1
|
Rani U, Ehrlich J, Fakhri G, Doklaijah M, Stewart T, Berry W, Imdad A. On the Knowledge and Prescription of Probiotics by Pediatric Providers: A Cross-Sectional Study. Nutrients 2025; 17:963. [PMID: 40289941 PMCID: PMC11945007 DOI: 10.3390/nu17060963] [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: 02/09/2025] [Revised: 03/06/2025] [Accepted: 03/07/2025] [Indexed: 04/30/2025] Open
Abstract
OBJECTIVE The therapeutic or prophylactic efficacy and safety of probiotics are not well established. The objective of this study was to assess the knowledge and practice of probiotic use in children among pediatric providers. METHODS This was a cross-sectional study of pediatric providers. A survey was sent to the members of the American Academy of Pediatrics, New York Chapter 1. RESULTS We received 168 responses. Participants were mostly females (70%) and with MD or equivalent education (93%). About 50% of responders did not select the correct definition of probiotics and confused probiotics with prebiotics and synbiotics. About 97% of practitioners were asked about the merits of probiotics by families, and 60% of respondents had prescribed probiotics in their clinical practice. The most common indication for prescription was for treatment of antibiotic-associated diarrhea. When asked about their recommendation for a family who had already started probiotics, 66% of the providers recommended continuing the probiotics. There was a significant association between the frequency of probiotics prescription and the type of practice (p < 0.05). However, this association disappeared after adjusting for age, gender, education, and years of practice. The more experienced the practitioner, the lower the odds were of prescribing probiotics (p < 0.05). CONCLUSIONS There was inadequate knowledge about probiotics among general pediatric providers. Of the pediatricians asked about probiotics, most recommended continuing them if a family was using probiotics for a specific condition. Studies with a larger nationally representative sample are required for future research.
Collapse
Affiliation(s)
- Uzma Rani
- State University of New York Upstate Medical University, Syracuse, NY 13210, USA; (U.R.); (J.E.); (G.F.); (M.D.); (T.S.); (W.B.)
- Division of General Pediatrics and Stead Family, Department of Pediatrics, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Julie Ehrlich
- State University of New York Upstate Medical University, Syracuse, NY 13210, USA; (U.R.); (J.E.); (G.F.); (M.D.); (T.S.); (W.B.)
- Department of Pediatrics, The University of Vermont Medical Center, Burlington, VT 05401, USA
| | - Ghina Fakhri
- State University of New York Upstate Medical University, Syracuse, NY 13210, USA; (U.R.); (J.E.); (G.F.); (M.D.); (T.S.); (W.B.)
| | - Mohammed Doklaijah
- State University of New York Upstate Medical University, Syracuse, NY 13210, USA; (U.R.); (J.E.); (G.F.); (M.D.); (T.S.); (W.B.)
| | - Telisa Stewart
- State University of New York Upstate Medical University, Syracuse, NY 13210, USA; (U.R.); (J.E.); (G.F.); (M.D.); (T.S.); (W.B.)
| | - Winter Berry
- State University of New York Upstate Medical University, Syracuse, NY 13210, USA; (U.R.); (J.E.); (G.F.); (M.D.); (T.S.); (W.B.)
