1
|
Slöcker-Barrio M, López-Herce Cid J, Solana-García MJ. The Interplay Between Nutrition and Microbiota and the Role of Probiotics and Symbiotics in Pediatric Infectious Diseases. Nutrients 2025; 17:1222. [PMID: 40218980 PMCID: PMC11990912 DOI: 10.3390/nu17071222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2025] [Revised: 03/28/2025] [Accepted: 03/28/2025] [Indexed: 04/14/2025] Open
Abstract
The interplay between nutrition and infectious diseases has been a central theme in health sciences for the last decades due to its great impact on the pediatric population, especially in immunocompromised patients and critically ill children. As conventional treatment and the development of antimicrobials for most infections standard treatment is either limited or not possible, alternative treatment options should be explored. Recent research shows that early enteral nutrition and nutritional supplements (such as probiotics and symbiotics) could have a pivotal role in promoting a healthy microbiome and subsequently preventing and improving outcomes for certain pediatric infectious diseases. However, understanding the specific mechanism of action and tailoring nutritional interventions remains a significant challenge. The optimal dose range for different probiotic strains and prebiotics and the most effective combination for each treatment indication needs further investigation and is yet to be defined. Additionally, in the era of personalized medicine, goal- and patient-directed treatment are key to optimizing and improving outcomes and minimizing potential complications and side effects, especially in complex and immunocompromised patients. The main objectives of this narrative review are 1. to explore the relationship and the complex interactions between microbiota and the human immune system; 2. to describe the influence of nutrition on infectious diseases; 3. to evaluate the impact of supplementation with probiotics and symbiotics in the prevention and treatment of the most relevant infections in children; and 4. to identify knowledge gaps and potential research priorities regarding the use of these supplements in pediatric patients.
Collapse
Affiliation(s)
- María Slöcker-Barrio
- Pediatric Intensive Care Department, Hospital General Universitario Gregorio Marañón, 28009 Madrid, Spain; (J.L.-H.C.); (M.J.S.-G.)
- Primary Care Interventions to Prevent Maternal and Child Chronic Diseases of Perinatal and Developmental Origin Network (RICORS-SAMID], RD24/0013/0012, Instituto de Salud Carlos III, 28029 Madrid, Spain
- Gregorio Marañón Biomedical Research Institute, 28009 Madrid, Spain
| | - Jesús López-Herce Cid
- Pediatric Intensive Care Department, Hospital General Universitario Gregorio Marañón, 28009 Madrid, Spain; (J.L.-H.C.); (M.J.S.-G.)
- Primary Care Interventions to Prevent Maternal and Child Chronic Diseases of Perinatal and Developmental Origin Network (RICORS-SAMID], RD24/0013/0012, Instituto de Salud Carlos III, 28029 Madrid, Spain
- Gregorio Marañón Biomedical Research Institute, 28009 Madrid, Spain
- Mother and Child and Public Health Department, School of Medicine, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - María José Solana-García
- Pediatric Intensive Care Department, Hospital General Universitario Gregorio Marañón, 28009 Madrid, Spain; (J.L.-H.C.); (M.J.S.-G.)
- Primary Care Interventions to Prevent Maternal and Child Chronic Diseases of Perinatal and Developmental Origin Network (RICORS-SAMID], RD24/0013/0012, Instituto de Salud Carlos III, 28029 Madrid, Spain
- Gregorio Marañón Biomedical Research Institute, 28009 Madrid, Spain
- Mother and Child and Public Health Department, School of Medicine, Universidad Complutense de Madrid, 28040 Madrid, Spain
| |
Collapse
|
2
|
Piazzesi A, Pane S, Romani L, Toto F, Scanu M, Marsiglia R, Del Chierico F, Cotugno N, Palma P, Putignani L. Gut Microbial Signatures Associated with Cryptosporidiosis: A Case Series. Microorganisms 2025; 13:342. [PMID: 40005709 PMCID: PMC11858469 DOI: 10.3390/microorganisms13020342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 01/24/2025] [Accepted: 01/26/2025] [Indexed: 02/27/2025] Open
Abstract
Cryptosporidium spp. are zoonotic protozoan parasites with a global prevalence, with both gastrointestinal and pulmonary involvement. Though symptoms can often be relatively mild, they can become severe and even fatal in children under five, the elderly, and in immunocompromised individuals, making cryptosporidiosis a leading cause of morbidity and mortality in fragile populations. Furthermore, there is an urgent clinical need for alternative therapies against cryptosporidiosis, as currently available FDA-approved treatments are ineffective in the immunocompromised. Recent evidence in animal models suggests that the gut microbiota (GM) can influence both host and parasite biology to influence the course of Cryptosporidium infection. Here, we present GM profiles in five cases of cryptosporidiosis, associated with varying underlying pathologies. We found that moderate-severe cryptosporidiosis was characterized by a reduction in alpha-diversity and an enrichment of Enterococcus spp., while decreases in Bifidobacterium, Gemmiger, and Blautia were detectable in the milder manifestations of the disease. Our results suggest that severe cryptosporidiosis is associated with a stronger change on the GM than is age or underlying pathology. Together with previously published studies in animal models, we believe that these results suggest that the GM could be a potential therapeutic target for human patients as well, particularly in the immunocompromised for whom anti-Cryptosporidium treatment remains largely ineffective.
Collapse
Affiliation(s)
- Antonia Piazzesi
- Management and Diagnostic Innovations & Clinical Pathways Research Area, Unit of Microbiome, Bambino Gesù Children’s Hospital, IRCCS, 00146 Rome, Italy; (A.P.); (F.T.); (M.S.); (R.M.); (F.D.C.)
| | - Stefania Pane
- Unit of Microbiology and Diagnostic Immunology, Unit of Microbiomics, Bambino Gesù Children’s Hospital, IRCCS, 00146 Rome, Italy;
| | - Lorenza Romani
- Infectious Diseases Unit, Bambino Gesù Children’s Hospital, IRCCS, 00146 Rome, Italy;
| | - Francesca Toto
- Management and Diagnostic Innovations & Clinical Pathways Research Area, Unit of Microbiome, Bambino Gesù Children’s Hospital, IRCCS, 00146 Rome, Italy; (A.P.); (F.T.); (M.S.); (R.M.); (F.D.C.)
| | - Matteo Scanu
- Management and Diagnostic Innovations & Clinical Pathways Research Area, Unit of Microbiome, Bambino Gesù Children’s Hospital, IRCCS, 00146 Rome, Italy; (A.P.); (F.T.); (M.S.); (R.M.); (F.D.C.)
| | - Riccardo Marsiglia
- Management and Diagnostic Innovations & Clinical Pathways Research Area, Unit of Microbiome, Bambino Gesù Children’s Hospital, IRCCS, 00146 Rome, Italy; (A.P.); (F.T.); (M.S.); (R.M.); (F.D.C.)
| | - Federica Del Chierico
- Management and Diagnostic Innovations & Clinical Pathways Research Area, Unit of Microbiome, Bambino Gesù Children’s Hospital, IRCCS, 00146 Rome, Italy; (A.P.); (F.T.); (M.S.); (R.M.); (F.D.C.)
| | - Nicola Cotugno
- Unit of Clinical Immunology and Vaccinology, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy; (N.C.); (P.P.)
| | - Paolo Palma
- Unit of Clinical Immunology and Vaccinology, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy; (N.C.); (P.P.)
- Department of Systems Medicine, University of Rome “Tor Vergata”, 00133 Rome, Italy
| | - Lorenza Putignani
- Unit of Microbiology and Diagnostic Immunology, Unit of Microbiomics and Management and Diagnostic Innovations & Clinical Pathways Research Area, Unit of Microbiome, Bambino Gesù Children’s Hospital, IRCCS, 00146 Rome, Italy
| |
Collapse
|
3
|
Minaz A, Alam R, Jiwani U, Vadsaria K, Khan A, Ishaq A, Sultan S, Mohsin M, Sharif A, Nisar YB, Das JK, Soofi S, Ariff S. Efficacy of probiotics for treatment of acute or persistent diarrhoea in children from birth till 10 years: Systematic review and meta-analysis. J Glob Health 2024; 14:04236. [PMID: 39703988 PMCID: PMC11659791 DOI: 10.7189/jogh.14.04236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2024] Open
Abstract
Background Numerous studies have investigated the efficacy of probiotics in treating acute and persistent diarrhoea. However, probiotics have not been established as a recommended management option for diarrhoeal illness by the World Health Organization (WHO). Therefore, we conducted a systematic review of randomised controlled trials to assess the efficacy of probiotics for the management of acute and persistent diarrhoea in children. Methods A systematic search on PubMed, CINAHL, Wiley Cochrane Library, Scopus, Clinicaltrials.gov, and WHO International Clinical Trials Registry Platform (ICTRP) was performed. All studies published in the year 2000 and onwards that assessed the use of probiotics in the management of acute and persistent diarrhoea in children aged 0-10 years were included. The risk of bias was assessed using the Cochrane Risk of Bias II (RoB-2) tool and the quality of evidence was assessed using the Grading of Recommendations, Assessment, Development, and Evaluations (GRADE) approach. This review was commissioned by WHO for revision of their guidelines for childhood diarrhoea management. Results The review included 98 studies with a total of 17 236 participants. Studies were categorised based on the WHO definition of diarrhoea or author-specified definition. In studies considering the WHO definition of diarrhoea, the probiotics group was more likely to achieve clinical cure (risk ratio = 1.12 (95% confidence interval (CI) = 1.01, 1.24, studies = 14)) and reduce the duration of diarrhoea (mean difference = -13.27 hours (95% CI = -16.72, -9.83, studies = 33)) than the control group in children with acute diarrhoea. However, the effect size was small, and statistical heterogeneity was very high, leading to low certainty of evidence. In children with persistent diarrhoea, probiotics reduced the duration of diarrhoea by 95 hours (mean difference = -96.45 (95% CI = -110.53, -82.37, studies = 2)), but the certainty of the evidence was very low. Conclusions The results from this systematic review suggest low certainty of evidence for the effect of probiotics on clinical cure and duration of diarrhoea in children. There was significant diversity in the genus, species, dosages, and duration of treatment in the trial and administration. High levels of heterogeneity reduced the certainty of evidence. Large-scale randomised clinical trials are needed to evaluate specific probiotic strains and doses. In addition, cost-effective analysis studies are needed to be explored in future research. Registration The protocol for this review was registered with the International Prospective Register of Systematic Reviews (PROSPERO: CRD42023449200).
Collapse
Affiliation(s)
- Anmol Minaz
- Centre of Excellence in Women and Child Health, Aga Khan University, Karachi, Pakistan
| | - Ridwa Alam
- Centre of Excellence in Women and Child Health, Aga Khan University, Karachi, Pakistan
| | - Uswa Jiwani
- Centre of Excellence in Women and Child Health, Aga Khan University, Karachi, Pakistan
| | - Khadija Vadsaria
- Centre of Excellence in Women and Child Health, Aga Khan University, Karachi, Pakistan
| | - Ahmad Khan
- Centre of Excellence in Women and Child Health, Aga Khan University, Karachi, Pakistan
| | - Aqsa Ishaq
- Centre of Excellence in Women and Child Health, Aga Khan University, Karachi, Pakistan
| | - Samar Sultan
- Centre of Excellence in Women and Child Health, Aga Khan University, Karachi, Pakistan
| | - Marium Mohsin
- Centre of Excellence in Women and Child Health, Aga Khan University, Karachi, Pakistan
| | - Ashraf Sharif
- University Library, Aga Khan University, Karachi, Pakistan
| | - Yasir Bin Nisar
- Department of Maternal, Newborn, Child and Adolescent Health and Ageing, World Health Organization, Geneva, Switzerland
| | - Jai K Das
- Institute for Global Health and Development, Aga Khan University, Karachi, Pakistan
| | - Sajid Soofi
- Centre of Excellence in Women and Child Health, Aga Khan University, Karachi, Pakistan
| | - Shabina Ariff
- Department of Paediatrics and Child Health, Aga Khan University, Karachi, Pakistan
| |
Collapse
|
4
|
Baek J, Kim BS, Kim Y, Bai J. Safety, Antagonistic Activity, and Probiotic Properties of Lactic Acid Bacteria Isolated from Jeotgal, Korean Fermented Seafoods. J Microbiol Biotechnol 2024; 35:e2411055. [PMID: 39663945 PMCID: PMC11813347 DOI: 10.4014/jmb.2411.11055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2024] [Revised: 11/29/2024] [Accepted: 12/06/2024] [Indexed: 12/13/2024]
Abstract
Probiotics are in high demand in the health functional food market as they effectively inhibit pathogens and improve host health. Therefore, in order to develop novel probiotic strains, new strains were isolated from various type of jeotgal, traditional Korean fermented seafood products, and their safety and probiotic properties have been evaluated. Based on 16S rRNA gene sequence analysis, six strains (JRD1, Pediococcus pentosaceus; JRD2, Lactiplantibacillus plantarum; JRD6, Pediococcus acidilactici; CLJ21, Lactiplantibacillus plantarum; CLJ24, Pediococcus pentosaceus; CLJ28, Leuconostoc mesenteroides subsp. dextranicum) were selected and subjected to further analysis. As a result, all six strains did not show hemolytic activity, antibiotics resistance, and cell cytotoxicity, confirming that they are safe for human use. Among them, JRD1, JRD6, and CLJ24 exhibited high survival rates under simulated gastrointestinal conditions. Additionally, these three strains demonstrated strong adhesion abilities on HT-29 cells, with values of 6.02, 5.77, and 5.86 log CFU/mL, respectively. Furthermore, JRD1, JRD6, and CLJ24 showed relatively high antagonistic activity against both Salmonella Typhimurium and Staphylococcus aureus through competition, exclusion, and displacement of their adhesion. Interestingly, cell-free supernatants (CFS) from three strains effectively inhibited the growth of both S. Typhimurium and S. aureus. Furthermore, CFS of CLJ24, JRD1, and JRD6 demonstrated anti-inflammatory effects in intestinal epithelial cells. The results suggest that CLJ24, JRD1, and JRD6 have potential to be development as functional probiotic strains with both antibacterial and anti-inflammatory activities.
