhou, China
| | - Weilin Jin
- Medical Frontier Innovation Research Center, Institute of Cancer Neuroscience, The First Hospital of Lanzhou University, The First Clinical Medical College of Lanzhou University, Lanzhou, China
| | - Xun Li
- Medical Frontier Innovation Research Center, Institute of Cancer Neuroscience, The First Hospital of Lanzhou University, The First Clinical Medical College of Lanzhou University, Lanzhou, China
| | - Xiangkai Li
- Intersection Laboratory of Life Medicine, School of Life Sciences, Lanzhou University, Lanzhou, China
| |
Collapse
|
26
|
Zafar H, Saier MH. Comparative Analyses of the Transport Proteins Encoded within the Genomes of nine Bifidobacterium Species. Microb Physiol 2022; 32:30-44. [PMID: 34555832 PMCID: PMC8940750 DOI: 10.1159/000518954] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Accepted: 08/02/2021] [Indexed: 01/03/2023]
Abstract
The human microbiome influences human health in both negative and positive ways. Studies on the transportomes of these organisms yield information that may be utilized for various purposes, including the identification of novel drug targets and the manufacture of improved probiotic strains. Moreover, these genomic analyses help to improve our understanding of the physiology and metabolic capabilities of these organisms. The present study is a continuation of our studies on the transport proteins of the major gut microbes. Bifidobacterium species are essential members of the human gut microbiome, and they initiate colonization of the gut at birth, providing health benefits that last a lifetime. In this study we analyze the transportomes of nine bifidobacterial species: B. adolescentis, B. animalis, B. bifidum, B. breve, B. catenulatum, B. dentium, B. longum subsp. infantis, B. longum subsp. longum, and B. pseudocatenulatum. All of these species have proven probiotic characteristics and exert beneficial effects on human health. Surprisingly, we found that all nine of these species have similar pore-forming toxins and drug exporters that may play roles in pathogenesis. These species have transporters for amino acids, carbohydrates, and proteins, essential for their organismal lifestyles and adaption to their respective ecological niches. The strictly probiotic species, B. bifidum, however, contains fewer such transporters, thus indicative of limited interactions with host cells and other gut microbial counterparts. The results of this study were compared with those of our previous studies on the transportomes of multiple species of Bacteroides, Escherichia coli/Salmonella, and Lactobacillus. Overall, bifidobacteria have larger transportomes (based on percentages of total proteins) than the previously examined groups of bacterial species, with a preference for primary active transport systems over secondary carriers. Taken together, these results provide useful information about the physiologies and pathogenic potentials of these probiotic organisms as reflected by their transportomes.
Collapse
Affiliation(s)
- Hassan Zafar
- Department of Molecular Biology, Division of Biological Sciences, University of California at San Diego, La Jolla, CA 92093-0116.,Central European Institute of Technology, Masaryk University, Kamenice 5, Brno, 625 00, Czech Republic.,Corresponding Authors HZ: Tel: +420773283624, ; MS: Tel: +1 858 534 4084, Fax: +1 858 534 7108,
| | - Milton H. Saier
- Department of Molecular Biology, Division of Biological Sciences, University of California at San Diego, La Jolla, CA 92093-0116.,Corresponding Authors HZ: Tel: +420773283624, ; MS: Tel: +1 858 534 4084, Fax: +1 858 534 7108,
| |
Collapse
|
27
|
Siregar GA, Tambunan AL. Role of Probiotic for Prevention and Management of COVID-19: A Literature Review. Open Access Maced J Med Sci 2021. [DOI: 10.3889/oamjms.2021.7435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Backgrounds: Coronavirus brought about by SARS-CoV-2 is a continuous worldwide pandemic. SARS-CoV-2 influences the human respiratory tracts’ epithelial cells, prompting a proinflammatory cytokine storm and ongoing lung inflammation. With various patients kicking the bucket day by day, an immunization and explicit antiviral medication regimens are being investigated. The choice to utilize this medication during the COVID-19 pandemic should be founded on cautious thought of the likely as preventive and curative in such context. Methods: A literature review changed into carried out through the PubMed, Science Direct, Medline, and Google Scholar search engines consist of probiotics preventive and management possibility in COVID-19. Results: The probiotics significantly affects the host insusceptible reaction, foundationally, and on invulnerable responses at close by mucosal locales, like the lung. Certain strains of probiotics have improved interferon type I levels, hoisting the number and capacity of antigen-introducing cells, regular executioner cells, and T cells, just as expanding the degree of specific antibodies at the fundamental and mucosal destinations. Conclusion: Probiotics have various advantages and potential in preventing and as the treatment of COVID-19, adjusting the arrangement of human gut microflora, reinforcing gut obstruction work, and defensive invulnerable reactions.