| | - Aamer Imdad
- Division of Gastroenterology, Hepatology, Pancreatology, and Nutrition, Stead Family Department of Pediatrics, University of Iowa, Iowa City, IA 52242, USA
| |
Collapse
|
2
|
Kenneth MJ, Wu CC, Fang CY, Hsu TK, Lin IC, Huang SW, Chiu YC, Hsu BM. Exploring the Impact of Chemotherapy on the Emergence of Antibiotic Resistance in the Gut Microbiota of Colorectal Cancer Patients. Antibiotics (Basel) 2025; 14:264. [PMID: 40149075 PMCID: PMC11939702 DOI: 10.3390/antibiotics14030264] [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: 01/13/2025] [Revised: 02/21/2025] [Accepted: 02/28/2025] [Indexed: 03/29/2025] Open
Abstract
With nearly half of colorectal cancer (CRC) patients diagnosed at advanced stages where surgery alone is insufficient, chemotherapy remains a cornerstone for this cancer treatment. To prevent infections and improve outcomes, antibiotics are often co-administered. However, chemotherapeutic interactions with the gut microbiota cause significant non-selective toxicity, affecting not only tumor and normal epithelial cells but also the gut microbiota. This toxicity triggers the bacterial SOS response and loss of microbial diversity, leading to bacterial mutations and dysbiosis. Consequently, pathogenic overgrowth and systemic infections increase, necessitating broad-spectrum antibiotics intervention. This review underscores how prolonged antibiotic use during chemotherapy, combined with chemotherapy-induced bacterial mutations, creates selective pressures that drive de novo antimicrobial resistance (AMR), allowing resistant bacteria to dominate the gut. This compromises the treatment efficacy and elevates the mortality risk. Restoring gut microbial diversity may mitigate chemotherapy-induced toxicity and improve therapeutic outcomes, and emerging strategies, such as fecal microbiota transplantation (FMT), probiotics, and prebiotics, show considerable promise. Given the global threat posed by antibiotic resistance to cancer treatment, prioritizing antimicrobial stewardship is essential for optimizing antibiotic use and preventing resistance in CRC patients undergoing chemotherapy. Future research should aim to minimize chemotherapy's impact on the gut microbiota and develop targeted interventions to restore microbial diversity affected during chemotherapy.
Collapse
Affiliation(s)
- Mutebi John Kenneth
- Department of Earth and Environmental Sciences, National Chung Cheng University, Chiayi 621, Taiwan
- Doctoral Program in Science, Technology, Environment and Mathematics, National Chung Cheng University, Chiayi 621, Taiwan
| | - Chin-Chia Wu
- Division of Colorectal Surgery, Dalin Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Chiayi 622, Taiwan
- College of Medicine, Tzu Chi University, Hualien 970, Taiwan
- School of Post-Baccalaureate Chinese Medicine, Tzu Chi University, Hualien 970, Taiwan
| | - Chuan-Yin Fang
- Division of Colon and Rectal Surgery, Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chiayi 600, Taiwan
| | - Tsui-Kang Hsu
- Department of Ophthalmology, Cheng Hsin General Hospital, Taipei 112, Taiwan
- School of Medicine, National Yang Ming Chiao Tung University, Hsinchu 300, Taiwan
| | - I-Ching Lin
- Department of Family Medicine, Asia University Hospital, Taichung 413, Taiwan
- Department of Kinesiology, Health and Leisure, Chienkuo Technology University, Changhua 500, Taiwan
| | - Shih-Wei Huang
- Center for Environmental Toxin and Emerging Contaminant Research, Cheng Shiu University, Kaohsiung 833, Taiwan
| | - Yi-Chou Chiu
- General Surgery, Surgical Department, Cheng Hsin General Hospital, Taipei 112, Taiwan
| | - Bing-Mu Hsu
- Department of Earth and Environmental Sciences, National Chung Cheng University, Chiayi 621, Taiwan
| |
Collapse
|
3
|
Kerek Á, Román I, Szabó Á, Pézsa NP, Jerzsele Á. Antibiotic Resistance Gene Expression in Veterinary Probiotics: Two Sides of the Coin. Vet Sci 2025; 12:217. [PMID: 40266902 PMCID: PMC11945515 DOI: 10.3390/vetsci12030217] [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: 01/26/2025] [Revised: 02/21/2025] [Accepted: 02/26/2025] [Indexed: 04/25/2025] Open
Abstract
The rapid proliferation of antimicrobial resistance has emerged as one of the most pressing animal and public health challenges of our time. Probiotics, extensively employed in human and veterinary medicine, are instrumental in maintaining a balanced microbiome and mitigating its disruption during antibiotic therapy. While their numerous benefits are well documented, probiotics also present potential risks, notably the capacity to harbor antimicrobial resistance genes. This genetic reservoir could contribute to the emergence and spread of antimicrobial resistance by facilitating the horizontal transfer of resistance genes to pathogenic bacteria within the gut. This review critically examines the presence of antimicrobial resistance genes in commonly used probiotic strains, explores the underlying mechanisms of resistance, and provides a balanced analysis of the benefits and risks associated with their use. By addressing these dual aspects, this paper highlights the need for vigilant evaluation of probiotics to preserve their therapeutic potential while minimizing public health risks.