Collapse
Affiliation(s)
- Jihyeon Baek
- Department of Food Science and Technology, Seoul Women’s University, Seoul 01797, Republic of Korea
| | - Bong Sun Kim
- Department of Food Science and Technology, Seoul Women’s University, Seoul 01797, Republic of Korea
| | - Yeonju Kim
- Department of Food Science and Technology, Seoul Women’s University, Seoul 01797, Republic of Korea
| | - Jaewoo Bai
- Department of Food Science and Technology, Seoul Women’s University, Seoul 01797, Republic of Korea
| |
Collapse
|
5
|
Long H, He G, He J, Du TF, Feng P, Zhu C. The protective effect and immunomodulatory ability of orally administrated Lacticaseibacillus rhamnosus GG against Mycoplasma pneumoniae infection in BALB/c mice. PLoS One 2024; 19:e0312318. [PMID: 39453930 PMCID: PMC11508164 DOI: 10.1371/journal.pone.0312318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2024] [Accepted: 08/27/2024] [Indexed: 10/27/2024] Open
Abstract
Mycoplasma pneumoniae represents one of the significant etiologies of community-acquired pneumonia in pediatric patients. However, clinical treatment of M. pneumoniae infection in children has encountered challenges due to the escalating resistance to quinolones. Numerous studies have highlighted the potential of probiotic lactobacillus administration in boosting immune responses to bacterial and viral respiratory infections. In this study, the protective efficacy of pre-oral administration of Lacticaseibacillus rhamnosus GG (LGG), Limosilactobacillus reuteri F275, Lactiplantibacillus plantarum NCIMB 8826, L. plantarum S1 or L. plantarum S2 was evaluated in the BALB/c mice model; it was observed that among these five strains of lactobacillus, the supplementation of LGG exhibited the most significant protective effect against M. pneumoniae infection. Moreover, when administered orally, both live LGG and heat-inactivated LGG have demonstrated efficacy in reducing the burden of M. pneumoniae in the lungs and alleviating pulmonary inflammation. Oral supplementation with LGG resulted in the inhibition of neutrophil recruitment into the lungs and increased recruitment of alveolar macrophages in M. pneumoniae-infected mice. Additionally, LGG supplementation led to increased production of IL-10 and secretory IgA (sIgA), while suppressing the levels of IL-1β, IL-6, IL-17A, and TNF-α in the lungs of mice infected with M. pneumoniae. The data suggests that supplementation with LGG can modulate immune responses, decrease pathogen load, and alleviate inflammatory injury in the lungs of M. pneumoniae-infected mice.
Collapse
Affiliation(s)
- Huanbing Long
- Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Institute of Pathogenic Biology, Hengyang Medical College, University of South China, Hengyang, Hunan, People’s Republic of China
| | - Guiting He
- Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Institute of Pathogenic Biology, Hengyang Medical College, University of South China, Hengyang, Hunan, People’s Republic of China
| | - Jiarong He
- Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Institute of Pathogenic Biology, Hengyang Medical College, University of South China, Hengyang, Hunan, People’s Republic of China
| | - Ting feng Du
- Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Institute of Pathogenic Biology, Hengyang Medical College, University of South China, Hengyang, Hunan, People’s Republic of China
| | - Pengxiao Feng
- Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Institute of Pathogenic Biology, Hengyang Medical College, University of South China, Hengyang, Hunan, People’s Republic of China
| | - Cuiming Zhu
- Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Institute of Pathogenic Biology, Hengyang Medical College, University of South China, Hengyang, Hunan, People’s Republic of China
| |
Collapse
|
6
|
Matar A, Damianos JA, Jencks KJ, Camilleri M. Intestinal Barrier Impairment, Preservation, and Repair: An Update. Nutrients 2024; 16:3494. [PMID: 39458489 PMCID: PMC11509958 DOI: 10.3390/nu16203494] [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: 09/19/2024] [Revised: 10/10/2024] [Accepted: 10/11/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND/OBJECTIVES Our objective was to review published studies of the intestinal barrier and permeability, the deleterious effects of dietary components (particularly fat), the impact of altered intestinal permeability in disease models and human diseases, the role of the microbiome and epigenomics in control of barrier function, and the opportunities to restore normal barrier function with dietary interventions and products of the microbiota. METHODS We conducted a literature review including the following keywords alone or in combination: intestinal barrier, permeability, microbiome, epigenomics, diet, irritable bowel syndrome, inflammatory bowel disease, probiotics. RESULTS Intestinal permeability is modified by a diet including fat, which increases permeability, and nutrients such as fiber, glutamine, zinc, vitamin D, polyphenols, emulsifiers, and anthocyanins, which decrease permeability. There is significant interaction of the microbiome and barrier function, including the inflammatory of luminal/bacterial antigens, and anti-inflammatory effects of commensals or probiotics and their products, including short-chain fatty acids. Epigenomic modification of barrier functions are best illustrated by effects on junction proteins or inflammation. Detailed documentation of the protective effects of diet, probiotics, prebiotics, and microbiota is provided. CONCLUSION intestinal permeability is a critical factor in protection against gastrointestinal diseases and is impacted by nutrients that preserve or heal and repair the barrier and nurture anti-inflammatory effects.
Collapse
Affiliation(s)
| | | | | | - Michael Camilleri
- Clinical Enteric Neuroscience Translational and Epidemiological Research (CENTER), Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN 55905, USA; (A.M.); (J.A.D.)
| |
Collapse
|
7
|
Mandal S, Mondal C, Lyndem LM. Probiotics: an alternative anti-parasite therapy. J Parasit Dis 2024; 48:409-423. [PMID: 39145362 PMCID: PMC11319687 DOI: 10.1007/s12639-024-01680-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 04/27/2024] [Indexed: 08/16/2024] Open
Abstract
This paper review about probiotic effects and mechanism of action against the gut and non-gut helminths and protozoan parasites. Gastrointestinal parasitic infections are considered a serious health problem and are widely distributed globally. The disease process which emanates from this parasite infection provides some of the many public and veterinary health problems in the tropical and sub-tropical countries. Prevention and control of the parasite disease is through antihelmintic and anti-protozoan drugs, but, due to the increasing emergence of such drug resistance, eradication of parasite infestation in human and livestock still lingers a challenge, which requires the development of new alternative strategies. The use of beneficial microorganisms i.e. probiotics is becoming interesting due to their prophylactic application against several diseases including parasite infections. Recent studies on the interactions between probiotics, parasites and host immune cells using animal models and in vitro culture systems has increased considerably and draw much attention, yet the mechanisms of actions mediating the positive effects of these beneficial microorganisms on the hosts remain unexplored. Therefore, the aim of the present review is to summarize the latest findings on the probiotic research against the gut and non-gut parasites of significance.
Collapse
Affiliation(s)
- Sudeshna Mandal
- Visva-Bharati, Parasitology Research Laboratory, Department of Zoology, Santiniketan, 731235 West Bengal India
| | - Chandrani Mondal
- Visva-Bharati, Parasitology Research Laboratory, Department of Zoology, Santiniketan, 731235 West Bengal India
| | - Larisha M. Lyndem
- Visva-Bharati, Parasitology Research Laboratory, Department of Zoology, Santiniketan, 731235 West Bengal India
| |
Collapse
|
8
|
Zheng B, Li M, Zhang T, Li B, Li Q, Saiding Q, Chen W, Guo M, Koo S, Ji X, Tao W. Functional modification of gut bacteria for disease diagnosis and treatment. MED 2024; 5:863-885. [PMID: 38964334 DOI: 10.1016/j.medj.2024.06.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 11/15/2023] [Accepted: 06/12/2024] [Indexed: 07/06/2024]
Abstract
Intestinal bacteria help keep humans healthy by regulating lipid and glucose metabolism as well as the immunological and neurological systems. Oral treatment using intestinal bacteria is limited by the high acidity of stomach fluids and the immune system's attack on foreign bacteria. Scientists have created coatings and workarounds to overcome these limitations and improve bacterial therapy. These preparations have demonstrated promising outcomes, with advances in synthetic biology and optogenetics improving their focused colonization and controlled release. Engineering bacteria preparations have become a revolutionary therapeutic approach that converts intestinal bacteria into cellular factories for medicinal chemical synthesis. The present paper discusses various aspects of engineering bacteria preparations, including wrapping materials, biomedical uses, and future developments.
Collapse
Affiliation(s)
- Bin Zheng
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin 300072, China
| | - Mengyi Li
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin 300072, China
| | - Tiange Zhang
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin 300072, China
| | - Bowen Li
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin 300072, China
| | - Qiuya Li
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin 300072, China
| | - Qimanguli Saiding
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Wei Chen
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Mingming Guo
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin 300072, China
| | - Seyoung Koo
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA; Department of Chemical and Molecular Engineering, Hanyang University ERICA, Ansan, Gyeonggi-do 15588, Republic of Korea.
| | - Xiaoyuan Ji
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin 300072, China.
| | - Wei Tao
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
9
|
Altomare A, Giovanetti M, Baldaro F, Ciccozzi M, Cicala M, Guarino MPL. The Prevention of Viral Infections: The Role of Intestinal Microbiota and Nutritional Factors. Nutrients 2024; 16:2445. [PMID: 39125326 PMCID: PMC11314041 DOI: 10.3390/nu16152445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 07/23/2024] [Accepted: 07/26/2024] [Indexed: 08/12/2024] Open
Abstract
Viral infections pose significant global challenges due to their rapid transmissibility. Therefore, preventing and treating these infections promptly is crucial to curbing their spread. This review focuses on the vital link between nutrition and viral infections, underscoring how dietary factors influence immune system modulation. Malnutrition, characterized by deficiencies in essential nutrients such as vitamins A, C, D, E, and zinc, can impair the immune system, thereby increasing vulnerability to viral infections and potentially leading to more severe health outcomes that complicate recovery. Additionally, emerging evidence highlights the role of commensal microbiota in immune regulation, which can affect hosts' susceptibility to infections. Specific dietary components, including bioactive compounds, vitamins, and probiotics, can beneficially modify gut microbiota, thus enhancing immune response and offering protection against viral infections. This review aims to elucidate the mechanisms by which dietary adjustments and gut microbiota impact the pathogenesis of viral infections, with a particular focus on strengthening the immune system.
Collapse
Affiliation(s)
- Annamaria Altomare
- Department of Sciences and Technologies for Sustainable Development and One Health, Università Campus Bio-Medico di Roma, 00128 Rome, Italy; (A.A.); (M.G.)
- Unit of Gastroenterology, Università Campus Bio-Medico di Roma, 00128 Rome, Italy; (F.B.); (M.P.L.G.)
| | - Marta Giovanetti
- Department of Sciences and Technologies for Sustainable Development and One Health, Università Campus Bio-Medico di Roma, 00128 Rome, Italy; (A.A.); (M.G.)
- Instituto Rene Rachou, Fundação Oswaldo Cruz, Belo Horizonte 30190-002, Brazil
- Climate Amplified Diseases and Epidemics (CLIMADE), Bairro Floresta 31110-370, Brazil
| | - Francesca Baldaro
- Unit of Gastroenterology, Università Campus Bio-Medico di Roma, 00128 Rome, Italy; (F.B.); (M.P.L.G.)
| | - Massimo Ciccozzi
- Unit of Medical Statistics and Molecular Epidemiology, Università Campus Bio-Medico di Roma, 00128 Rome, Italy;
| | - Michele Cicala
- Unit of Gastroenterology, Università Campus Bio-Medico di Roma, 00128 Rome, Italy; (F.B.); (M.P.L.G.)
- Unit of Gastroenterology and Digestive Endoscopy, Fondazione Policlinico Campus Bio-Medico di Roma, 00128 Rome, Italy
| | - Michele Pier Luca Guarino
- Unit of Gastroenterology, Università Campus Bio-Medico di Roma, 00128 Rome, Italy; (F.B.); (M.P.L.G.)
- Unit of Gastroenterology and Digestive Endoscopy, Fondazione Policlinico Campus Bio-Medico di Roma, 00128 Rome, Italy
| |
Collapse
|
10
|
Leser T, Baker A. Molecular Mechanisms of Lacticaseibacillus rhamnosus, LGG ® Probiotic Function. Microorganisms 2024; 12:794. [PMID: 38674738 PMCID: PMC11051730 DOI: 10.3390/microorganisms12040794] [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: 02/26/2024] [Revised: 04/10/2024] [Accepted: 04/12/2024] [Indexed: 04/28/2024] Open
Abstract
To advance probiotic research, a comprehensive understanding of bacterial interactions with human physiology at the molecular and cellular levels is fundamental. Lacticaseibacillus rhamnosus LGG® is a bacterial strain that has long been recognized for its beneficial effects on human health. Probiotic effector molecules derived from LGG®, including secreted proteins, surface-anchored proteins, polysaccharides, and lipoteichoic acids, which interact with host physiological processes have been identified. In vitro and animal studies have revealed that specific LGG® effector molecules stimulate epithelial cell survival, preserve intestinal barrier integrity, reduce oxidative stress, mitigate excessive mucosal inflammation, enhance IgA secretion, and provide long-term protection through epigenetic imprinting. Pili on the cell surface of LGG® promote adhesion to the intestinal mucosa and ensure close contact to host cells. Extracellular vesicles produced by LGG® recapitulate many of these effects through their cargo of effector molecules. Collectively, the effector molecules of LGG® exert a significant influence on both the gut mucosa and immune system, which promotes intestinal homeostasis and immune tolerance.
Collapse
Affiliation(s)
- Thomas Leser
- Future Labs, Human Health Biosolutions, Novonesis, Kogle Alle 6, 2970 Hoersholm, Denmark;
| | | |
Collapse
|
11
|
Kanauchi O, Low ZX, Jounai K, Tsuji R, AbuBakar S. Overview of anti-viral effects of probiotics via immune cells in pre-, mid- and post-SARS-CoV2 era. Front Immunol 2023; 14:1280680. [PMID: 38116008 PMCID: PMC10728489 DOI: 10.3389/fimmu.2023.1280680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 11/20/2023] [Indexed: 12/21/2023] Open
Abstract
The COVID-19 outbreak has caused significant global changes and increased public awareness of SARS-CoV-2. Substantial progress in developing vaccines, enhancing sanitation practices, and implementing various measures to combat the virus, including the utilization of probiotics has been made. This comprehensive review examined the medical impact of clinically proven probiotics on infectious diseases, considering three crucial time periods: before (pre-), during (mid-), and after (post-) COVID-19 pandemic era. This review also showed a perspective on the use of probiotics to stimulate the innate immune system and prevent infectious diseases. In pre-COVID-19 era, several probiotic strains were found to be clinically effective in addressing gastrointestinal infectious diseases, the common cold and flu. However, the mechanism by which probiotics exerted their antiviral effects remained relatively unclear during that period. Nevertheless, probiotics, Lactococcus lactis strain Plasma (LC-Plasma), and others have gained attention for their unique ability to modulate the immune system and demonstrate antiviral properties. While some probiotics have shown promise in alleviating gastrointestinal symptoms linked to COVID-19, their direct effectiveness in treating or preventing COVID-19 progression has not yet been conclusively established. As we transition into the post-COVID-19 era, the relationship between COVID-19 and plasmacytoid dendritic cells (pDCs), a vital component of the innate immune system, has been gradually elucidated. These findings are now being applied in developing novel vaccines and treatments involving interferons and in immune activation research using probiotics as adjuvants, comparable to CpG-DNA through TLR9. The role of the local innate immune system, including pDCs, as the first line of defense against viral infections has gained increasing interest. Moving forward, insight of the immune system and the crosstalk between probiotics and the innate immune system is expected to highlight the role of probiotics in adjunctive immunoregulatory therapy. In combination with drug treatments, probiotics may play a more substantial role in enhancing immune responses. The immunoregulatory approach using probiotics such as LC-Plasma, which can induce anti-infectious factors such as interferons, holds promise as a viable therapeutic and prophylactic option against viral infectious diseases due to their good safety profile and protective efficacy.