Collapse
|
28
|
Tóth AG, Csabai I, Judge MF, Maróti G, Becsei Á, Spisák S, Solymosi N. Mobile Antimicrobial Resistance Genes in Probiotics. Antibiotics (Basel) 2021; 10:antibiotics10111287. [PMID: 34827225 PMCID: PMC8614787 DOI: 10.3390/antibiotics10111287] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 10/14/2021] [Accepted: 10/20/2021] [Indexed: 01/19/2023] Open
Abstract
Even though people worldwide tend to consume probiotic products for their beneficial health effects on a daily basis, recently, concerns were outlined regarding the uptake and potential intestinal colonisation of the bacteria that they carry. These bacteria are capable of executing horizontal gene transfer (HGT) which facilitates the movement of various genes, including antimicrobial resistance genes (ARGs), among the donor and recipient bacterial populations. Within our study, 47 shotgun sequencing datasets deriving from various probiotic samples (isolated strains and metagenomes) were bioinformatically analysed. We detected more than 70 ARGs, out of which rpoB mutants conferring resistance to rifampicin, tet(W/N/W) and potentially extended-spectrum beta-lactamase (ESBL) coding TEM-116 were the most common. Numerous ARGs were associated with integrated mobile genetic elements, plasmids or phages promoting the HGT. Our findings raise clinical and public health concerns as the consumption of probiotic products may lead to the transfer of ARGs to human gut bacteria.
Collapse
Affiliation(s)
- Adrienn Gréta Tóth
- Health Services Management Training Centre, Semmelweis University, 1125 Budapest, Hungary;
| | - István Csabai
- Department of Phyisics of Complex Systems, Eötvös Loránd University, 1117 Budapest, Hungary; (I.C.); (Á.B.)
| | - Maura Fiona Judge
- Centre for Bioinformatics, University of Veterinary Medicine Budapest, 1078 Budapest, Hungary;
| | - Gergely Maróti
- Institute of Plant Biology, Biological Research Center, 6726 Szeged, Hungary;
- Faculty of Water Sciences, University of Public Service, 6500 Baja, Hungary
| | - Ágnes Becsei
- Department of Phyisics of Complex Systems, Eötvös Loránd University, 1117 Budapest, Hungary; (I.C.); (Á.B.)
| | - Sándor Spisák
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA;
| | - Norbert Solymosi
- Centre for Bioinformatics, University of Veterinary Medicine Budapest, 1078 Budapest, Hungary;
- Correspondence: ; Tel.: +36-30-9347-069
| |
Collapse
|
29
|
Viganò C, Mulinacci G, Palermo A, Barisani D, Pirola L, Fichera M, Invernizzi P, Massironi S. Impact of COVID-19 on inflammatory bowel disease practice and perspectives for the future. World J Gastroenterol 2021; 27:5520-5535. [PMID: 34588749 PMCID: PMC8433611 DOI: 10.3748/wjg.v27.i33.5520] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 05/13/2021] [Accepted: 08/02/2021] [Indexed: 02/06/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19) is an infectious disease caused by the novel coronavirus severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2); since its first description in December 2019, it has rapidly spread to a global pandemic. Specific concerns have been raised concerning patients with inflammatory bowel diseases (IBD), which are chronic autoimmune inflammatory disorders of the gut that frequently require immunosuppressive and biological therapies to control their activity. Accumulating evidence has so far demonstrated that patients with IBD are not at increased risk of contracting severe acute respiratory syndrome coronavirus 2 infection. As for the general population, the identified risk factors for severe COVID-19 course among IBD patients have been established to be advanced age and the presence of comorbidities. Treatment with high-dose corticosteroids has also been associated with an increased risk of death in IBD patients with COVID-19. Information on COVID-19 is constantly evolving, with data growing at a rapid pace. This will guarantee better knowledge and stronger evidence to help physicians in the choice of the best therapeutic approach for each patient, concurrently controlling for the risk of IBD disease under treatment and the risk of COVID-19 adverse outcomes and balancing the two. Moreover, the impact of the enormous number of severe respiratory patients on healthcare systems and facilities has led to an unprecedented redeployment of healthcare resources, significantly impacting the care of patients with chronic diseases. In this newly changed environment, the primary aim is to avoid harm whilst still providing adequate management. Telemedicine has been applied and is strongly encouraged for patients without the necessity of infusion therapy and whose conditions are stable. The severe acute respiratory syndrome coronavirus 2 pandemic has already revolutionized the management of patients with chronic immune-mediated diseases such as IBD. Direct and indirect effects of the COVID-19 pandemic will be present for some time. This is the reason why continuous research, rapid solutions and constantly updated guidelines are of utmost importance. The aim of the present review is, therefore, to point out what has been learned so far as well as to pinpoint the unanswered questions and perspectives for the future.