Collapse
Affiliation(s)
- Ádám Kerek
- Department of Pharmacology and Toxicology, University of Veterinary Medicine, István utca 2, H-1078 Budapest, Hungary; (I.R.); (Á.S.); (N.P.P.); (Á.J.)
- National Laboratory of Infectious Animal Diseases, Antimicrobial Resistance, Veterinary Public Health and Food Chain Safety, University of Veterinary Medicine, István utca 2, H-1078 Budapest, Hungary
| | - István Román
- Department of Pharmacology and Toxicology, University of Veterinary Medicine, István utca 2, H-1078 Budapest, Hungary; (I.R.); (Á.S.); (N.P.P.); (Á.J.)
| | - Ábel Szabó
- Department of Pharmacology and Toxicology, University of Veterinary Medicine, István utca 2, H-1078 Budapest, Hungary; (I.R.); (Á.S.); (N.P.P.); (Á.J.)
| | - Nikolett Palkovicsné Pézsa
- Department of Pharmacology and Toxicology, University of Veterinary Medicine, István utca 2, H-1078 Budapest, Hungary; (I.R.); (Á.S.); (N.P.P.); (Á.J.)
| | - Ákos Jerzsele
- Department of Pharmacology and Toxicology, University of Veterinary Medicine, István utca 2, H-1078 Budapest, Hungary; (I.R.); (Á.S.); (N.P.P.); (Á.J.)
- National Laboratory of Infectious Animal Diseases, Antimicrobial Resistance, Veterinary Public Health and Food Chain Safety, University of Veterinary Medicine, István utca 2, H-1078 Budapest, Hungary
| |
Collapse
|
4
|
Zahedifard Z, Mahmoodi S, Ghasemian A. Genetically Engineered Bacteria as a Promising Therapeutic Strategy Against Cancer: A Comprehensive Review. Biotechnol Appl Biochem 2025. [PMID: 39985148 DOI: 10.1002/bab.2738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Accepted: 02/06/2025] [Indexed: 02/24/2025]
Abstract
As a significant cause of global mortality, the cancer has also economic impacts. In the era of cancer therapy, mitigating side effects and costs and overcoming drug resistance is crucial. Microbial species can grow inside the tumor microenvironment and inhibit cancer growth through direct killing of tumor cells and immunoregulatory effects. Although microbiota or their products have demonstrated anticancer effects, the possibility of acting as pathogens and exerting side effects in certain individuals is a risk. Hence, several genetically modified/engineered bacteria (GEB) have been developed to this aim with ability of diagnosing and selective targeting and destruction of cancers. Additionally, GEB are expected to be considerably more efficient, safer, more permeable, less costly, and less invasive theranostic approaches compared to wild types. Potential GEB strains such as Escherichia coli (Nissle 1917, and MG1655), Salmonella typhimurium YB1 SL7207 (aroA gene deletion), VNP20009 (∆msbB/∆purI) and ΔppGpp (PTet and PBAD), and Listeria monocytogenes Lmat-LLO have been developed to combat cancer cells. When used in tandem with conventional treatments, GEB substantially improve the efficacy of anticancer therapy outcomes. In addition, public acceptance, optimal timing (s), duration (s), dose (s), and strains identification, interactions with other strains and the host cells, efficacy, safety and quality, and potential risks and ethical dilemmas include major challenges.