Collapse
Affiliation(s)
- Osamu Kanauchi
- Tropical Infectious Disease Research and Education Centre (TIDREC), Institute for Advanced Studies, University of Malaya, Kuala Lumpur, Malaysia
- Institute of Health Sciences, Kirin Holdings Co., Ltd., Fujisawa, Japan
| | - Zhao Xuan Low
- Tropical Infectious Disease Research and Education Centre (TIDREC), Institute for Advanced Studies, University of Malaya, Kuala Lumpur, Malaysia
| | - Kenta Jounai
- Institute of Health Sciences, Kirin Holdings Co., Ltd., Fujisawa, Japan
| | - Ryohei Tsuji
- Institute of Health Sciences, Kirin Holdings Co., Ltd., Fujisawa, Japan
| | - Sazaly AbuBakar
- Tropical Infectious Disease Research and Education Centre (TIDREC), Institute for Advanced Studies, University of Malaya, Kuala Lumpur, Malaysia
| |
Collapse
|
12
|
Liu X, Wang J, Liu J, Li X, Guan Y, Qian S, Jia X. Cryptosporidiosis diagnosed using metagenomic next-generation sequencing in a healthy child admitted to pediatric intensive care unit: a case report. Front Cell Infect Microbiol 2023; 13:1269963. [PMID: 37965251 PMCID: PMC10641789 DOI: 10.3389/fcimb.2023.1269963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 10/12/2023] [Indexed: 11/16/2023] Open
Abstract
Background Cryptosporidium infections in humans typically result in symptoms such as abdominal pain and diarrhea. When the diarrhea is severe, it can cause serious complications and even be life-threatening, especially in patients with compromised immune systems. Case presentation Here, we reported the use of metagenomic next-generation sequencing (mNGS) to assist in the diagnosis and treatment of a 10-year-old boy with severe Cryptosporidium infection. Despite the absence of any history of immunocompromise, the infection still resulted in severe symptoms, including shock, as well as damage to his pancreas and kidneys. The mNGS tests detected the presence of Cryptosporidium parvum when conventional methods failed. The patient received anti-parasite treatment along with supportive care to manage the condition. With disease surveillance based on regular clinical tests and sequential mNGS tests, the child recovered from the severe conditions. Conclusion Our study emphasized the importance of recognizing the potential severity of Cryptosporidium infection, even among individuals with normal immune systems. Timely diagnosis and ongoing monitoring are essential for patient prognosis.
Collapse
Affiliation(s)
- Xiaoyi Liu
- Pediatric Intensive Care Unit, Beijing Children's Hospital, Capital Medical University, Beijing, China
| | - Jing Wang
- Department of Scientific Affairs, Hugobiotech Co., Ltd., Beijing, China
| | - Jun Liu
- Pediatric Intensive Care Unit, Beijing Children's Hospital, Capital Medical University, Beijing, China
| | - Xuming Li
- Department of Scientific Affairs, Hugobiotech Co., Ltd., Beijing, China
| | - Yuanlin Guan
- Department of Scientific Affairs, Hugobiotech Co., Ltd., Beijing, China
| | - Suyun Qian
- Pediatric Intensive Care Unit, Beijing Children's Hospital, Capital Medical University, Beijing, China
| | - Xinlei Jia
- Pediatric Intensive Care Unit, Beijing Children's Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
13
|
Piazzesi A, Pane S, Russo A, Del Chierico F, Francalanci P, Cotugno N, Rossi P, Locatelli F, Palma P, Putignani L. Case Report: The impact of severe cryptosporidiosis on the gut microbiota of a pediatric patient with CD40L immunodeficiency. Front Cell Infect Microbiol 2023; 13:1281440. [PMID: 37965266 PMCID: PMC10641285 DOI: 10.3389/fcimb.2023.1281440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 09/15/2023] [Indexed: 11/16/2023] Open
Abstract
Cryptosporidium parvum is a protozoan parasite and one of the leading causes of gastroenteritis in the world, primarily affecting very young children and immunocompromised patients. While infection is usually self-limiting, it can become chronic and even lethal in these vulnerable populations, in whom Cryptosporidium treatments are generally ineffective, due to their acting in concert with a functioning immune system. Here, we describe a case of chronic cryptosporidiosis in a European child with severe CD40L immunodeficiency infected with Cryptosporidium parvum of the IIa20G1 subgenotype, a lineage which has thus far only ever been described in the Middle East. After years of on-off treatment with conventional and non-conventional anti-parasitic drugs failed to clear parasitosis, we performed targeted metagenomics to observe the bacterial composition of the patient's gut microbiota (GM), and to evaluate fecal microbiota transplantation (FMT) as a potential treatment option. We found that C. parvum infection led to significant shifts in GM bacterial composition in our patient, with consequent shifts in predicted intestinal functional signatures consistent with a state of persistent inflammation. This, combined with the patient's poor prognosis and increasing parasitic burden despite many rounds of anti-parasitic drug treatments, made the patient a potential candidate for an experimental FMT procedure. Unfortunately, given the many comorbidities that were precipitated by the patient's immunodeficiency and chronic C. parvum infection, FMT was postponed in favor of more urgently necessary liver and bone marrow transplants. Tragically, after the first liver transplant failed, the patient lost his life before undergoing FMT and a second liver transplant. With this case report, we present the first description of how cryptosporidiosis can shape the gut microbiota of a pediatric patient with severe immunodeficiency. Finally, we discuss how both our results and the current scientific literature suggest that GM modulations, either by probiotics or FMT, can become novel treatment options for chronic Cryptosporidium infection and its consequent complications, especially in those patients who do not respond to the currently available anti-parasitic therapies.
Collapse
Affiliation(s)
- Antonia Piazzesi
- Unit of Human Microbiome, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Stefania Pane
- Unit of Microbiomics, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Alessandra Russo
- Unit of Microbiomics, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | | | - Paola Francalanci
- Department of Pathology, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Nicola Cotugno
- Unit of Clinical Immunology and Vaccinology, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
- Department of Systems Medicine, Chair of Pediatrics, University of Rome “Tor Vergata”, Rome, Italy
| | - Paolo Rossi
- Department of Systems Medicine, Chair of Pediatrics, University of Rome “Tor Vergata”, Rome, Italy
- Academic Department of Pediatrics, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Franco Locatelli
- Department of Haematology/Oncology and Cell and Gene Therapy, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
- Department of Life Sciences and Public Health, Catholic University of the Sacred Heart, Rome, Italy
| | - Paolo Palma
- Unit of Clinical Immunology and Vaccinology, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
- Department of Systems Medicine, Chair of Pediatrics, University of Rome “Tor Vergata”, Rome, Italy
| | - Lorenza Putignani
- Unit of Microbiomics and Unit of Human Microbiome, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| |
Collapse
|
14
|
Battistini C, Herkenhoff ME, de Souza Leite M, Vieira ADS, Bedani R, Saad SMI. Brewer's Spent Grain Enhanced the Recovery of Potential Probiotic Strains in Fermented Milk After Exposure to In Vitro-Simulated Gastrointestinal Conditions. Probiotics Antimicrob Proteins 2023; 15:326-337. [PMID: 34491541 DOI: 10.1007/s12602-021-09839-8] [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] [Accepted: 08/19/2021] [Indexed: 11/28/2022]
Abstract
Brewer's spent grain (BSG) is a beer industry by-product with interesting functional properties by its high fiber content and bioactive compounds, which may be possibly employed as a prebiotic ingredient. The fermentability of BSG by ten probiotics and two starter cultures was evaluated, and the co-culture of Lacticaseibacillus paracasei subsp. paracasei F-19® (probiotic) and Streptococcus thermophilus TH-4® (starter) was selected to produce a potentially probiotic fermented milk (FM). Four formulations of FM were studied: FM1 (control), FM2 (probiotic - /BSG +), FM3 (probiotic + /BSG -), and FM4 (probiotic + /BSG +). The viability of the microorganisms in the FM was monitored throughout 28 days of storage. The resistance of the microorganisms in the FM to in vitro-simulated gastrointestinal tract (GIT) conditions was also evaluated. Even though the BSG did not influence the fermentation kinetics or increase the populations of both microorganisms in the FM, a significant improvement on the survival of TH-4® against in vitro-simulated GIT stress was observed in the formulations containing BSG alone or in combination with F-19®. All formulations showed potential as probiotic FM, since total probiotic populations were kept above 1010 CFU in a daily portion of 200 mL, and a minimum of 1010 and 108 CFU equivalent of, respectively, TH-4® and F-19® was recovered after the GIT stress. Therefore, TH-4® has potential as a probiotic strain in addition to its starter feature, while BSG may be employed as a possible prebiotic ingredient in a synbiotic approach. Nonetheless, further studies to evaluate possible health benefits are needed.
Collapse
Affiliation(s)
- Carolina Battistini
- Department of Biochemical and Pharmaceutical Technology, School of Pharmaceutical Sciences, University of São Paulo (Universidade de São Paulo, USP), São Paulo, Av. Professor Lineu Prestes, SP, 58005508-000, Brazil.
- Food Research Center FoRC, University of São Paulo (USP), Rua Do Lago, 250, Ed. Semi Industrial, Bloco C, São Paulo, SP, 05508-000, Brazil.
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois At Chicago, Chicago, 840 S Wood Street, Room 1044 CSB, IL, 60612, USA.
| | - Marcos Edgar Herkenhoff
- Department of Biochemical and Pharmaceutical Technology, School of Pharmaceutical Sciences, University of São Paulo (Universidade de São Paulo, USP), São Paulo, Av. Professor Lineu Prestes, SP, 58005508-000, Brazil
- Food Research Center FoRC, University of São Paulo (USP), Rua Do Lago, 250, Ed. Semi Industrial, Bloco C, São Paulo, SP, 05508-000, Brazil
| | - Marcela de Souza Leite
- Department of Biochemical and Pharmaceutical Technology, School of Pharmaceutical Sciences, University of São Paulo (Universidade de São Paulo, USP), São Paulo, Av. Professor Lineu Prestes, SP, 58005508-000, Brazil
- Food Research Center FoRC, University of São Paulo (USP), Rua Do Lago, 250, Ed. Semi Industrial, Bloco C, São Paulo, SP, 05508-000, Brazil
| | - Antonio Diogo Silva Vieira
- Department of Biochemical and Pharmaceutical Technology, School of Pharmaceutical Sciences, University of São Paulo (Universidade de São Paulo, USP), São Paulo, Av. Professor Lineu Prestes, SP, 58005508-000, Brazil
- Food Research Center FoRC, University of São Paulo (USP), Rua Do Lago, 250, Ed. Semi Industrial, Bloco C, São Paulo, SP, 05508-000, Brazil
| | - Raquel Bedani
- Department of Biochemical and Pharmaceutical Technology, School of Pharmaceutical Sciences, University of São Paulo (Universidade de São Paulo, USP), São Paulo, Av. Professor Lineu Prestes, SP, 58005508-000, Brazil
- Food Research Center FoRC, University of São Paulo (USP), Rua Do Lago, 250, Ed. Semi Industrial, Bloco C, São Paulo, SP, 05508-000, Brazil
| | - Susana Marta Isay Saad
- Department of Biochemical and Pharmaceutical Technology, School of Pharmaceutical Sciences, University of São Paulo (Universidade de São Paulo, USP), São Paulo, Av. Professor Lineu Prestes, SP, 58005508-000, Brazil.
- Food Research Center FoRC, University of São Paulo (USP), Rua Do Lago, 250, Ed. Semi Industrial, Bloco C, São Paulo, SP, 05508-000, Brazil.
| |
Collapse
|
15
|
Săsăran MO, Mărginean CO, Adumitrăchioaiei H, Meliț LE. Pathogen-Specific Benefits of Probiotic and Synbiotic Use in Childhood Acute Gastroenteritis: An Updated Review of the Literature. Nutrients 2023; 15:643. [PMID: 36771350 PMCID: PMC9919199 DOI: 10.3390/nu15030643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Revised: 01/24/2023] [Accepted: 01/25/2023] [Indexed: 01/29/2023] Open
Abstract
Probiotics represent viable microorganisms which are found within the normal gut microbiota, that exert strain-specific benefits in the management of several gastrointestinal disorders in children, including acute gastroenteritis. This review aims to evaluate the pathogen-specific role of probiotic supplementation in childhood diarrhea. A search of scientific databases was conducted to identify studies which investigated efficacy of probiotics and synbiotics in influencing outcome of acute gastroenteritis of known etiology. We identified 32 studies, most of which analyzed impact of probiotic supplementation in rotavirus gastroenteritis, while a very limited number of these conducted a separate analysis on bacterial diarrhea. Lactobacillus rhamnosus (L. rhamnosus), L. reuteri and S. boulardii still remain the most researched strains, with a proven role in decreasing diarrhea and hospitalization duration, especially in the setting of rotavirus infection. Combined products containing at least one of the aforementioned strains also performed similarly and might also influence rotavirus fecal shedding. Rotavirus immunization status has also been proposed as a significant influencing factor of probiotic use impact. The paucity of research focusing on bacterial etiologies, as well as of clinical trials conducted within ambulatory care units leaves room for further research on the matter, which needs to include larger cohort studies.
Collapse
Affiliation(s)
- Maria Oana Săsăran
- Department of Pediatrics III Faculty of Medicine in English, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Târgu Mureș, Gheorghe Marinescu Street No 38, 540136 Târgu Mureș, Romania
| | - Cristina Oana Mărginean
- Department of Pediatrics I, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Târgu Mureș, Gheorghe Marinescu Street No 38, 540136 Târgu Mureș, Romania
| | - Heidrun Adumitrăchioaiei
- Doctoral School of Medicine, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Târgu Mureș, Gheorghe Marinescu Street No 38, 540136 Târgu Mureș, Romania
| | - Lorena Elena Meliț
- Department of Pediatrics I, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Târgu Mureș, Gheorghe Marinescu Street No 38, 540136 Târgu Mureș, Romania
| |
Collapse
|
16
|
Mead JR. Early immune and host cell responses to Cryptosporidium infection. FRONTIERS IN PARASITOLOGY 2023; 2:1113950. [PMID: 37325809 PMCID: PMC10269812 DOI: 10.3389/fpara.2023.1113950] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 01/13/2023] [Indexed: 06/17/2023]
Abstract
Cryptosporidium spp. are opportunistic protozoan parasites that infect epithelial cells of the small intestine and cause diarrheal illness in both immunocompetent and immunodeficient individuals. These infections may be more severe in immunocompromised individuals and young children, especially in children under 2 in developing countries. The parasite has a global distribution and is an important cause of childhood diarrhea where it may result in cognitive impairment and growth deficits. Current therapies are limited with nitazoxanide being the only FDA-approved drug. However, it is not efficacious in immunocompromised patients. Additionally, there are no vaccines for cryptosporidiosis available. While acquired immunity is needed to clear Cryptosporidium parasites completely, innate immunity and early responses to infection are important in keeping the infection in check so that adaptive responses have time to develop. Infection is localized to the epithelial cells of the gut. Therefore, host cell defenses are important in the early response to infection and may be triggered through toll receptors or inflammasomes which induce a number of signal pathways, interferons, cytokines, and other immune mediators. Chemokines and chemokine receptors are upregulated which recruit immune cells such neutrophils, NK cells, and macrophages to the infection site to help in host cell defense as well as dendritic cells that are an important bridge between innate and adaptive responses. This review will focus on the host cell responses and the immune responses that are important in the early stages of infection.