Collapse
Affiliation(s)
- Chiara Viganò
- Division of Gastroenterology and Center for Autoimmune Liver Diseases, Department of Medicine and Surgery, University of Milano-Bicocca, Monza 20900, Italy
- European Reference Network on Hepatological Diseases, San Gerardo Hospital, Monza 20900, Italy
| | - Giacomo Mulinacci
- Division of Gastroenterology and Center for Autoimmune Liver Diseases, Department of Medicine and Surgery, University of Milano-Bicocca, Monza 20900, Italy
- European Reference Network on Hepatological Diseases, San Gerardo Hospital, Monza 20900, Italy
| | - Andrea Palermo
- Division of Gastroenterology and Center for Autoimmune Liver Diseases, Department of Medicine and Surgery, University of Milano-Bicocca, Monza 20900, Italy
- European Reference Network on Hepatological Diseases, San Gerardo Hospital, Monza 20900, Italy
| | - Donatella Barisani
- School of Medicine and Surgery, University of Milano-Bicocca, Monza 20900, Italy
| | - Lorena Pirola
- Division of Gastroenterology and Center for Autoimmune Liver Diseases, Department of Medicine and Surgery, University of Milano-Bicocca, Monza 20900, Italy
- European Reference Network on Hepatological Diseases, San Gerardo Hospital, Monza 20900, Italy
| | - Maria Fichera
- Division of Gastroenterology and Center for Autoimmune Liver Diseases, Department of Medicine and Surgery, University of Milano-Bicocca, Monza 20900, Italy
- European Reference Network on Hepatological Diseases, San Gerardo Hospital, Monza 20900, Italy
| | - Pietro Invernizzi
- Division of Gastroenterology and Center for Autoimmune Liver Diseases, Department of Medicine and Surgery, University of Milano-Bicocca, Monza 20900, Italy
- European Reference Network on Hepatological Diseases, San Gerardo Hospital, Monza 20900, Italy
| | - Sara Massironi
- Division of Gastroenterology and Center for Autoimmune Liver Diseases, Department of Medicine and Surgery, University of Milano-Bicocca, Monza 20900, Italy
- European Reference Network on Hepatological Diseases, San Gerardo Hospital, Monza 20900, Italy
| |
Collapse
|
30
|
Nezametdinova VZ, Yunes RA, Dukhinova MS, Alekseeva MG, Danilenko VN. The Role of the PFNA Operon of Bifidobacteria in the Recognition of Host's Immune Signals: Prospects for the Use of the FN3 Protein in the Treatment of COVID-19. Int J Mol Sci 2021; 22:ijms22179219. [PMID: 34502130 PMCID: PMC8430577 DOI: 10.3390/ijms22179219] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 08/21/2021] [Accepted: 08/24/2021] [Indexed: 12/11/2022] Open
Abstract
Bifidobacteria are some of the major agents that shaped the immune system of many members of the animal kingdom during their evolution. Over recent years, the question of concrete mechanisms underlying the immunomodulatory properties of bifidobacteria has been addressed in both animal and human studies. A possible candidate for this role has been discovered recently. The PFNA cluster, consisting of five core genes, pkb2, fn3, aaa-atp, duf58, tgm, has been found in all gut-dwelling autochthonous bifidobacterial species of humans. The sensory region of the species-specific serine-threonine protein kinase (PKB2), the transmembrane region of the microbial transglutaminase (TGM), and the type-III fibronectin domain-containing protein (FN3) encoded by the I gene imply that the PFNA cluster might be implicated in the interaction between bacteria and the host immune system. Moreover, the FN3 protein encoded by one of the genes making up the PFNA cluster, contains domains and motifs of cytokine receptors capable of selectively binding TNF-α. The PFNA cluster could play an important role for sensing signals of the immune system. Among the practical implications of this finding is the creation of anti-inflammatory drugs aimed at alleviating cytokine storms, one of the dire consequences resulting from SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Venera Z. Nezametdinova
- Laboratory of Bacterial Genetics, The Vavilov Institute of General Genetics, 117971 Moscow, Russia; (V.Z.N.); (R.A.Y.); (M.G.A.)