Collapse
Affiliation(s)
- Zahra Zahedifard
- Department of Medical Biotechnology, School of Medicine, Fasa University of Medical Sciences, Fasa, Iran
| | - Shirin Mahmoodi
- Department of Medical Biotechnology, School of Medicine, Fasa University of Medical Sciences, Fasa, Iran
| | - Abdolmajid Ghasemian
- Noncommunicable Diseases Research Center, Fasa University of Medical Sciences, Fasa, Iran
| |
Collapse
|
5
|
Slykerman RF, Davies N, Donohoe R. Evaluating the scientific evidence to support mental health and well-being claims made on probiotic products. Nutr Health 2025:2601060241305682. [PMID: 39828955 DOI: 10.1177/02601060241305682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
BACKGROUND Increasing interest in probiotics for mental health has led to products marketed to consumers claiming psychological benefits. This study aimed to identify mental health claims on probiotic supplements and to provide a narrative review of the scientific evidence to support these claims. METHODS Probiotic supplements with mental health claims on the product label were identified. A search of the scientific literature was performed using Google Scholar, PubMed and Cochrane Central Register of Controlled Trials. Studies were included if the primary outcome was mental health or a biomarker of mental health, if they investigated the exact probiotic strain or combination of strains as present in the product, and if the study was a trial of probiotics conducted in either humans or animals. Included studies were assessed for risk of bias. RESULTS Fourteen probiotic products with mental health claims were identified containing five different strains or strain combinations. Eighteen studies relevant to the products and investigating mental health outcomes met inclusion criteria. Ten studies investigated the same combination of probiotics and provided evidence to support the claims made. Statistically significant results were not universally present for all products. CONCLUSION The amount and quality of evidence to support claims of mental health benefits made on probiotic products is variable. The results support the need for a grading system to facilitate informed consumer decision-making when choosing a probiotic supplement to support mental health.
Collapse
Affiliation(s)
- Rebecca F Slykerman
- Department of Psychological Medicine, The University of Auckland, Auckland, New Zealand
| | - Naomi Davies
- Department of Psychological Medicine, The University of Auckland, Auckland, New Zealand
| | - Robert Donohoe
- Department of Psychological Medicine, The University of Auckland, Auckland, New Zealand
| |
Collapse
|
6
|
Almabruk BA, Bajafar AA, Mohamed AN, Al-Zahrani SA, Albishi NM, Aljarwan R, Aljaser RA, Alghamdi LI, Almutairi TS, Alsolami AS, Alghamdi JK. Efficacy of Probiotics in the Management of Irritable Bowel Syndrome: A Systematic Review and Meta-Analysis. Cureus 2024; 16:e75954. [PMID: 39830577 PMCID: PMC11741147 DOI: 10.7759/cureus.75954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/18/2024] [Indexed: 01/22/2025] Open
Abstract
Irritable bowel syndrome (IBS) significantly impacts quality of life. Probiotics offer relief by modulating gut microbiota, but variability in outcomes necessitates a systematic evaluation of their efficacy. This study aims to evaluate the efficacy of probiotics in improving symptoms of IBS through a systematic review and meta-analysis. A comprehensive search of PubMed and Google Scholar identified studies published between 2014 and 2018. Inclusion criteria focused on randomized controlled trials evaluating probiotics in adult IBS patients diagnosed using standardized criteria. Statistical analysis utilized random effects models to account for heterogeneity, with subgroup analysis performed for IBS subtypes. This review included 23 studies involving 3,288 participants. Probiotics significantly reduced abdominal pain (mean difference = -1.66, 95% CI = -2.39 to -0.93, p < 0.0001) and bloating (mean difference = -2.13, 95% CI = -3.96 to -0.30, p = 0.0224). Improvement in stool habits was significant (mean difference = -1.52, 95% CI = -2.15 to -0.88, p < 0.0001), particularly in diarrhea-predominant IBS. Quality of life improved significantly, with a mean increase of 8.77 points (95% CI = 0.91 to 16.64, p = 0.028). Adverse events were mild and infrequent. However, heterogeneity was high (I² > 90%), reflecting variability in study protocols. Probiotics are effective in reducing IBS symptoms and improving quality of life, mainly in diarrhea-predominant IBS. More research should be conducted that focuses on standardized, long-term trials to refine treatment strategies.