Collapse
Affiliation(s)
- Jan R. Mead
- Department of Pediatrics, Children’s Healthcare Organization of Atlanta, Emory University, Atlanta, GA, United States
- Atlanta Veterans Affairs Medical Center, Decatur, GA, United States
| |
Collapse
|
17
|
Zheng Y, Zhang Z, Tang P, Wu Y, Zhang A, Li D, Wang CZ, Wan JY, Yao H, Yuan CS. Probiotics fortify intestinal barrier function: a systematic review and meta-analysis of randomized trials. Front Immunol 2023; 14:1143548. [PMID: 37168869 PMCID: PMC10165082 DOI: 10.3389/fimmu.2023.1143548] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Accepted: 04/10/2023] [Indexed: 05/13/2023] Open
Abstract
Background Probiotics play a vital role in treating immune and inflammatory diseases by improving intestinal barrier function; however, a comprehensive evaluation is missing. The present study aimed to explore the impact of probiotics on the intestinal barrier and related immune function, inflammation, and microbiota composition. A systematic review and meta-analyses were conducted. Methods Four major databases (PubMed, Science Citation Index Expanded, CENTRAL, and Embase) were thoroughly searched. Weighted mean differences were calculated for continuous outcomes with corresponding 95% confidence intervals (CIs), heterogeneity among studies was evaluated utilizing I2 statistic (Chi-Square test), and data were pooled using random effects meta-analyses. Results Meta-analysis of data from a total of 26 RCTs (n = 1891) indicated that probiotics significantly improved gut barrier function measured by levels of TER (MD, 5.27, 95% CI, 3.82 to 6.72, P < 0.00001), serum zonulin (SMD, -1.58, 95% CI, -2.49 to -0.66, P = 0.0007), endotoxin (SMD, -3.20, 95% CI, -5.41 to -0.98, P = 0.005), and LPS (SMD, -0.47, 95% CI, -0.85 to -0.09, P = 0.02). Furthermore, probiotic groups demonstrated better efficacy over control groups in reducing inflammatory factors, including CRP, TNF-α, and IL-6. Probiotics can also modulate the gut microbiota structure by boosting the enrichment of Bifidobacterium and Lactobacillus. Conclusion The present work revealed that probiotics could improve intestinal barrier function, and alleviate inflammation and microbial dysbiosis. Further high-quality RCTs are warranted to achieve a more definitive conclusion. Clinical trial registration https://www.crd.york.ac.uk/PROSPERO/display_record.php?RecordID=281822, identifier CRD42021281822.
Collapse
Affiliation(s)
- Yanfei Zheng
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
- National Institute of Traditional Chinese Medicine (TCM) Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Zengliang Zhang
- Traditional Chinese Medicine College, Inner Mongolia Medical University, Inner Mongolia, China
| | - Ping Tang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
- National Institute of Traditional Chinese Medicine (TCM) Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Yuqi Wu
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
- National Institute of Traditional Chinese Medicine (TCM) Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Anqi Zhang
- National Institute of Traditional Chinese Medicine (TCM) Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Delong Li
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
- National Institute of Traditional Chinese Medicine (TCM) Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Chong-Zhi Wang
- Tang Center for Herbal Medicine Research, The University of Chicago, Chicago, IL, United States
- Department of Anesthesia and Critical Care, The University of Chicago, Chicago, IL, United States
| | - Jin-Yi Wan
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
- National Institute of Traditional Chinese Medicine (TCM) Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing, China
- *Correspondence: Jin-Yi Wan, ; Haiqiang Yao,
| | - Haiqiang Yao
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
- National Institute of Traditional Chinese Medicine (TCM) Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing, China
- *Correspondence: Jin-Yi Wan, ; Haiqiang Yao,
| | - Chun-Su Yuan
- Tang Center for Herbal Medicine Research, The University of Chicago, Chicago, IL, United States
- Department of Anesthesia and Critical Care, The University of Chicago, Chicago, IL, United States
| |
Collapse
|
18
|
Steyer A, Mičetić-Turk D, Fijan S. The Efficacy of Probiotics as Antiviral Agents for the Treatment of Rotavirus Gastrointestinal Infections in Children: An Updated Overview of Literature. Microorganisms 2022; 10:microorganisms10122392. [PMID: 36557645 PMCID: PMC9781831 DOI: 10.3390/microorganisms10122392] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 11/27/2022] [Accepted: 11/29/2022] [Indexed: 12/09/2022] Open
Abstract
Enteric viruses, including the rotavirus, norovirus, and adenoviruses, are the most common cause of acute gastroenteritis. The rotavirus disease is especially prevalent among children, and studies over the past decade have revealed complex interactions between rotaviruses and the gut microbiota. One way to treat and prevent dysbiosis is the use of probiotics as an antiviral agent. This review focuses on the latest scientific evidence on the antiviral properties of probiotics against rotavirus gastroenteric infections in children. A total of 19 studies exhibited a statistically significant antiviral effect of probiotics. The main probiotics that were effective were Saccharomyces cerevisiae var. boulardii, Lacticaseibacillus rhamnosus GG, and various multi-strain probiotics. The underlying mechanism of the probiotics against rotavirus gastroenteric infections in children included immune enhancement and modulation of intestinal microbiota leading to shortening of diarrhoea. However, several clinical studies also found no significant difference in the probiotic group compared to the placebo group even though well-known strains were used, thus showing the importance of correct dosage, duration of treatment, quality of probiotics and the possible influence of other factors, such as the production process of probiotics and the influence of immunisation on the effect of probiotics. Therefore, more robust, well-designed clinical studies addressing all factors are warranted.
Collapse
Affiliation(s)
- Andrej Steyer
- National Laboratory of Health, Environment and Food, Division of Public Health Microbiology, Grablovičeva 44, 1000 Ljubljana, Slovenia
| | - Dušanka Mičetić-Turk
- Department of Paediatrics, Faculty of Medicine, University of Maribor, Taborska ulica 8, 2000 Maribor, Slovenia
- Institute for Health and Nutrition, Faculty of Health Sciences, University of Maribor, Žitna ulica 15, 2000 Maribor, Slovenia
| | - Sabina Fijan
- Institute for Health and Nutrition, Faculty of Health Sciences, University of Maribor, Žitna ulica 15, 2000 Maribor, Slovenia
- Correspondence:
| |
Collapse
|
19
|
Differential Response to the Course of Cryptosporidium parvum Infection and Its Impact on Epithelial Integrity in Differentiated versus Undifferentiated Human Intestinal Enteroids. Infect Immun 2022; 90:e0039722. [PMID: 36286526 PMCID: PMC9671013 DOI: 10.1128/iai.00397-22] [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] [Indexed: 11/20/2022] Open
Abstract
Cryptosporidium is a leading cause of diarrhea and death in young children and untreated AIDS patients and causes waterborne outbreaks. Pathogenic mechanisms underlying diarrhea and intestinal dysfunction are poorly understood. We previously developed stem-cell derived human intestinal enteroid (HIE) models for Cryptosporidium parvum which we used in this study to investigate the course of infection and its effect on intestinal epithelial integrity. By immunofluorescence and confocal microscopy, there was robust infection of undifferentiated and differentiated HIEs in two and three-dimensional (2D, 3D) models. Infection of differentiated HIEs in the 2D model was greater than that of undifferentiated HIEs but lasted only for 3 days, whereas infection persisted for 21 days and resulted in completion of the life cycle in undifferentiated HIEs. Infection of undifferentiated HIE monolayers suggest that C. parvum infects LGR5+ stem cells. Transepithelial electrical resistance measurement of HIEs in the 2D model revealed that infection resulted in decreased epithelial integrity which persisted in differentiated HIEs but recovered in undifferentiated HIEs. Compromised epithelial integrity was reflected in disorganization of the tight and adherens junctions as visualized using the markers ZO-1 and E-cadherin, respectively. Quantitation using the image analysis tools Tight Junction Organizational Rate and Intercellular Junction Organization Quantification, measurement of monolayer height, and RNA transcripts of both proteins by quantitative reverse transcription PCR confirmed that disruption persisted in differentiated HIEs but recovered in undifferentiated HIEs. These models, which more accurately recapitulate human infection, will be useful tools to dissect pathogenic mechanisms underlying diarrhea and intestinal dysfunction in cryptosporidiosis.
Collapse
|
20
|
Vecchio AL, Nunziata F, Bruzzese D, Conelli ML, Guarino A. Rotavirus immunisation status affects the efficacy of Lacticaseibacillus rhamnosus GG for the treatment of children with acute diarrhoea: a meta-analysis. Benef Microbes 2022; 13:283-294. [PMID: 36004717 DOI: 10.3920/bm2022.0024] [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] [Indexed: 11/19/2022]
Abstract
The efficacy of Lacticaseibacillus rhamonosus GG (LGG) for the treatment of children with acute gastroenteritis has been debated based on most recent evidence. Previous evidence demonstrated that LGG mainly benefits children with Rotavirus infection compared to other aetiologies. However, Rotavirus immunisation (RVI) has been implemented worldwide since 2006. We aimed to investigate whether the efficacy of LGG in children with gastroenteritis vary according to RVI status. The MEDLINE, Embase and Cochrane library databases were searched for relevant randomised controlled trials (RCT) up to April 2022. The duration of diarrhoea and episodes lasting >48 h were considered as primary outcomes. The date of vaccine introduction and RVI coverage were reviewed for all countries where trials were conducted. Among the 15 RCTs included in the analysis (n=3,465), only 5 showed a low risk of bias. In RCT conducted before the introduction of RVI (n=2,932), LGG was effective in reducing the duration of diarrhoea compared with placebo or standard care (Median -23.80 h, 95% confidence interval (CI) -36.59 to -11.02]). Only 2 RCTs (n=1,072) reported data of populations partially immunised against Rotavirus with an overall coverage of 44 and 67%, respectively. In this population, LGG showed no efficacy in reducing the duration of diarrhoea (Median -5.34, 95%CI -12.9 to 2.22). Similarly, LGG reduced the risk of diarrhoea lasting >48 h in children not immunised against Rotavirus (RR 0.73, 95%CI 0.54-0.99), but not in population partially immunised (RR 0.98, 95%CI 0.87 to 1.11). The implementation of RVI might affect the efficacy of LGG modifying local epidemiology and susceptibility of the target population to selected probiotics.
Collapse
Affiliation(s)
- A Lo Vecchio
- Department of Translational Medical Sciences - Section of Pediatrics, University of Naples Federico II, Via Pansini 5, 80131 Naples, Italy
| | - F Nunziata
- Department of Translational Medical Sciences - Section of Pediatrics, University of Naples Federico II, Via Pansini 5, 80131 Naples, Italy
| | - D Bruzzese
- Department of Public Health, University of Naples Federico II, Via Pansini 5, 80131 Naples, Italy
| | - M L Conelli
- Department of Translational Medical Sciences - Section of Pediatrics, University of Naples Federico II, Via Pansini 5, 80131 Naples, Italy
| | - A Guarino
- Department of Translational Medical Sciences - Section of Pediatrics, University of Naples Federico II, Via Pansini 5, 80131 Naples, Italy
| |
Collapse
|
21
|
Ross AG, Khanam F, Islam MT, Chowdhury F, Sleigh AC. Diagnosis and Management of Acute Enteropathogens in Returning Travelers. Int J Infect Dis 2022; 123:34-40. [DOI: 10.1016/j.ijid.2022.08.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 07/29/2022] [Accepted: 08/01/2022] [Indexed: 10/16/2022] Open
|
22
|
Xavier-Santos D, Scharlack NK, Pena FDL, Antunes AEC. Effects of Lacticaseibacillus rhamnosus GG supplementation, via food and non-food matrices, on children's health promotion: A scoping review. Food Res Int 2022; 158:111518. [PMID: 35840226 DOI: 10.1016/j.foodres.2022.111518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Revised: 06/11/2022] [Accepted: 06/13/2022] [Indexed: 11/04/2022]
Abstract
The literature considers children both a risk group for administering probiotic strains and one of the populations that can most benefit from it. Due to the health benefits associated to probiotic supplementation, this scope review sought to formulate a critical evaluation of how Lacticaseibacillus rhamnosus GG, carried in food and non-food matrices, and experimental design may affect the health promotion of infants and children. In this study, a literature search was conducted in three scientific databases: PubMed, Web of Science, and SciELO to retrieve research, published in English or Spanish, which administered L. rhamnosus GG to infants and children with any disease or in eutrophic condition. Three reviewers with an expert supervision screened 540 articles, published between 2001 and 2022, which were retrieved from the databases. The data extracted was compiled and shown in this scoping review. In total, was included, after criteria observation, 44 articles in this review. Intestinal disorders were the most frequent outcome in these studies (36.4%) and capsules, the most common vehicle for administering the probiotic strain (40.9%). Probiotic strain dose ranged from 105 to 1012 cfu/dose of L. rhamnosus GG and intervention length extended from one to more than 6 months. Food matrix showed health effects in 57.1% of the clinical trials and non-food matrix 46.7%, which indicates that the health-promoting effect of the probiotic GG strain may be equivalent between the two forms of delivery. However, the highly heterogeneous experimental designs prevent further analysis and a systematic review and meta-analysis is recommended to address just the outcomes of studies and achieve data homogeneity in order to determine which vehicle is the most suitable for health promoting.
Collapse
Affiliation(s)
- Douglas Xavier-Santos
- School of Applied Sciences (FCA), State University of Campinas, 300 Pedro Zaccaria St, 13484-350 Limeira, SP, Brazil
| | - Nayara Kastem Scharlack
- School of Applied Sciences (FCA), State University of Campinas, 300 Pedro Zaccaria St, 13484-350 Limeira, SP, Brazil
| | - Fabíola de Lima Pena
- School of Applied Sciences (FCA), State University of Campinas, 300 Pedro Zaccaria St, 13484-350 Limeira, SP, Brazil
| | | |
Collapse
|
23
|
Barta DG, Cornea-Cipcigan M, Margaoan R, Vodnar DC. Biotechnological Processes Simulating the Natural Fermentation Process of Bee Bread and Therapeutic Properties-An Overview. Front Nutr 2022; 9:871896. [PMID: 35571893 PMCID: PMC9097220 DOI: 10.3389/fnut.2022.871896] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Accepted: 03/21/2022] [Indexed: 12/12/2022] Open
Abstract
Recent signs of progress in functional foods and nutraceuticals highlighted the favorable impact of bioactive molecules on human health and longevity. As an outcome of the fermentation process, an increasing interest is developed in bee products. Bee bread (BB) is a different product intended for humans and bees, resulting from bee pollen's lactic fermentation in the honeycombs, abundant in polyphenols, nutrients (vitamins and proteins), fatty acids, and minerals. BB conservation is correlated to bacteria metabolites, mainly created by Pseudomonas spp., Lactobacillus spp., and Saccharomyces spp., which give lactic acid bacteria the ability to outperform other microbial groups. Because of enzymatic transformations, the fermentation process increases the content of new compounds. After the fermentation process is finalized, the meaningful content of lactic acid and several metabolites prevent the damage caused by various pathogens that could influence the quality of BB. Over the last few years, there has been an increase in bee pollen fermentation processes to unconventional dietary and functional supplements. The use of the chosen starters improves the bioavailability and digestibility of bioactive substances naturally found in bee pollen. As a consequence of enzymatic changes, the fermentation process enhances BB components and preserves them against loss of characteristics. In this aspect, the present review describes the current biotechnological advancements in the development of BB rich in beneficial components derived from bee pollen fermentation and its use as a food supplement and probiotic product with increased shelf life and multiple health benefits.