| | - Roman A. Yunes
- Laboratory of Bacterial Genetics, The Vavilov Institute of General Genetics, 117971 Moscow, Russia; (V.Z.N.); (R.A.Y.); (M.G.A.)
| | - Marina S. Dukhinova
- International Institute ‘Solution Chemistry of Advanced Materials and Technologies’, ITMO University, 197101 Saint-Petersburg, Russia;
| | - Maria G. Alekseeva
- Laboratory of Bacterial Genetics, The Vavilov Institute of General Genetics, 117971 Moscow, Russia; (V.Z.N.); (R.A.Y.); (M.G.A.)
| | - Valery N. Danilenko
- Laboratory of Bacterial Genetics, The Vavilov Institute of General Genetics, 117971 Moscow, Russia; (V.Z.N.); (R.A.Y.); (M.G.A.)
- Correspondence:
| |
Collapse
|
31
|
Chen ZR, Liu J, Liao ZG, Zhou J, Peng HW, Gong F, Hu JF, Zhou Y. COVID-19 and gastroenteric manifestations. World J Clin Cases 2021; 9:4990-4997. [PMID: 34307549 PMCID: PMC8283602 DOI: 10.12998/wjcc.v9.i19.4990] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 03/14/2021] [Accepted: 05/15/2021] [Indexed: 02/06/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19), caused by the infection of a novel coronavirus [severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)], has become a pandemic. The infection has resulted in about one hundred million COVID-19 cases and millions of deaths. Although SARS-CoV-2 mainly spreads through the air and impairs the function of the respiratory system, it also attacks the gastrointestinal epithelial cells through the same receptor, angiotensin converting enzyme 2 receptor, which results in gastroenteric symptoms and potential fecal-oral transmission. Besides the infection of SARS-CoV-2, the treatments of COVID-19 also contribute to the gastroenteric manifestations due to the adverse drug reactions of anti-COVID-19 drugs. In this review, we update the clinical features, basic studies, and clinical practices of COVID-19-associated gastroenteric manifestations.
Collapse
Affiliation(s)
- Zhang-Ren Chen
- Department of Pharmacy, The First Affiliated Hospital of Nanchang University, Nanchang 330100, Jiangxi Province, China
| | - Jing Liu
- Department of Pharmacy, The First Affiliated Hospital of Nanchang University, Nanchang 330100, Jiangxi Province, China
| | - Zhi-Guo Liao
- Department of Pharmacy, The First Affiliated Hospital of Nanchang University, Nanchang 330100, Jiangxi Province, China
| | - Jian Zhou
- Department of Pharmacy, The First Affiliated Hospital of Nanchang University, Nanchang 330100, Jiangxi Province, China
| | - Hong-Wei Peng
- Department of Pharmacy, The First Affiliated Hospital of Nanchang University, Nanchang 330100, Jiangxi Province, China
| | - Fei Gong
- Department of Pharmacy, The First Affiliated Hospital of Nanchang University, Nanchang 330100, Jiangxi Province, China
| | - Jin-Fang Hu
- Department of Pharmacy, The First Affiliated Hospital of Nanchang University, Nanchang 330100, Jiangxi Province, China
| | - Ying Zhou
- Department of Pharmacy, The First Affiliated Hospital of Nanchang University, Nanchang 330100, Jiangxi Province, China
| |
Collapse
|
32
|
Mirzaei R, Attar A, Papizadeh S, Jeda AS, Hosseini-Fard SR, Jamasbi E, Kazemi S, Amerkani S, Talei GR, Moradi P, Jalalifar S, Yousefimashouf R, Hossain MA, Keyvani H, Karampoor S. The emerging role of probiotics as a mitigation strategy against coronavirus disease 2019 (COVID-19). Arch Virol 2021; 166:1819-1840. [PMID: 33745067 PMCID: PMC7980799 DOI: 10.1007/s00705-021-05036-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Accepted: 01/28/2021] [Indexed: 02/06/2023]
Abstract
COVID-19 is an acute respiratory infection accompanied by pneumonia caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), which has affected millions of people globally. To date, there are no highly efficient therapies for this infection. Probiotic bacteria can interact with the gut microbiome to strengthen the immune system, enhance immune responses, and induce appropriate immune signaling pathways. Several probiotics have been confirmed to reduce the duration of bacterial or viral infections. Immune fitness may be one of the approaches by which protection against viral infections can be reinforced. In general, prevention is more efficient than therapy in fighting viral infections. Thus, probiotics have emerged as suitable candidates for controlling these infections. During the COVID-19 pandemic, any approach with the capacity to induce mucosal and systemic reactions could potentially be useful. Here, we summarize findings regarding the effectiveness of various probiotics for preventing virus-induced respiratory infectious diseases, especially those that could be employed for COVID-19 patients. However, the benefits of probiotics are strain-specific, and it is necessary to identify the bacterial strains that are scientifically established to be beneficial.