Collapse
Affiliation(s)
| | - Ali A Bajafar
- General Practice, King Abdullah Medical Complex, Jeddah, SAU
| | - Ahmed N Mohamed
- Critical Care Medicine, SEHA Salma Rehabilitation Hospital, Abu Dhabi, ARE
| | - Saleh A Al-Zahrani
- College of Medicine, King Saud Bin Abdulaziz University for Health Sciences, Jeddah, SAU
| | - Naif M Albishi
- Nursing, Imam Abdulrahman Bin Faisal University, Dammam, SAU
| | - Rafeef Aljarwan
- College of Medicine, King Saud Bin Abdulaziz University for Health Sciences, Riyadh, SAU
| | | | | | | | - Almas S Alsolami
- College of Applied Medical Sciences, King Saud Bin Abdulaziz University for Health Sciences, Jeddah, SAU
| | - Joud K Alghamdi
- Faculty of Medicine, King Abdulaziz University Hospital, Jeddah, SAU
| |
Collapse
|
7
|
Anderson JL, Tierney AC, Miles C, Kotsimbos T, King SJ. Probiotic knowledge of adults with cystic fibrosis is limited but is associated with probiotic use: A cross-sectional survey study. Nutr Health 2024; 30:697-706. [PMID: 36366805 DOI: 10.1177/02601060221136653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Background: Probiotics are used by people with cystic fibrosis (CF) and other chronic diseases to manage gastrointestinal symptoms. Aim: To describe probiotic knowledge; its relationship with probiotic use, probiotic information sources and factors influencing choice in adults with CF and a general population control group. Methods: A cross-sectional questionnaire study was conducted in adults with CF (n = 205) and Controls (n = 158). Probiotic knowledge was compared between CF and Controls using a knowledge score (maximum 5) based on predefined criteria: (1a) bacteria/microorganism; (1b) live; (2a) administered; (2b) adequate dose and (3) health benefit, using independent samples t-test. Two-way analysis of variance explored knowledge scores between CF and Control and between Ever User and Never User groups. Chi-square and Fisher's exact tests compared knowledge criterion, probiotic sources and influences on probiotic choice between groups. Thematic analysis of open-text responses explored probiotic-related knowledge and influences on probiotic decision making. Results: Knowledge scores (mean ± SD) did not differ between CF (1.70 ± 1.12) and Controls (1.89 ± 0.99), p = 0.13. Probiotic use was associated with knowledge score (p < 0.001). More CF Ever Users than Never Users correctly identified criteria 1a (65% vs. 38%), 1b (16% vs. 0%), 2a (45% vs. 22%) and 3 (73% vs. 42%) (all p < 0.005). CF participants considered 'dairy yoghurt' (69%), 'live cultures' (64%) and 'fermented foods' (37%) as 'all/mostly' probiotic sources. The internet was the commonest source of probiotic-related information. Conclusion: Probiotic knowledge and use were associated in adults with CF. Understanding of probiotic characteristics and sources were limited. Education is needed to help guide patient probiotic decision making.
Collapse
Affiliation(s)
- Jacqueline L Anderson
- Discipline of Food, Nutrition and Dietetics, School of Allied Health, La Trobe University, Bundoora, Australia
| | - Audrey C Tierney
- Discipline of Food, Nutrition and Dietetics, School of Allied Health, La Trobe University, Bundoora, Australia
- School of Allied Health, Health Implementation Science and Technology Centre, Health Research Institute, University of Limerick, Limerick, Ireland
| | - Caitlin Miles
- Nutrition and Dietetics Department, Monash Health, Clayton, Australia
| | - Tom Kotsimbos
- Cystic Fibrosis Service, Department of Allergy, Immunology and Respiratory Medicine, Alfred Health, Melbourne, Australia
- Department of Medicine, Monash University, Melbourne, Australia
| | - Susannah J King
- Discipline of Food, Nutrition and Dietetics, School of Allied Health, La Trobe University, Bundoora, Australia
- Nutrition Department, Alfred Health, Melbourne, Australia
| |
Collapse
|
8
|
Martinez Guevara D, Vidal Cañas S, Palacios I, Gómez A, Estrada M, Gallego J, Liscano Y. Effectiveness of Probiotics, Prebiotics, and Synbiotics in Managing Insulin Resistance and Hormonal Imbalance in Women with Polycystic Ovary Syndrome (PCOS): A Systematic Review of Randomized Clinical Trials. Nutrients 2024; 16:3916. [PMID: 39599701 PMCID: PMC11597640 DOI: 10.3390/nu16223916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 11/12/2024] [Accepted: 11/14/2024] [Indexed: 11/29/2024] Open
Abstract