Collapse
Affiliation(s)
- Daniel Gabriel Barta
- Institute of Life Sciences, University of Agricultural Sciences and Veterinary Medicine Cluj-Napoca, Cluj-Napoca, Romania.,Faculty of Food Science and Technology, University of Agricultural Sciences and Veterinary Medicine Cluj-Napoca, Cluj-Napoca, Romania
| | - Mihaiela Cornea-Cipcigan
- Advanced Horticultural Research Institute of Transylvania, University of Agricultural Sciences and Veterinary Medicine Cluj-Napoca, Cluj-Napoca, Romania
| | - Rodica Margaoan
- Advanced Horticultural Research Institute of Transylvania, University of Agricultural Sciences and Veterinary Medicine Cluj-Napoca, Cluj-Napoca, Romania
| | - Dan Cristian Vodnar
- Institute of Life Sciences, University of Agricultural Sciences and Veterinary Medicine Cluj-Napoca, Cluj-Napoca, Romania.,Faculty of Food Science and Technology, University of Agricultural Sciences and Veterinary Medicine Cluj-Napoca, Cluj-Napoca, Romania
| |
Collapse
|
24
|
Pane S, Putignani L. Cryptosporidium: Still Open Scenarios. Pathogens 2022; 11:pathogens11050515. [PMID: 35631036 PMCID: PMC9143492 DOI: 10.3390/pathogens11050515] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 04/20/2022] [Accepted: 04/23/2022] [Indexed: 01/27/2023] Open
Abstract
Cryptosporidiosis is increasingly identified as a leading cause of childhood diarrhea and malnutrition in both low-income and high-income countries. The strong impact on public health in epidemic scenarios makes it increasingly essential to identify the sources of infection and understand the transmission routes in order to apply the right prevention or treatment protocols. The objective of this literature review was to present an overview of the current state of human cryptosporidiosis, reviewing risk factors, discussing advances in the drug treatment and epidemiology, and emphasizing the need to identify a government system for reporting diagnosed cases, hitherto undervalued.
Collapse
Affiliation(s)
- Stefania Pane
- Department of Diagnostic and Laboratory Medicine, Bambino Gesù Children’s Hospital, IRCCS, Unit of Microbiology and Diagnostic Immunology, Unit of Microbiomics, 00146 Rome, Italy;
| | - Lorenza Putignani
- Department of Diagnostic and Laboratory Medicine, Bambino Gesù Children’s Hospital, IRCCS, Unit of Microbiology and Diagnostic Immunology, Unit of Microbiomics and Multimodal Laboratory Medicine Research Area, Unit of Human Microbiome, 00146 Rome, Italy
- Correspondence:
| |
Collapse
|
25
|
Debnath N, Kumar A, Yadav AK. Probiotics as a biotherapeutics for the management and prevention of respiratory tract diseases. Microbiol Immunol 2022; 66:277-291. [DOI: 10.1111/1348-0421.12980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Revised: 03/20/2022] [Accepted: 04/11/2022] [Indexed: 11/27/2022]
Affiliation(s)
- Nabendu Debnath
- Centre for Molecular Biology Central University of Jammu Samba 181143 Jammu and Kashmir (UT) India
| | - Ashwani Kumar
- Department of Nutrition Biology Central University of Haryana, Mahendergarh Jant‐Pali 123031 Haryana India
| | - Ashok Kumar Yadav
- Centre for Molecular Biology Central University of Jammu Samba 181143 Jammu and Kashmir (UT) India
| |
Collapse
|
26
|
Tai YH, Chang ML, Chu PH, Yeh CC, Cherng YG, Chen TL, Liao CC. Risk of Acute Myocardial Infarction in Patients with Gastroenteritis: A Nationwide Case-Control Study. J Clin Med 2022; 11:1341. [PMID: 35268431 PMCID: PMC8911228 DOI: 10.3390/jcm11051341] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 02/22/2022] [Accepted: 02/25/2022] [Indexed: 01/27/2023] Open
Abstract
Gastroenteritis promotes the development of systemic inflammation and a hypercoagulable state. There are limited data regarding the association between gastroenteritis and acute myocardial infarction (AMI). We aimed to evaluate the risk of AMI after an episode of gastroenteritis. In this nested case-control study, we selected patients who were hospitalized for AMI (N = 103,584) as a case group during 2010-2017 and performed propensity score matching (case-control ratio 1:1) to select eligible controls from insurance research data in Taiwan. We applied multivariable logistic regressions to calculate adjusted odds ratios (ORs) with 95% confidence intervals (CIs) for the risk of AMI associated with recent gastroenteritis within 14 days before AMI. We also compared the outcomes after AMI in patients with or without gastroenteritis. A total of 1381 patients (1.3%) with AMI had a prior episode of gastroenteritis compared to 829 (0.8%) among the controls. Gastroenteritis was significantly associated with a subsequent risk of AMI (adjusted OR: 1.68, 95% CI: 1.54-1.83), which was augmented in hospitalizations for gastroenteritis (adjusted OR: 2.50, 95% CI: 1.20-5.21). The outcomes after AMI were worse in patients with gastroenteritis than in those without gastroenteritis, including increased 30-day in-hospital mortality (adjusted OR: 1.28, 95% CI: 1.08-1.52), medical expenditure, and length of hospital stay. Gastroenteritis may act as a trigger for AMI and correlates with worse post-AMI outcomes. Strategies of aggressive hydration and/or increased antithrombotic therapies for this susceptible population should be further developed.
Collapse
Affiliation(s)
- Ying-Hsuan Tai
- Department of Anesthesiology, Shuang Ho Hospital, Taipei Medical University, New Taipei City 235, Taiwan; (Y.-H.T.); (Y.-G.C.)
- Department of Anesthesiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan;
| | - Ming-Long Chang
- Department of Emergency Medicine, Taipei Medical University Hospital, Taipei 110, Taiwan;
- Department of Emergency Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
| | - Pao-Hsien Chu
- Division of Cardiology, Department of Internal Medicine, Linkou Chang Gung Memorial Hospital, Taoyuan 333, Taiwan;
| | - Chun-Chieh Yeh
- Department of Surgery, China Medical University Hospital, Taichung 404, Taiwan;
- Department of Surgery, University of Illinois, Chicago, IL 60637, USA
| | - Yih-Giun Cherng
- Department of Anesthesiology, Shuang Ho Hospital, Taipei Medical University, New Taipei City 235, Taiwan; (Y.-H.T.); (Y.-G.C.)
- Department of Anesthesiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan;
| | - Ta-Liang Chen
- Department of Anesthesiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan;
- Department of Anesthesiology, Wan Fang Hospital, Taipei Medical University, Taipei 116, Taiwan
- Anesthesiology and Health Policy Research Center, Taipei Medical University Hospital, Taipei 110, Taiwan
| | - Chien-Chang Liao
- Department of Anesthesiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan;
- Anesthesiology and Health Policy Research Center, Taipei Medical University Hospital, Taipei 110, Taiwan
- Department of Anesthesiology, Taipei Medical University Hospital, Taipei 110, Taiwan
- Research Center of Big Data and Meta-Analysis, Wan Fang Hospital, Taipei Medical University, Taipei 116, Taiwan
- School of Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung 404, Taiwan
| |
Collapse
|
27
|
Zhu Y, Wang X, Zhu L, Tu Y, Chen W, Gong L, Pan T, Lin H, Lin J, Sun H, Ge Y, Wei L, Guo Y, Lu C, Chen Y, Xu L. Lactobacillus rhamnosus GG combined with inosine ameliorates alcohol-induced liver injury through regulation of intestinal barrier and Treg/Th1 cells. Toxicol Appl Pharmacol 2022; 439:115923. [PMID: 35176292 DOI: 10.1016/j.taap.2022.115923] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 02/08/2022] [Accepted: 02/11/2022] [Indexed: 02/06/2023]
Abstract
BACKGROUND Intestinal epithelial barrier disruption and bacterial translocation exacerbates the progression of alcoholic liver disease. Lactobacillus rhamnosus GG (LGG), a probiotic, has been shown benefits in chronic liver disease and in regulating gut dysbiosis. Previous studies showed the protective roles of LGG in ethanol-disrupted gut barrier functions and liver injury. Inosine, a metabolite produced by intestinal bacteria, has the anti-inflammatory and immunregulatory functions. In this study, the synergistic effect of LGG and inosine was investigated in a mouse model of alcohol-induced liver disease (ALD). METHODS Male C57BL/6 mice were fed with a Lieber-DeCarli diet containing 5% alcohol for four weeks to establish a model of alcohol-induced liver injury. LGG or a combination of LGG and inosine were administrated orally to explore a new therapeutic method for alcohol-induced liver disease and to investigate the underlying mechanisms. Liver damage was evaluated by transaminases and pathological changes. Tight junction proteins, composition of the gut microbiome, cytokines, lipopolysaccharides (LPS), glutathione (GSH), superoxide dismutase (SOD), malondialdehyde (MDA), F4/80+ macrophages, as well as p38, Jun N-terminal kinase (JNK), were determined by qRT-PCR, RNAseq, ELISA, IHC and western blot. Regulatory T (Treg) cells were characterized by positive staining of CD4, CD25 and Foxp3 using flow cytometry. IFN-γ-producing CD4+ T (Th1) cells were examined by intracellular cytokine staining. RESULTS Alcohol consumption induced elevated liver enzymes, steatosis and inflammation, while LGG combined with inosine treatment was more significant to ameliorate these symptoms compared with LGG alone. When LGG combined with inosine were administered to ALD mice, intestinal microecology significantly improved reflected by intestinal villi and tight junction proteins recovery and the restoration of intestinal flora. Combined therapy inhibited phosphorylation of p38 and JNK to alleviate hepatic inflammation. Moreover, flow cytometry analysis showed that long-term excessive alcohol consumption reduced Tregs population while increased Th1 population, which was restored by a combination of LGG and inosine treatment. CONCLUSIONS The findings from the study indicate that the combined LGG and inosine treatment ameliorates ALD by improving the gut ecosystem, intestinal barrier function, immune homeostasis and liver injury.
Collapse
Affiliation(s)
- Yin Zhu
- Department of Infectious Diseases and liver Diseases, Ningbo Medical Centre Lihuili Hospital, Affiliated Lihuili Hospital of Ningbo University, Ningbo Institute of Innovation for Combined Medicine and Engineering, Ningbo 315040, China; Department of Infectious Diseases, The First Affiliated Hospital of Wenzhou Medical University, Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, Hepatology Institute of Wenzhou Medical University, Wenzhou 325025, China
| | - Xiaozhi Wang
- Department of General and Gastrointestinal Surgery, The Affiliated Mindong Hospital of Fujian Medical University, Fujian 355000, China
| | - Lujian Zhu
- Department of Infectious Diseases, The First Affiliated Hospital of Wenzhou Medical University, Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, Hepatology Institute of Wenzhou Medical University, Wenzhou 325025, China
| | - Yulu Tu
- Department of Infectious Diseases, The First Affiliated Hospital of Wenzhou Medical University, Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, Hepatology Institute of Wenzhou Medical University, Wenzhou 325025, China
| | - Wanting Chen
- Department of Infectious Diseases and liver Diseases, Ningbo Medical Centre Lihuili Hospital, Affiliated Lihuili Hospital of Ningbo University, Ningbo Institute of Innovation for Combined Medicine and Engineering, Ningbo 315040, China
| | - Lingwen Gong
- Department of Infectious Diseases and liver Diseases, Ningbo Medical Centre Lihuili Hospital, Affiliated Lihuili Hospital of Ningbo University, Ningbo Institute of Innovation for Combined Medicine and Engineering, Ningbo 315040, China
| | - Tongtong Pan
- Department of Infectious Diseases, The First Affiliated Hospital of Wenzhou Medical University, Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, Hepatology Institute of Wenzhou Medical University, Wenzhou 325025, China
| | - Hongwei Lin
- Department of Infectious Diseases, The First Affiliated Hospital of Wenzhou Medical University, Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, Hepatology Institute of Wenzhou Medical University, Wenzhou 325025, China
| | - Jing Lin
- Department of Infectious Diseases, The First Affiliated Hospital of Wenzhou Medical University, Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, Hepatology Institute of Wenzhou Medical University, Wenzhou 325025, China
| | - Huiling Sun
- Department of Infectious Diseases and Liver Diseases, Lishui City People's Hospital, Lishui 323020, China
| | - Yuli Ge
- Department of Infectious Diseases and Liver Diseases, Lishui City People's Hospital, Lishui 323020, China
| | - Li Wei
- Department of Infectious Diseases and liver Diseases, Ningbo Medical Centre Lihuili Hospital, Affiliated Lihuili Hospital of Ningbo University, Ningbo Institute of Innovation for Combined Medicine and Engineering, Ningbo 315040, China
| | - Yu Guo
- Department of Infectious Diseases and liver Diseases, Ningbo Medical Centre Lihuili Hospital, Affiliated Lihuili Hospital of Ningbo University, Ningbo Institute of Innovation for Combined Medicine and Engineering, Ningbo 315040, China
| | - Caide Lu
- Department of Hepatobiliary and Pancreatic Surgery, Ningbo Medical Centre Lihuili Hospital, Affiliated Lihuili Hospital of Ningbo University, Ningbo 315040, China.
| | - Yongping Chen
- Department of Infectious Diseases, The First Affiliated Hospital of Wenzhou Medical University, Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, Hepatology Institute of Wenzhou Medical University, Wenzhou 325025, China.
| | - Lanman Xu
- Department of Infectious Diseases and liver Diseases, Ningbo Medical Centre Lihuili Hospital, Affiliated Lihuili Hospital of Ningbo University, Ningbo Institute of Innovation for Combined Medicine and Engineering, Ningbo 315040, China; Department of Infectious Diseases, The First Affiliated Hospital of Wenzhou Medical University, Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, Hepatology Institute of Wenzhou Medical University, Wenzhou 325025, China.
| |
Collapse
|
28
|
Li X, Wang Q, Wu D, Zhang DW, Li SC, Zhang SW, Chen X, Li W. The effect of a novel anticonvulsant chemical Q808 on gut microbiota and hippocampus neurotransmitters in pentylenetetrazole-induced seizures in rats. BMC Neurosci 2022; 23:7. [PMID: 35114941 PMCID: PMC8812211 DOI: 10.1186/s12868-022-00690-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Accepted: 01/28/2022] [Indexed: 11/18/2022] Open
Abstract
Background The gut microbiota can modulate brain function and behavior and is increasingly recognized as an important factor in mediating the risk of epilepsy and the effects of seizure interventions. Drug therapy is one of the factors that influence the composition of the intestinal microbiota. Q808 is an innovative chemical with strong anticonvulsant activity and low neurotoxicity. However, studies evaluating the effect of Q808 on gut microbial communities are lacking. In this study, we aimed to evaluate the anticonvulsant activity of Q808 on a pentylenetetrazol (PTZ)—induced seizure model and analyze and compare the intestinal microbiota composition of non-PTZ vehicle control group, the PTZ-induced seizure model rats with and without Q808, through 16S rDNA sequencing. Neurotransmitter levels in the hippocampus were quantitatively estimated using HPLC–MS. Results The results suggest that Q808 effectively alleviates seizures in chronic PTZ-kindled model rats. Additionally, based on the analyzed abundance of the gut microbiota, dysbacteriosis of model rats was found to be corrected after Q808 treatment at the phylum level. The unique bacterial taxa (e.g., Lactobacillus) that are associated with acetylcholine production, were significantly increased. Several short-chain fatty acids (SCFAs)-producing bacteria, including Roseburia, Alloprevptella, Prevotellaceae_NK3B31_group, Prevotellaceae_UCG-001, and Prevotella_9, were enriched. In the hippocampus, the contents of acetylcholine increased, whereas the levels of 3-methoxytyramine, glutamine, and 5-hydroxyindole acetic acid (5-HIAA) decreased after Q808 treatment. Conclusions This study demonstrates that Q808 can be used to remodel the dysbiosis of the gut microbiome and influence neurotransmitter levels in the hippocampus of PTZ-induced seizure model rats. We hope that these novel findings prompt further research on the interaction between gut microbiota and seizures and the mechanism of Q808. Supplementary Information The online version contains supplementary material available at 10.1186/s12868-022-00690-3.