Collapse
Affiliation(s)
- Rasoul Mirzaei
- Department of Microbiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
- Student Research Committee, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Adeleh Attar
- Department of Microbiology and Virology, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Saher Papizadeh
- Department of Microbiology and Virology, School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Ali Salimi Jeda
- Department of Virology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Seyed Reza Hosseini-Fard
- Department of Clinical Biochemistry, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Elaheh Jamasbi
- Department of Anatomical Sciences, Kermanshah University of Medical Science, Kermanshah, Iran
| | - Sima Kazemi
- Department of Microbiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Saman Amerkani
- Department of Microbiology and Virology, School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Gholam Reza Talei
- Department of Virology, School of Medicine, Lorestan University of Medical Sciences, Khorramabad, Lorestan, Iran
| | - Pouya Moradi
- Department of Virology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Saba Jalalifar
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Rasoul Yousefimashouf
- Department of Microbiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
- Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Mohammad Akhter Hossain
- The Florey University of Neuroscience and Mental Health, University of Melbourne, Melbourne, VIC, 3010, Australia.
| | - Hossein Keyvani
- Department of Virology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
- Gastrointestinal and Liver Diseases Research Center, Iran University of Medical Sciences, Tehran, Iran.
| | - Sajad Karampoor
- Department of Virology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
- Gastrointestinal and Liver Diseases Research Center, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
33
|
Li Q, Cheng F, Xu Q, Su Y, Cai X, Zeng F, Zhang Y. The role of probiotics in coronavirus disease-19 infection in Wuhan: A retrospective study of 311 severe patients. Int Immunopharmacol 2021; 95:107531. [PMID: 33714884 PMCID: PMC7934664 DOI: 10.1016/j.intimp.2021.107531] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 02/08/2021] [Accepted: 02/22/2021] [Indexed: 01/06/2023]
Abstract
Purpose Researches revealed that probiotics maybe a potential strategy for COVID-19, whereas there is a lack of related evidence. This study aims to analyze the role of probiotics on severe COVID-19 patients. Methods In the current retrospective single-center study, we collected data of 311 consecutive severe patients with confirmed COVID-19 in Wuhan Union Hospital from Feb 3rd to Feb 20th, 2020. Epidemiological, clinical and medication characteristics were compared and analyzed between patients with or without probiotics. Results In total, 93 of the 123 patients (75.61%) who were treated with probiotics survived to hospital discharge with the median inpatient day of 32 days and mean virus clearance time of 23 days, which were significantly longer than those of patients without probiotics. There were no bias in laboratory parameters, except for IL-6 and ESR, which were significantly higher in patients treated probiotics. We tracked the dynamic changes of 8 selected laboratory parameters (IL-6, CRP, total T lymphocytes, NK cells, B lymphocyte, CD4 + T cells, CD8 + T cells and CD4/CD8 ratio) and found that probiotics could not reduce the increased IL-6 levels but possessed the ability to moderate the immunity and decreased the incidence of secondary infection in COVID-19 patients. Conclusions Probiotics could be an effective strategy for the treatment of COVID-19 patients to reduce the secondary infection and moderated the immunity.
Collapse
Affiliation(s)
- Qiang Li
- Department of Pharmacy, Union Hospital, Tongji Medical College, Hua Zhong University of Science and Technology, Wuhan 430022, China; Hubei Province Clinical Research Center for Precision Medicine for Critical Illness, Wuhan 430022, China
| | - Fang Cheng
- Department of Pharmacy, Union Hospital, Tongji Medical College, Hua Zhong University of Science and Technology, Wuhan 430022, China; Hubei Province Clinical Research Center for Precision Medicine for Critical Illness, Wuhan 430022, China
| | - Qiling Xu
- Department of Pharmacy, Union Hospital, Tongji Medical College, Hua Zhong University of Science and Technology, Wuhan 430022, China; Hubei Province Clinical Research Center for Precision Medicine for Critical Illness, Wuhan 430022, China
| | - Yuyong Su
- Department of Pharmacy, Union Hospital, Tongji Medical College, Hua Zhong University of Science and Technology, Wuhan 430022, China; Hubei Province Clinical Research Center for Precision Medicine for Critical Illness, Wuhan 430022, China
| | - Xuefeng Cai
- Department of Pharmacy, Union Hospital, Tongji Medical College, Hua Zhong University of Science and Technology, Wuhan 430022, China; Hubei Province Clinical Research Center for Precision Medicine for Critical Illness, Wuhan 430022, China
| | - Fang Zeng
- Department of Pharmacy, Union Hospital, Tongji Medical College, Hua Zhong University of Science and Technology, Wuhan 430022, China; Hubei Province Clinical Research Center for Precision Medicine for Critical Illness, Wuhan 430022, China.