Background/Objectives: Polycystic ovary syndrome is a common endocrine disorder in women of reproductive age characterized by insulin resistance and hormonal imbalances. Recent research suggests that probiotics and synbiotics may improve these parameters by modulating the gut microbiota. This study systematically reviewed randomized clinical trials evaluating the impact of probiotic, prebiotic, and synbiotic supplementation on insulin resistance and hormonal parameters in women with PCOS. Methods: Exhaustive searches were conducted in PubMed, Cochrane CENTRAL, Scopus, Web of Science, and Embase, following PRISMA guidelines. Randomized trials assessing supplementation with probiotics, prebiotics, or synbiotics for at least 8 weeks in women diagnosed with PCOS according to the Rotterdam criteria were included. Data on participants, interventions, and outcomes related to insulin resistance and hormones were extracted. Results: Eleven studies from Iran involving overweight or obese women aged 15 to 48 were included. Probiotic and synbiotic supplementation showed significant improvements in insulin resistance (reductions in HOMA-IR, fasting glucose, and insulin), lipid profiles (decreased LDL and triglycerides; increased HDL), and hormonal balance (increased SHBG, decreased total testosterone). Synbiotics had more pronounced effects than probiotics or prebiotics alone. Adherence was high, and side effects were minimal. Conclusions: Despite promising results, limitations such as small sample sizes, homogeneous populations, and short intervention durations limit the generalization of the findings. Larger, longer, multicenter trials with diverse populations and standardized methodologies are needed to confirm the efficacy and safety of synbiotics in managing PCOS. Integrating these interventions could improve clinical management and quality of life for affected women, but additional evidence is required to support widespread use.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Yamil Liscano
- Grupo de Investigación en Salud Integral (GISI), Departamento Facultad de Salud, Universidad Santiago de Cali, Cali 5183000, Colombia; (D.M.G.); (S.V.C.); (I.P.); (A.G.); (M.E.); (J.G.)
| |
Collapse
|
9
|
Lopes CM, de Jesus Monteiro CS, Duarte AP, dos Santos JL. Probiotics and Prebiotics for the Treatment of Irritable Bowel Syndrome-A Narrative Review. J Clin Med 2024; 13:6337. [PMID: 39518476 PMCID: PMC11546470 DOI: 10.3390/jcm13216337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 10/17/2024] [Accepted: 10/21/2024] [Indexed: 11/16/2024] Open
Abstract
Background/Objectives: Gastrointestinal functional disorders (GFDs), including irritable bowel syndrome (IBS), are imbalances in the gut-brain axis characterized by persistence of symptoms in the abdominal area. Probiotics are live microorganisms that provide benefits to the health of their hosts when administered in adequate amounts, while prebiotics are a substrate that is selectively used by host microorganisms. This narrative review aimed to evaluate the effectiveness of prebiotics and probiotics mostly in irritable bowel syndrome, particularly on issues such as the interaction between these products and the gut microbiota, the duration of supplementation and long-term effects, the definition of ideal dosages, and the regulation and quality control of these products. Methods: A bibliographic search was carried out in indexed databases and articles published within 10 years before the beginning of the study and publications in English language, which investigated the specific theme of the study were considered. Papers dealing with topics not covered by the research questions, or presenting errors related with the wrong population or the wrong methods, as well as experimental studies and case reviews were excluded. Fifty-five articles were selected, initially in isolation by the authors and, afterward, under consensus. Results: It was possible to observe the effectiveness mainly of probiotics, in improving specific symptoms of the respective disorder; however, the available data remain unclear due to limitations concerning samples and methods of the studies evaluated. Conclusions: Despite evidence suggestive of therapeutic efficacy, additional multicenter randomized controlled trials (RCTs) with better defined protocols are still necessary to fill in the gaps in this subject, define measures to ensure the safe administration of these products, and confirm their therapeutic potential.
Collapse
Affiliation(s)
- Carolina Marques Lopes
- Faculdade de Ciências da Saúde, Universidade da Beira Interior, 6200-506 Covilhã, Portugal; (C.M.L.); (C.S.d.J.M.); (A.P.D.)