Collapse
Affiliation(s)
- Xiang Li
- Department of Pharmacology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin, China
| | - Qing Wang
- Academy of Chinese Medical Sciences of Jilin Province, Changchun, Jilin, China
| | - Di Wu
- Academy of Chinese Medical Sciences of Jilin Province, Changchun, Jilin, China
| | - Dian-Wen Zhang
- Academy of Chinese Medical Sciences of Jilin Province, Changchun, Jilin, China
| | | | - Si-Wei Zhang
- Department of Pharmacology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin, China
| | - Xia Chen
- Department of Pharmacology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin, China.
| | - Wei Li
- Academy of Chinese Medical Sciences of Jilin Province, Changchun, Jilin, China.
| |
Collapse
|
29
|
Advances in therapeutic and vaccine targets for Cryptosporidium: Challenges and possible mitigation strategies. Acta Trop 2022; 226:106273. [PMID: 34906550 DOI: 10.1016/j.actatropica.2021.106273] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 12/07/2021] [Accepted: 12/08/2021] [Indexed: 12/14/2022]
Abstract
Cryptosporidium is known to be the second most common diarrheal pathogen in children, causing potentially fatal diarrhea and associated with long-term growth stunting and cognitive deficits. The only Food and Drug Administration-approved treatment for cryptosporidiosis is nitazoxanide, but this drug has not shown potentially effective results in susceptible hosts. Therefore, a safe and effective drug for cryptosporidiosis is urgently needed. Cryptosporidium genome sequencing analysis may help develop an effective drug, but both in vitro and in vivo approaches to drug evaluation are not fully standardized. On the other hand, the development of partial immunity after exposure suggests the possibility of a successful and effective vaccine, but protective surrogates are not precise. In this review, we present our current perspectives on novel cryptosporidiosis therapies, vaccine targets and efficacies, as well as potential mitigation plans, recommendations and perceived challenges.
Collapse
|
30
|
Misra S, Pandey P, Dalbhagat CG, Mishra HN. Emerging Technologies and Coating Materials for Improved Probiotication in Food Products: a Review. FOOD BIOPROCESS TECH 2022; 15:998-1039. [PMID: 35126801 PMCID: PMC8800850 DOI: 10.1007/s11947-021-02753-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 12/16/2021] [Indexed: 12/29/2022]
Abstract
From the past few decades, consumers' demand for probiotic-based functional and healthy food products is rising exponentially. Encapsulation is an emerging field to protect probiotics from unfavorable conditions and to deliver probiotics at the target place while maintaining the controlled release in the colon. Probiotics have been encapsulated for decades using different encapsulation methods to maintain their viability during processing, storage, and digestion and to give health benefits. This review focuses on novel microencapsulation techniques of probiotic bacteria including vacuum drying, microwave drying, spray freeze drying, fluidized bed drying, impinging aerosol technology, hybridization system, ultrasonication with their recent advancement, and characteristics of the commonly used polymers have been briefly discussed. Other than novel techniques, characterization of microcapsules along with their mechanism of release and stability have shown great interest recently in developing novel functional food products with synergetic effects, especially in COVID-19 outbreak. A thorough discussion of novel processing technologies and applications in food products with the incorporation of recent research works is the novelty and highlight of this review paper.
Collapse
Affiliation(s)
- Sourav Misra
- Agricultural and Food Engineering Department, Indian Institute of Technology Kharagpur, Kharagpur, West Bengal 721 302 India
| | - Pooja Pandey
- Agricultural and Food Engineering Department, Indian Institute of Technology Kharagpur, Kharagpur, West Bengal 721 302 India
| | - Chandrakant Genu Dalbhagat
- Agricultural and Food Engineering Department, Indian Institute of Technology Kharagpur, Kharagpur, West Bengal 721 302 India
| | - Hari Niwas Mishra
- Agricultural and Food Engineering Department, Indian Institute of Technology Kharagpur, Kharagpur, West Bengal 721 302 India
| |
Collapse
|
31
|
Suda K, Matsuda K. How Microbes Affect Depression: Underlying Mechanisms via the Gut-Brain Axis and the Modulating Role of Probiotics. Int J Mol Sci 2022; 23:ijms23031172. [PMID: 35163104 PMCID: PMC8835211 DOI: 10.3390/ijms23031172] [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] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 01/19/2022] [Accepted: 01/20/2022] [Indexed: 02/04/2023] Open
Abstract
Accumulating evidence suggests that the gut microbiome influences the brain functions and psychological state of its host via the gut-brain axis, and gut dysbiosis has been linked to several mental illnesses, including major depressive disorder (MDD). Animal experiments have shown that a depletion of the gut microbiota leads to behavioral changes, and is associated with pathological changes, including abnormal stress response and impaired adult neurogenesis. Short-chain fatty acids such as butyrate are known to contribute to the up-regulation of brain-derived neurotrophic factor (BDNF), and gut dysbiosis causes decreased levels of BDNF, which could affect neuronal development and synaptic plasticity. Increased gut permeability causes an influx of gut microbial components such as lipopolysaccharides, and the resultant systemic inflammation may lead to neuroinflammation in the central nervous system. In light of the fact that gut microbial factors contribute to the initiation and exacerbation of depressive symptoms, this review summarizes the current understanding of the molecular mechanisms involved in MDD onset, and discusses the therapeutic potential of probiotics, including butyrate-producing bacteria, which can mediate the microbiota-gut-brain axis.
Collapse
|
32
|
Mirashrafi S, Moravejolahkami AR, Balouch Zehi Z, Hojjati Kermani MA, Bahreini-Esfahani N, Haratian M, Ganjali Dashti M, Pourhossein M. The efficacy of probiotics on virus titres and antibody production in virus diseases: A systematic review on recent evidence for COVID-19 treatment. Clin Nutr ESPEN 2021; 46:1-8. [PMID: 34857182 PMCID: PMC8539817 DOI: 10.1016/j.clnesp.2021.10.016] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Revised: 09/14/2021] [Accepted: 10/18/2021] [Indexed: 02/08/2023]
Abstract
BACKGROUND & AIMS There are some studies indicating the effects of probiotic-containing foods or supplements on viral diseases. We aimed to conduct a rapid review of probiotics with specific emphasis on their potential for early administration in patients at greater risk of SARS-CoV-2 infection. METHODS We searched on PubMed, EMBASE, Google Scholar, Science Direct, Scopus and Web of Science up to February 2021 to identify interventional and observational studies documenting the effects of probiotics strains on interleukins, virus titers, and antibody production with a focus on probiotic-containing foods (PROSPERO Registration ID. CRD42020181453) RESULTS: From a total of 163 records, 21 studies were classified into three domains based on the efficacy of probiotics on 1) the level of interleukins (n = 7), 2) virus titers (n = 2), and 3) interferon (IFN) and antibody production (n = 12). The suppuration of pro-inflammatory interleukins and type I INF production seemed to be the main anti-viral effect of probiotics. Nine studies also indicated the beneficial effects of probiotics and fermented foods on viral diseases. CONCLUSION Based on evidence, some probiotic strains may be useful in viral infections; randomized trials are needed to confirm these findings.
Collapse
Affiliation(s)
- Shahrzad Mirashrafi
- Department of Clinical Nutrition, School of Nutrition & Food Science, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Amir Reza Moravejolahkami
- Department of Clinical Nutrition, School of Nutrition & Food Science, Isfahan University of Medical Sciences, Isfahan, Iran,Corresponding author. Department of Clinical Nutrition, School of Nutrition and Food Sciences, Isfahan University of Medical Sciences, Hezar-Jerib Ave, Isfahan, Iran. P.O. Box 81746-73461. Fax: +98(31)36681378
| | - Zakiyeh Balouch Zehi
- Department of Nutrition, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Ali Hojjati Kermani
- Clinical Tuberculosis and Epidemiology Research Center, National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Nimah Bahreini-Esfahani
- Department of Food Science and Technology, School of Nutrition and Food Science, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mohsen Haratian
- Department of Nutrition, Vice Chancellor of Health, Hamadan University of Medical Sciences, Hamedan, Iran
| | - Marjan Ganjali Dashti
- Department of Biological Sciences, University of Texas at Dallas, 800 W Campbell Road, Richardson, TX, USA
| | - Meraj Pourhossein
- Department of Food Science and Technology, School of Nutrition and Food Science, Isfahan University of Medical Sciences, Isfahan, Iran,Corresponding author. Department of Food Science and Technology, Food Security Research Center, School of Nutrition & Food Science, Isfahan University of Medical Sciences, Hezar-Jerib Ave, Isfahan, Iran. P.O. Box 81746-73461. Fax: +1 519 721. 7430
| |
Collapse
|
33
|
Huang R, Xing HY, Liu HJ, Chen ZF, Tang BB. Efficacy of probiotics in the treatment of acute diarrhea in children: a systematic review and meta-analysis of clinical trials. Transl Pediatr 2021; 10:3248-3260. [PMID: 35070839 PMCID: PMC8753473 DOI: 10.21037/tp-21-511] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 12/01/2021] [Indexed: 11/07/2022] Open
Abstract
BACKGROUND If acute diarrhea in children is not treated promptly and effectively, it can lead to severe dehydration and serious sequelae. Due to the imbalance of intestinal bacteria in children with acute diarrhea, the supplementation with probiotics is important, which can improve the intestinal microenvironment, promote immunity, and enhance resistance. This meta-analysis provides further evidence and discussion of the therapeutic effect of probiotics on acute diarrhea in children. METHODS MEDLINE, EMBASE, PubMed, and the Cochrane Library databases were searched by rapid matching. The input keywords were as follows: (probiotics/synbiotics) and (child/children) and (acute diarrhea/acute gastroenteritis). Articles reporting on randomized controlled trials (RCTs) of probiotics in treating acute diarrhea in children were retrieved. The studies were published from 2010 to 2020. After screening and quality evaluation, Stata 16.0 software was used for the analysis. RESULTS Twelve articles with 744 patients were included in the study, and the overall quality of the articles was excellent. Meta-analysis showed that the duration of diarrhea in the probiotics group was shorter than that in the control group [standard mean difference (SMD) =-0.74, 95% CI: -1.11 to -0.37, Z=-3.935, P=0.000], the 2-day treatment efficacy for diarrhea in the probiotics group was greater than that in the control group [odds ratio (OR) =2.12, 95% CI: 1.47-3.05, Z=3.998, P=0.000], and the length of hospital stay in the probiotics group was shorter than that of the control group (SMD =-0.60, 95% CI: -0.74 to -0.47, Z=-8.781, P=0.000). In the subgroup analysis, combined probiotics shortened the duration of diarrhea compared with single probiotic use, and Lactobacillus reuteri and Saccharomyces boulardii had a better therapeutic effect than Lactobacillus rhamnosus or Lactobacillus acidophilus. DISCUSSION In the treatment of acute diarrhea in children, the addition of probiotics can shorten the duration of diarrhea, increase treatment efficacy after 2 days of treatment, and shorten the length of hospital stay. However, because of possible publication bias in the current study, further high-quality RCT studies in clinical settings are needed to verify the current results and continue the exploration of this topic.
Collapse
Affiliation(s)
- Rao Huang
- Department of Pediatrics, Haikou Hospital of Maternal and Child Health, Haikou, China
| | - Hong-Yi Xing
- Department of Emergency, Haikou Hospital of Maternal and Child Health, Haikou, China
| | - Hong-Juan Liu
- Department of Pediatrics, Haikou Hospital of Maternal and Child Health, Haikou, China
| | - Ze-Fu Chen
- Department of Pediatrics, Hainan General Hospital (Hainan Affiliated Hospital of Hainan Medical University), Haikou, China
| | - Bi-Bo Tang
- Department of Pediatrics, Hainan West Central Hospital, Danzhou, China
| |
Collapse
|
34
|
Javanshir N, Hosseini GNG, Sadeghi M, Esmaeili R, Satarikia F, Ahmadian G, Allahyari N. Evaluation of the Function of Probiotics, Emphasizing the Role of their Binding to the Intestinal Epithelium in the Stability and their Effects on the Immune System. Biol Proced Online 2021; 23:23. [PMID: 34847891 PMCID: PMC8903605 DOI: 10.1186/s12575-021-00160-w] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 11/04/2021] [Indexed: 12/11/2022] Open
Abstract
Due to the importance of using cost-effective methods for therapeutic purposes, the function of probiotics as safe microorganisms and the study of their relevant functional mechanisms have recently been in the spotlight. Finding the mechanisms of attachment and stability and their beneficial effects on the immune system can be useful in identifying and increasing the therapeutic effects of probiotics. In this review, the functional mechanisms of probiotics were comprehensively investigated. Relevant articles were searched in scientific sources, documents, and databases, including PubMed, NCBI, Bactibace, OptiBac, and Bagel4. The most important functional mechanisms of probiotics and their effects on strengthening the epithelial barrier, competitive inhibition of pathogenic microorganisms, production of antimicrobials, binding and interaction with the host, and regulatory effects on the immune system were discussed. In this regard, the attachment of probiotics to the epithelium is very important because the prerequisite for their proper functioning is to establish a proper connection to the epithelium. Therefore, more attention should be paid to the binding effect of probiotics, including sortase A, a significant factor involved in the expression of sortase-dependent proteins (SDP), on their surface as mediators of intestinal epithelial cell binding. In general, by investigating the functional mechanisms of probiotics, it was concluded that the mechanism by which probiotics regulate the immune system and adhesion capacity can directly and indirectly have preventive and therapeutic effects on a wide range of diseases. However, further study of these mechanisms requires extensive research on various aspects.