| | - Yu Zhang
- Department of Pharmacy, Union Hospital, Tongji Medical College, Hua Zhong University of Science and Technology, Wuhan 430022, China; Hubei Province Clinical Research Center for Precision Medicine for Critical Illness, Wuhan 430022, China.
| |
Collapse
|
34
|
Abstract
Oral booster-single strain probiotic bifidobacteria could be a potential strategy for SARS-CoV-2. This study aims to evaluate the role of oral probiotic Bifidobacterium on moderate/severe SARS-CoV-2 inpatients. In this single-center study, we analyzed data of 44 moderate/severe inpatients with diagnosed COVID-19 in Istanbul Maltepe University Medical Faculty Hospital, 2020 from 1 November 2020 to 15 December 2020. Clinical and medication features were compared and analyzed between patients with or without probiotic. In result, 19 of the 44 patients (43.18%) who were administrated with oral booster-single strain probiotic were discharged with the median inpatient day of 7.6 days which were significantly shorter than those of patients without probiotic. There were significant differences in inpatient days, radiological improvement at day 6 and week 3, and reduction in interleukin-6 levels in those receiving oral probiotic therapy. Although the mortality rate was 5% in the probiotic group, it was 25% in the non-probiotic group. Booster-single strain probiotic bifidobacteria could be an effective treatment strategy for moderate/severe SARS-CoV-2 inpatients to reduce the mortality and length of stay in hospital.
Collapse
Affiliation(s)
- Hüseyin S Bozkurt
- Faculty of Medical, Clinic of Gastroenterology, Istanbul Maltepe University, Istanbul, Turkey
| | - Ömer Bilen
- Faculty of Design, Bursa Technical University, Bursa, Turkey
| |
Collapse
|
35
|
Bozkurt HS, Bilen Ö. Gastrointestinal symptoms in COVID-19 could be associated with severe lung involvement and increased readmission rates. EUR J INFLAMM 2021; 19. [DOI: 10.1177/20587392211048259] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/17/2025] Open
Abstract
Introduction SARS-CoV-2 virus manifests itself with primary lung damage but also has intestinal involvement. In this study, we aimed to investigate the frequency of gastrointestinal symptoms (GIS) and the relationship of GIS with readmission to the hospital within 30 days in SARS-CoV-2 infected patients who were hospitalized in a specified pandemic hospital. Materials and Methods Symptomatic patients diagnosed with rapid antibody positivity with real-time polymerase chain reaction and typical thorax computed tomography findings were included in this retrospective cohort observational study. Demographic and clinical data were obtained from electronic medical records. Hospital-associated GIS were considered as experiencing at least one of the GIS such as gas, bloating, diarrhea, and constipation developing within72 h after hospital admission. Results The mean age of the patients was 58 ± 14.4 years and 60.7% were men. 82% of hospitalizations were a moderate and severe disease. 71.4% of patients without GIS had at least one of the GIS after hospitalization. As the severity of the disease increased, the frequency of the severity of gastrointestinal symptom increased. GIS bowel disorders were more prominent in patients with moderate and severe disease. Antibiotic and specific treatment (anti-Il-1, anti-Il-6) contributed to the occurrence of gastrointestinal symptom in SARS-CoV-2 inpatients. Conclusion According to our observations of the second wave of the pandemic, the presence, frequency, and severity of gastrointestinal symptom in inpatient is associated with severity of lung disease and increased readmission rate after discharge.
Collapse
Affiliation(s)
- Hüseyin S Bozkurt
- Clinic of Gastroenterology, Istanbul Maltepe University Faculty Of Medicine, Istanbul, Turkey
| | - Ömer Bilen
- Bursa Techinical University, Bursa, Turkey
| |
Collapse
|