- CICS-UBI-Health Sciences Research Centre, Faculdade de Ciências da Saúde, Universidade da Beira Interior, 6200-506 Covilhã, Portugal
- Academic Clinical Center of Beiras, Faculdade de Ciências da Saúde, Universidade da Beira Interior, 6200-506 Covilhã, Portugal
| | - Cristina Sofia de Jesus Monteiro
- Faculdade de Ciências da Saúde, Universidade da Beira Interior, 6200-506 Covilhã, Portugal; (C.M.L.); (C.S.d.J.M.); (A.P.D.)
- CICS-UBI-Health Sciences Research Centre, Faculdade de Ciências da Saúde, Universidade da Beira Interior, 6200-506 Covilhã, Portugal
- Academic Clinical Center of Beiras, Faculdade de Ciências da Saúde, Universidade da Beira Interior, 6200-506 Covilhã, Portugal
- UFBI—Pharmacovigilance Unit of Beira Interior, Faculdade de Ciências da Saúde, Universidade da Beira Interior, 6200-506 Covilhã, Portugal
| | - Ana Paula Duarte
- Faculdade de Ciências da Saúde, Universidade da Beira Interior, 6200-506 Covilhã, Portugal; (C.M.L.); (C.S.d.J.M.); (A.P.D.)
- CICS-UBI-Health Sciences Research Centre, Faculdade de Ciências da Saúde, Universidade da Beira Interior, 6200-506 Covilhã, Portugal
- Academic Clinical Center of Beiras, Faculdade de Ciências da Saúde, Universidade da Beira Interior, 6200-506 Covilhã, Portugal
- UFBI—Pharmacovigilance Unit of Beira Interior, Faculdade de Ciências da Saúde, Universidade da Beira Interior, 6200-506 Covilhã, Portugal
| | - Jorge Luiz dos Santos
- Faculdade de Ciências da Saúde, Universidade da Beira Interior, 6200-506 Covilhã, Portugal; (C.M.L.); (C.S.d.J.M.); (A.P.D.)
- CICS-UBI-Health Sciences Research Centre, Faculdade de Ciências da Saúde, Universidade da Beira Interior, 6200-506 Covilhã, Portugal
- Academic Clinical Center of Beiras, Faculdade de Ciências da Saúde, Universidade da Beira Interior, 6200-506 Covilhã, Portugal
- UFBI—Pharmacovigilance Unit of Beira Interior, Faculdade de Ciências da Saúde, Universidade da Beira Interior, 6200-506 Covilhã, Portugal
| |
Collapse
|
10
|
Loy MH. From plate to planet: culturally responsive culinary practices for health system innovation. Front Nutr 2024; 11:1476503. [PMID: 39483786 PMCID: PMC11525790 DOI: 10.3389/fnut.2024.1476503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Accepted: 09/30/2024] [Indexed: 11/03/2024] Open
Abstract
The field of culinary medicine has gained significant attention for its potential to improve health outcomes through the integration of nutrition and medical practice. However, the cultural dimensions of this interdisciplinary field remain underexplored. Emphasizing the role of sociocultural practices, the paper highlights how culturally appreciative culinary practices can meet the sextuple aim of healthcare system innovation. By examining diverse cultural traditions and their contributions to culinary medicine, this review underscores the importance of culturally attuned approaches in promoting human health. The integration of cultural food wisdom into healthcare practices offers a pathway to more effective and personalized care, stronger patient-provider relationships, diversity/equity/inclusion/belonging, and sustainable food systems.