Collapse
Affiliation(s)
- Nahid Javanshir
- Department of Industrial and Environmental Biotechnology, National Institute of Genetic Engineering and Biotechnology. (NIGEB), P.O. Box: 14155-6343, Tehran, Iran
| | | | - Mahdieh Sadeghi
- Department of Science, Islamic Azad University - Parand Branch, Parand, Iran
| | | | - Fateme Satarikia
- Department of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Gholamreza Ahmadian
- Department of Industrial and Environmental Biotechnology, National Institute of Genetic Engineering and Biotechnology. (NIGEB), P.O. Box: 14155-6343, Tehran, Iran.
| | - Najaf Allahyari
- Department of Industrial and Environmental Biotechnology, National Institute of Genetic Engineering and Biotechnology. (NIGEB), P.O. Box: 14155-6343, Tehran, Iran.
| |
Collapse
|
35
|
Li Z, Zhu G, Li C, Lai H, Liu X, Zhang L. Which Probiotic Is the Most Effective for Treating Acute Diarrhea in Children? A Bayesian Network Meta-Analysis of Randomized Controlled Trials. Nutrients 2021; 13:4319. [PMID: 34959871 PMCID: PMC8706888 DOI: 10.3390/nu13124319] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 11/22/2021] [Accepted: 11/25/2021] [Indexed: 12/13/2022] Open
Abstract
Acute diarrhea is a major cause of morbidity and mortality in children under five. Probiotics are beneficial for treating acute diarrhea in children, but unclear which specific probiotic is the most effective. We performed a Bayesian network meta-analysis to examine the comparative effectiveness of probiotics. By searching EMBASE, PubMed, and the Cochrane Library up to 31 March 2021, randomized clinical trials (RCTs) on probiotics for treating acute diarrhea in children were included. Primary outcomes included the duration of diarrhea and diarrhea lasting ≥2 days, and secondary outcomes included the mean stool frequency on day 2 and duration of hospitalization, fever, and vomiting. We assessed the certainty of the evidence of outcomes according to Grading of Recommendations Assessment, Development, and Evaluation (GRADE) guideline. Eighty-four studies with twenty-one different interventions in 13,443 children were included. For the primary outcomes, moderate evidence indicated that, Lactobacillus reuteri [mean difference (MD) = -0.84 day; 95% confidence interval (CI), -1.39, -0.29], Bifidobacterium lactis (MD = -0.98 day; 95%CI, -1.82, -0.14), Saccharomyces boulardii (MD = -1.25 day; 95%CI, -1.59, -0.91), Lactobacillus species (spp.) plus Bifidobacterium spp. plus Saccharomyces spp. (MD = -1.19 day; 95%CI, -1.81, -0.58), and Bacillus spp. plus Enterococcus spp. plus Clostridium spp. (MD = -1.1 day; 95%CI, -1.84, -0.35) significantly reduced the duration of diarrhea when compared with placebo. Saccharomyces boulardii [Odds ratio (OR) = 0.22; 95%CI, 0.11, 0.41] and Lactobacillus reuteri (OR = 0.23; 95%CI, 0.090, 0.60) significantly reduced the risk of diarrhea lasting ≥2 days when compared with placebo or no treatment, with moderate evidence. Among all probiotics, Saccharomyces boulardii may be the most effective in reducing both duration of diarrhea (compared with placebo) and risk of diarrhea lasting ≥2 days (compared with placebo or no treatment), with moderate evidence. To be conclusive, Saccharomyces boulardii may be the most effective probiotic for treating acute diarrhea in children, followed by several other single-strain and multi-strain probiotics.
Collapse
Affiliation(s)
- Zengbin Li
- China-Australia Joint Research Center for Infectious Diseases, School of Public Health, Xi’an Jiaotong University Health Science Center, Xi’an 710061, China; (Z.L.); (G.Z.); (H.L.)
| | - Guixian Zhu
- China-Australia Joint Research Center for Infectious Diseases, School of Public Health, Xi’an Jiaotong University Health Science Center, Xi’an 710061, China; (Z.L.); (G.Z.); (H.L.)
| | - Chao Li
- Department of Epidemiology and Biostatistics, School of Public Health, Global Health Institute, Xi’an Jiaotong University Health Science Center, Xi’an 710061, China; (C.L.); (X.L.)
| | - Hao Lai
- China-Australia Joint Research Center for Infectious Diseases, School of Public Health, Xi’an Jiaotong University Health Science Center, Xi’an 710061, China; (Z.L.); (G.Z.); (H.L.)
| | - Xin Liu
- Department of Epidemiology and Biostatistics, School of Public Health, Global Health Institute, Xi’an Jiaotong University Health Science Center, Xi’an 710061, China; (C.L.); (X.L.)
| | - Lei Zhang
- China-Australia Joint Research Center for Infectious Diseases, School of Public Health, Xi’an Jiaotong University Health Science Center, Xi’an 710061, China; (Z.L.); (G.Z.); (H.L.)
- Melbourne Sexual Health Centre, Alfred Health, Melbourne, VIC 3053, Australia
- Central Clinical School, Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, VIC 3800, Australia
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University, Zhengzhou 450001, China
| |
Collapse
|
36
|
Abstract
Viral infections represent a major health problem worldwide. Due to the wide variety of etiological agents and their increasing resistance to anti-virals and antibiotics treatments, new strategies for effective therapies need to be developed. Scientific evidence suggests that probiotics may have prophylactic and therapeutic effects in viral diseases. Indeed, these microorganisms interact harmoniously with the intestinal microbiota and protect the integrity of the intestinal barrier as well as modulate the host immune system. Currently, clinical trials with probiotics have been documented in respiratory tract infections, infections caused by human immunodeficiency viruses, herpes, human papillomavirus and hepatic encephalopathy. However, the benefits documented so far are difficult to extrapolate, due to the strain-dependent effect. In addition, the dose of the microorganism used as well as host characteristics are other parameters that should be consider when advocating the use of probiotics to treat viral infections. This review addresses the scientific evidence of the efficacy of probiotics in clinical strains perspective in viral infectious diseases in the last 10 years.
Collapse
|
37
|
Baradaran Ghavami S, Pourhamzeh M, Farmani M, Keshavarz H, Shahrokh S, Shpichka A, Asadzadeh Aghdaei H, Hakemi-Vala M, Hossein-khannazer N, Timashev P, Vosough M. Cross-talk between immune system and microbiota in COVID-19. Expert Rev Gastroenterol Hepatol 2021; 15:1281-1294. [PMID: 34654347 PMCID: PMC8567289 DOI: 10.1080/17474124.2021.1991311] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Accepted: 10/06/2021] [Indexed: 02/08/2023]
Abstract
INTRODUCTION Human gut microbiota plays a crucial role in providing protective responses against pathogens, particularly by regulating immune system homeostasis. There is a reciprocal interaction between the gut and lung microbiota, called the gut-lung axis (GLA). Any alteration in the gut microbiota or their metabolites can cause immune dysregulation, which can impair the antiviral activity of the immune system against respiratory viruses such as severe acute respiratory syndrome coronavirus (SARS-CoV) and SARS-CoV-2. AREAS COVERED This narrative review mainly outlines emerging data on the mechanisms underlying the interactions between the immune system and intestinal microbial dysbiosis, which is caused by an imbalance in the levels of essential metabolites. The authors will also discuss the role of probiotics in restoring the balance of the gut microbiota and modulation of cytokine storm. EXPERT OPINION Microbiota-derived signals regulate the immune system and protect different tissues during severe viral respiratory infections. The GLA's equilibration could help manage the mortality and morbidity rates associated with SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Shaghayegh Baradaran Ghavami
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Thran, Iran
| | - Mahsa Pourhamzeh
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
- Division of Neuroscience, Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Maryam Farmani
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Thran, Iran
| | - Hediye Keshavarz
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Thran, Iran
| | - Shabnam Shahrokh
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Thran, Iran
| | - Anastasia Shpichka
- Institute for Regenerative Medicine, Sechenov First Moscow State Medical University, Moscow, Russia
- World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov First Moscow State Medical University, Moscow, Russia
- Chemistry Department, Lomonosov Moscow State University, Moscow, Russia
| | - Hamid Asadzadeh Aghdaei
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Thran, Iran
| | - Mojdeh Hakemi-Vala
- Department of Microbiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Nikoo Hossein-khannazer
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Thran, Iran
| | - Peter Timashev
- Institute for Regenerative Medicine, Sechenov First Moscow State Medical University, Moscow, Russia
- World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov First Moscow State Medical University, Moscow, Russia
- Chemistry Department, Lomonosov Moscow State University, Moscow, Russia
| | - Massoud Vosough
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| |
Collapse
|
38
|
Tan W, Zhou Z, Li W, Lu H, Qiu Z. Lactobacillus rhamnosus GG for Cow's Milk Allergy in Children: A Systematic Review and Meta-Analysis. Front Pediatr 2021; 9:727127. [PMID: 34746056 PMCID: PMC8569903 DOI: 10.3389/fped.2021.727127] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Accepted: 09/20/2021] [Indexed: 12/13/2022] Open
Abstract
Objective: Cow's milk allergy (CMA) is a common allergic disease. Probiotics have been suggested as a treatment for CMA, with Lactobacillus rhamnosus GG (LGG) being one of the important predominant choices. Despite reports on this topic, the effectiveness of application in CMA remains to be firmly established. Methods: To assess the effects of LGG on CMA in children, the PubMed/Medline, Embase, Cochrane Library, and Web of Science databases were searched for studies on LGG in treatment of CMA, which were published in the English language. Results: Ten studies were finally included. Significantly higher tolerability rates favoring LGG over controls were observed [risk ratio (RR), 2.22; 95% confidence interval (CI), 1.86-2.66; I 2 = 0.00; moderate-quality evidence]. There were no significant differences in SCORAD values favoring LGG over the placebo (mean difference, 1.41; 95% CI, -4.99-7.82; p = 0.67; very low-quality evidence), and LGG may have improved fecal occult blood (risk ratio, 0.36; 95% CI, 0.14-0.92; p = 0.03; low-quality evidence). Conclusion: We found that LGG may have moderate-quality evidence to promote oral tolerance in children with CMA and may facilitate recovery from intestinal symptoms. However, this finding must be treated with caution, and more gpowerful RCTs are needed to evaluate the most effective dose and treatment time for children with CMA. Registration number: CRD42021237221.
Collapse
Affiliation(s)
- Weifu Tan
- Department of Pediatrics, Dongguan Binhaiwan Central Hospital, Jinan University, Dongguan, China
| | - Zhicong Zhou
- Department of Pediatrics, The First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Wei Li
- Department of Pediatrics, Dongguan Binhaiwan Central Hospital, Jinan University, Dongguan, China
| | - Han Lu
- Department of Obstotrics and Gynocology, The First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Zemin Qiu
- Department of Pediatrics, The First Affiliated Hospital, Jinan University, Guangzhou, China
| |
Collapse
|
39
|
Treatment of human intestinal cryptosporidiosis: A review of published clinical trials. INTERNATIONAL JOURNAL FOR PARASITOLOGY-DRUGS AND DRUG RESISTANCE 2021; 17:128-138. [PMID: 34562754 PMCID: PMC8473663 DOI: 10.1016/j.ijpddr.2021.09.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/13/2021] [Revised: 09/06/2021] [Accepted: 09/09/2021] [Indexed: 12/01/2022]
Abstract
The global burden of diarrhea caused by Cryptosporidium parasite is underestimated. In immunocompromised hosts, chronic and severe presentation of intestinal cryptosporidiosis can result in long-term morbidity and high illness costs. The evidence of effective treatments for cryptosporidiosis has been lacking. We reviewed the published clinical trials to bring forward the feasible therapeutic options of human cryptosporidiosis in various populations and settings according to clinical improvement and parasite clearance rates. A total of 42 studies consisting of the use of nitazoxanide, paromomycin, macrolides, somatostatin analogues, letrazuril, albendazole, rifaximin, miltefosine, clofazimine, and colostrum were included in the review. The trials were mostly conducted in small number of individuals infected with human immunodeficiency virus (HIV), and there is inadequate data of controlled trials to suggest the use of these treatment modalities. Nitazoxanide was reported to be highly efficacious only in immunocompetent hosts and was found to be superior to paromomycin in the same group of patients. Macrolides showed no effective results in both clinical and parasitological improvement. Human bovine colostrum should possibly be administered as one of complementary therapeutic modalities along with other antimicrobials to reach optimal parasite eradication. Other trials of therapeutic modalities were terminated due to futility. Currently, available data is intended to aid the development of strategies for improving access to treatments in different clinical settings, as well as to help guide further studies on treatments of human intestinal cryptosporidiosis.
Collapse
|
40
|
Effectiveness of probiotics and synbiotics in reducing duration of acute infectious diarrhea in pediatric patients in developed countries: a systematic review and meta-analysis. Eur J Pediatr 2021; 180:2907-2920. [PMID: 33825068 DOI: 10.1007/s00431-021-04046-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 03/11/2021] [Accepted: 03/21/2021] [Indexed: 02/07/2023]
Abstract
Acute diarrhea is one of the most frequent causes of doctor visits and hospital admissions for children. Our objective was to evaluate the association between probiotics administration and reduction of acute infectious diarrhea duration in children dwelling in developed countries. Bibliographic databases, gray literature, and reference lists were searched up to September 29, 2019. Double-blind, randomized controlled trials that examined probiotics efficacy in children with acute infectious diarrhea residing in developed countries were included. Data were synthesized by generic inverse variance method using fixed- and random-effects model. Twenty trials met the eligibility criteria (n = 3469 patients) and were included in the qualitative synthesis, and 19 studies in meta-analysis. Twelve trials (n = 840) were assessed as high/unclear risk of bias and eight (n = 2629) as low risk of bias. Comparisons revealed a moderate effectiveness of probiotics in low risk of bias studies (MD = - 13.45 h; 95% CI - 24.26, - 2.62; p = 0.02, Bayesian meta-analysis pooled effect MD = - 0.38, 95% CrI - 2.3, 1.58) and a notable effect in studies with high/unclear risk for bias (MD = - 19.70 h; 95% CI - 28.09, - 11.31; p = 0.0004). In trials of optimal methodological quality (n = 1989), probiotics effect was absent (MD = - 3.32 h; 95% CI - 8.78, 2.13, p = 0.23).Conclusion: Outcomes suggest that probiotics do not demonstrate sufficient clinical impact in reducing diarrhea duration in children in the developed countries.Systematic Review Registration: This review is registered at PROSPERO (ID: CRD42020152966). What is Known: • Probiotics, due to the conflicting study results, are administered without adequate evidence as an adjuvant therapeutic agent for eliminating duration of acute infectious diarrhea in pediatric patients. What is New: • In developed countries, probiotics are demonstrated as ineffective in reducing the duration of acute infectious diarrhea in children.
Collapse
|
41
|
Deoxynivalenol exposure induces liver damage in mice: Inflammation and immune responses, oxidative stress, and protective effects of Lactobacillus rhamnosus GG. Food Chem Toxicol 2021; 156:112514. [PMID: 34400200 DOI: 10.1016/j.fct.2021.112514] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 07/28/2021] [Accepted: 08/12/2021] [Indexed: 02/08/2023]
Abstract
Deoxynivalenol (DON), one of the most common environmental pollutants, substantially affects human and animal health. Much attention has been paid to the ability of probiotics to modulate inflammation and immune responses. In this work, the toxic effects of DON on the liver and the protective effects of Lactobacillus rhamnosus GG (LGG) were investigated. We treated mice with oral gavage of DON (2.4 mg/kg bw/day), LGG (1 × 109 CFU/mouse/day) or both for 28 days. The results showed that DON triggered liver inflammation, reflected by pathological changes and liver function damage but LGG oral administration significantly attenuated these changes. Notably, DON treatment activated the TLR4/NF-κB signaling pathway which contribute to produce inflammatory cytokines, but oral administration of LGG inhibited all the effects of DON. DON treatment can also induce oxidative stress and activate Keap1-Nrf2 signaling pathway, leading to the activation of Nrf2 and the downstream genes, while LGG treatment can improve the antioxidant capacity of liver and protected mice from DON injury. In conclusion, LGG was able to negate the detrimental effects of DON on the liver and may contribute as a potential dietary intervention strategy to reduce mycotoxicity.
Collapse
|
42
|
The Interaction Between Viruses and Intestinal Microbiota: A Review. Curr Microbiol 2021; 78:3597-3608. [PMID: 34350485 PMCID: PMC8336530 DOI: 10.1007/s00284-021-02623-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 07/28/2021] [Indexed: 02/07/2023]
Abstract
As the main pathogen threatening human and animal health, viruses can affect the immunity and metabolism of bodies. There are innate microbial barriers in the digestive tract of the body to preserve the homeostasis of the animal body, which directly or indirectly influences the host defence against viral infection. Understanding the interaction between viruses and intestinal microbiota or probiotics is helpful to study the pathogenesis of diseases. Here, we review recent studies on the interaction mechanism between intestinal microbiota and viruses. The interaction can be divided into two aspects: inhibition of viral infection by microbiota and promotion of viral infection by microbiota. The treatment of viral infection by probiotics is summarized.
Collapse
|
43
|
McFarland LV, Srinivasan R, Setty RP, Ganapathy S, Bavdekar A, Mitra M, Raju B, Mohan N. Specific Probiotics for the Treatment of Pediatric Acute Gastroenteritis in India: A Systematic Review and Meta-Analysis. JPGN REPORTS 2021; 2:e079. [PMID: 37205949 PMCID: PMC10191489 DOI: 10.1097/pg9.0000000000000079] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Accepted: 03/08/2021] [Indexed: 05/21/2023]
Abstract
Pediatric acute gastroenteritis (PAGE) is a significant cause of morbidity, mortality and healthcare costs in many countries, but differences in PAGE vary from country-to-country; thus, we limited our analysis to 1 country. Probiotics have been recommended as an adjunct to standard treatment, but the choice of probiotic is unclear. PubMed, Google Scholar, and reviews were searched from inception to May 2020 for randomized controlled trials (RCTs) in India using probiotics for a treatment for PAGE. Meta-analyses using subgroups of identical probiotic types (≥2 RCT/type) were conducted for primary outcomes (duration of diarrhea, cured by day 3, rapidity of response, and length of hospital stay). Twenty-two RCTs were included in the systematic review (N = 4059 participants) including 5 single-strained probiotics and 3 multi-strained mixtures. For the meta-analyses, 17 RCT (20 treatment arms) were included. Saccharomyces boulardii CNCM I-745 had the strongest effect on shortening the duration of diarrhea (standardized mean difference, -1.86 d; 95% confidence interval, -2.8 to -0.9), while both Lactobacillus rhamnosus GG and a mixture of 4 Bacillus clausii strains (O/C, SIN, N/R, T) significantly reduced the duration of diarrhea (-1.7 and -1.4 d, respectively). S. boulardii and L. rhamnosus GG significantly reduced hospital stays (-1.8 and -1.1 d, respectively), while B. clausii had no effect. The frequency of stools/day was significantly reduced by day 4 for S. boulardii and by day 5 for L. rhamnosus GG. In India, 2 types of probiotics (S. boulardii CNCM I-745 and L. rhamnosus GG) significantly shortened both the duration of diarrhea and hospitalization stays in pediatric patients with PAGE. While these 2 probiotic strains were safe and effective for children in India, further research is needed to confirm if other probiotic strains or mixtures may be effective.
Collapse
Affiliation(s)
- Lynne V. McFarland
- From the Department of Medicinal Chemistry, University of Washington, Seattle, WA
| | - Ramesh Srinivasan
- Department of Gastroenterology, Apollo Hospitals, Jubilee Hills, Hyderabad, India
| | - Rajendra P. Setty
- Department of Pediatric Gastroenterology, Hepatology and Nutrition, Pure Bliss Hospital, Panchkula, India
| | | | | | - Monjori Mitra
- Department of Pediatrics, Institute of Child Health, Kolkata, India
| | - Bhaskar Raju
- Department of Gastroenterology, Dr. Mehta’s Children’s Hospital, Chennai, India
| | - Neelam Mohan
- Department of Pediatric Gastroenterology, Medanta Medicity, Gurgaon, India
| |
Collapse
|
44
|
Double-Barrel Shotgun: Probiotic Lactic Acid Bacteria with Antiviral Properties Modified to Serve as Vaccines. Microorganisms 2021; 9:microorganisms9081565. [PMID: 34442644 PMCID: PMC8401918 DOI: 10.3390/microorganisms9081565] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 07/07/2021] [Accepted: 07/08/2021] [Indexed: 12/17/2022] Open
Abstract
Contrary to the general belief that the sole function of probiotics is to keep intestinal microbiota in a balanced state and stimulate the host’s immune response, several studies have shown that certain strains of lactic acid bacteria (LAB) have direct and/or indirect antiviral properties. LAB can stimulate the innate antiviral immune defence system in their host, produce antiviral peptides, and release metabolites that prevent either viral replication or adhesion to cell surfaces. The SARS-CoV (COVID-19) pandemic shifted the world’s interest towards the development of vaccines against viral infections. It is hypothesised that the adherence of SARS-CoV spike proteins to the surface of Bifidobacterium breve could elicit an immune response in its host and trigger the production of antibodies. The question now remains as to whether probiotic LAB could be genetically modified to synthesize viral antigens and serve as vaccines—this concept and the role that LAB play in viral infection are explored in this review.
Collapse
|
45
|
Varsha KK, Maheshwari AP, Nampoothiri KM. Accomplishment of probiotics in human health pertaining to immunoregulation and disease control. Clin Nutr ESPEN 2021; 44:26-37. [PMID: 34330476 DOI: 10.1016/j.clnesp.2021.06.020] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Accepted: 06/23/2021] [Indexed: 12/17/2022]
Abstract
It is a well-established fact that the microbiome harboring the human body plays a critical role in maintaining human health and can influence treatments against various ailments. Human microbiome-based research contemplates the possibility of selecting and administering specific commensal bacterial strains to modulate the gut microbiota to attain favorable outcomes to the therapies. Consumption of probiotics and probiotic-based dietary supplements as functional foods has been a promising treatment strategy against various diseases. Clinical studies demonstrate that probiotic administration alters gut microbiota composition and instigates immune modulation in the host. The benefits of probiotics are reported to be strain-specific and depend on the host's baseline immune competence. This review explores the role of probiotics in alleviating symptoms of allergy, cancer, cardio vascular (CV) diseases, diabetes mellitus (DM), bowel diseases (IBD and IBS), periodontal disease, diseases affecting liver and kidney, neuroinflammatory diseases, and viral infections. Also, it surveyed the broad spectrum bioactive compounds produced by probiotics and possible mechanisms that trigger the immune system.
Collapse
Affiliation(s)
- Kontham Kulangara Varsha
- Microbial Processes and Technology Division (MPTD), CSIR- National Institute for Interdisciplinary Science and Technology (NIIST), Thiruvananthapuram, 695 019, Kerala, India
| | - Arun Padmakumar Maheshwari
- Microbial Processes and Technology Division (MPTD), CSIR- National Institute for Interdisciplinary Science and Technology (NIIST), Thiruvananthapuram, 695 019, Kerala, India
| | - Kesavan Madhavan Nampoothiri
- Microbial Processes and Technology Division (MPTD), CSIR- National Institute for Interdisciplinary Science and Technology (NIIST), Thiruvananthapuram, 695 019, Kerala, India.
| |
Collapse
|
46
|
Probiotics-based foods and beverages as future foods and their overall safety and regulatory claims. FUTURE FOODS 2021. [DOI: 10.1016/j.fufo.2021.100013] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
|
47
|
Abdalla AK, Ayyash MM, Olaimat AN, Osaili TM, Al-Nabulsi AA, Shah NP, Holley R. Exopolysaccharides as Antimicrobial Agents: Mechanism and Spectrum of Activity. Front Microbiol 2021; 12:664395. [PMID: 34093478 PMCID: PMC8170130 DOI: 10.3389/fmicb.2021.664395] [Citation(s) in RCA: 98] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 04/20/2021] [Indexed: 01/16/2023] Open
Abstract
Exopolysaccharides (EPSs) are metabolites synthesized and excreted by a variety of microorganisms, including lactic acid bacteria (LAB). EPS serve several biological functions such as interactions between bacteria and their environments, protection against hostile conditions including dehydration, the alleviation of the action of toxic compounds (bile salts, hydrolyzing enzymes, lysozyme, gastric, and pancreatic enzymes, metal ions, antibiotics), and stresses (changing pH, osmolarity), and evasion of the immune response and phage attack. Bacterial EPSs are considered valuable by the food, pharmaceutical, and nutraceutical industries, owing to their health-promoting benefits and rheological impacts. Numerous studies have reported the unusual antimicrobial activities of various EPS against a wide variety of pathogenic microbes (bacteria, virus, and fungi). This review aims to provide a comprehensive examination of the in vitro and in vivo antimicrobial activities of different EPSs, mainly against foodborne bacterial, fungal, and viral pathogens. The mechanism of EPS action against these pathogens as well as the methods used to measure antimicrobial activities are critically reviewed.
Collapse
Affiliation(s)
| | - Mutamed M. Ayyash
- Department of Food Science, College of Food and Agriculture, United Arab Emirates University (UAEU), Al Ain, United Arab Emirates
| | - Amin N. Olaimat
- Department of Clinical Nutrition and Dietetics, Faculty of Applied Medical Sciences, The Hashemite University, Zarqa, Jordan
| | - Tareq M. Osaili
- Department Clinical Nutrition and Dietetics, University of Sharjah, Sharjah, United Arab Emirates
- Department of Nutrition and Food Technology, Jordan University of Science and Technology, Irbid, Jordan
| | - Anas A. Al-Nabulsi
- Department of Nutrition and Food Technology, Jordan University of Science and Technology, Irbid, Jordan
| | - Nagendra P. Shah
- Food and Nutritional Science, School of Biological Sciences, The University of Hong Kong, Hong Kong, Hong Kong
| | - Richard Holley
- Department of Food and Human Nutritional Sciences, University of Manitoba, Winnipeg, MB, Canada
| |
Collapse
|
48
|
Phillips M, Dunlap BF, Baldridge MT, Karst SM. Enteric Viruses and the Intestinal Microbiota. Virology 2021. [DOI: 10.1002/9781119818526.ch6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
49
|
Su S, Zhang ZF, Wang X, Wang YM, Wang BM. Mechanism of Lactobacillus rhamnosus in treatment of irritable bowel syndrome. Shijie Huaren Xiaohua Zazhi 2021; 29:366-371. [DOI: 10.11569/wcjd.v29.i7.366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Irritable bowel syndrome (IBS) is a clinically common functional gastrointestinal disease, which affects the quality of life of patients. Therefore, it is of great significance to explore effective treatment methods for IBS. Probiotics can improve the symptoms of IBS patients and their quality of life. Lactobacillus rhamnosus is one of the most studied probiotics and has attracted much attention. . Lactobacillus rhamnosus has been used to treat IBS, and much progress has been made in recent years. Lactobacillus rhamnosus can improve the symptoms of IBS by regulating the imbalance of the intestinal flora, protecting the intestinal barrier function, exerting anti-inflammatory activity, regulating the intestinal immunity, improving visceral hypersensitivity, and inhibiting bacteria. This review aims to elucidate the possible mechanism of Lactobacillus rhamnosus in the treatment of IBS.
Collapse
Affiliation(s)
- Shuai Su
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Zhi-Fang Zhang
- Department of Neurology, Tianjin Xiqing Hospital, Tianjin 300380, China
| | - Xin Wang
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Yu-Ming Wang
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Bang-Mao Wang
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin 300052, China
| |
Collapse
|
50
|
Brink LR, Chichlowski M, Pastor N, Thimmasandra Narayanappa A, Shah N. In the Age of Viral Pandemic, Can Ingredients Inspired by Human Milk and Infant Nutrition Be Repurposed to Support the Immune System? Nutrients 2021; 13:870. [PMID: 33800961 PMCID: PMC7999376 DOI: 10.3390/nu13030870] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Revised: 02/27/2021] [Accepted: 03/01/2021] [Indexed: 12/14/2022] Open
Abstract
In 2020, with the advent of a pandemic touching all aspects of global life, there is a renewed interest in nutrition solutions to support the immune system. Infants are vulnerable to infection and breastfeeding has been demonstrated to provide protection. As such, human milk is a great model for sources of functional nutrition ingredients, which may play direct roles in protection against viral diseases. This review aims to summarize the literature around human milk (lactoferrin, milk fat globule membrane, osteopontin, glycerol monolaurate and human milk oligosaccharides) and infant nutrition (polyunsaturated fatty acids, probiotics and postbiotics) inspired ingredients for support against viral infections and the immune system more broadly. We believe that the application of these ingredients can span across all life stages and thus apply to both pediatric and adult nutrition. We highlight the opportunities for further research in this field to help provide tangible nutrition solutions to support one's immune system and fight against infections.
Collapse
Affiliation(s)
- Lauren R. Brink
- Medical and Scientific Affairs, Nutrition, Reckitt Benckiser, Evansville, IN 47721, USA; (M.C.); (N.P.)
| | - Maciej Chichlowski
- Medical and Scientific Affairs, Nutrition, Reckitt Benckiser, Evansville, IN 47721, USA; (M.C.); (N.P.)
| | - Nitida Pastor
- Medical and Scientific Affairs, Nutrition, Reckitt Benckiser, Evansville, IN 47721, USA; (M.C.); (N.P.)
| | | | - Neil Shah
- Medical and Scientific Affairs, Nutrition, Reckitt Benckiser, Slough SL1 3UH, UK;
- University College London, Great Ormond Street, London WC1N 3JH, UK
| |
Collapse
|