Collapse
Affiliation(s)
- Michelle H. Loy
- Department of Medicine and Pediatrics, Weill Cornell Medicine, Cornell University, New York, NY, United States
| |
Collapse
|
11
|
da Silva TF, Glória RDA, Americo MF, Freitas ADS, de Jesus LCL, Barroso FAL, Laguna JG, Coelho-Rocha ND, Tavares LM, le Loir Y, Jan G, Guédon É, Azevedo VADC. Unlocking the Potential of Probiotics: A Comprehensive Review on Research, Production, and Regulation of Probiotics. Probiotics Antimicrob Proteins 2024; 16:1687-1723. [PMID: 38539008 DOI: 10.1007/s12602-024-10247-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/12/2024] [Indexed: 10/02/2024]
Abstract
This review provides a comprehensive overview of the current state of probiotic research, covering a wide range of topics, including strain identification, functional characterization, preclinical and clinical evaluations, mechanisms of action, therapeutic applications, manufacturing considerations, and future directions. The screening process for potential probiotics involves phenotypic and genomic analysis to identify strains with health-promoting properties while excluding those with any factor that could be harmful to the host. In vitro assays for evaluating probiotic traits such as acid tolerance, bile metabolism, adhesion properties, and antimicrobial effects are described. The review highlights promising findings from in vivo studies on probiotic mitigation of inflammatory bowel diseases, chemotherapy-induced mucositis, dysbiosis, obesity, diabetes, and bone health, primarily through immunomodulation and modulation of the local microbiota in human and animal models. Clinical studies demonstrating beneficial modulation of metabolic diseases and human central nervous system function are also presented. Manufacturing processes significantly impact the growth, viability, and properties of probiotics, and the composition of the product matrix and supplementation with prebiotics or other strains can modify their effects. The lack of regulatory oversight raises concerns about the quality, safety, and labeling accuracy of commercial probiotics, particularly for vulnerable populations. Advancements in multi-omics approaches, especially probiogenomics, will provide a deeper understanding of the mechanisms behind probiotic functionality, allowing for personalized and targeted probiotic therapies. However, it is crucial to simultaneously focus on improving manufacturing practices, implementing quality control standards, and establishing regulatory oversight to ensure the safety and efficacy of probiotic products in the face of increasing therapeutic applications.
Collapse
Affiliation(s)
- Tales Fernando da Silva
- Institute of Biological Sciences, Department of Genetics, Ecology, and Evolution, Federal University of Minas Gerais, Belo Horizonte, Brazil
- UMR1253, INRAE, L'Institut Agro Rennes Angers, STLO, Rennes, France
| | - Rafael de Assis Glória
- Institute of Biological Sciences, Department of Genetics, Ecology, and Evolution, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Monique Ferrary Americo
- Institute of Biological Sciences, Department of Genetics, Ecology, and Evolution, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Andria Dos Santos Freitas
- Institute of Biological Sciences, Department of Genetics, Ecology, and Evolution, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Luis Claudio Lima de Jesus
- Institute of Biological Sciences, Department of Genetics, Ecology, and Evolution, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Fernanda Alvarenga Lima Barroso
- Institute of Biological Sciences, Department of Genetics, Ecology, and Evolution, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Juliana Guimarães Laguna
- Institute of Biological Sciences, Department of Genetics, Ecology, and Evolution, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Nina Dias Coelho-Rocha
- Institute of Biological Sciences, Department of Genetics, Ecology, and Evolution, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Laisa Macedo Tavares
- Institute of Biological Sciences, Department of Genetics, Ecology, and Evolution, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Yves le Loir
- UMR1253, INRAE, L'Institut Agro Rennes Angers, STLO, Rennes, France
| | - Gwénaël Jan
- UMR1253, INRAE, L'Institut Agro Rennes Angers, STLO, Rennes, France
| | - Éric Guédon
- UMR1253, INRAE, L'Institut Agro Rennes Angers, STLO, Rennes, France
| | - Vasco Ariston de Carvalho Azevedo
- Institute of Biological Sciences, Department of Genetics, Ecology, and Evolution, Federal University of Minas Gerais, Belo Horizonte, Brazil.
| |
Collapse
|
12
|
Campos LL, Oliveira SRM, Amaral MNS, Gallotti B, Oliveira AF, Arantes RME, Ribeiro-Souza S, Vital KD, Fernandes SOA, Cardoso VN, Nicoli JR, Martins FS. Oral Treatment with Saccharomyces cerevisiae CNCM I-3856 Mitigates the Inflammatory Response Experimentally Induced by Salmonella enterica subsp. enterica Serovar Typhimurium in Mice. Probiotics Antimicrob Proteins 2024:10.1007/s12602-024-10359-4. [PMID: 39243351 DOI: 10.1007/s12602-024-10359-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/03/2024] [Indexed: 09/09/2024]
Abstract
Salmonella spp. are intracellular, Gram-negative pathogens responsible for a range of diarrheal diseases, which can present either as self-limited (gastroenteritis) or as a systemic form (typhoid fever), characterizing a serious public health problem. In this study, we investigated the therapeutic effects of oral administration of Saccharomyces cerevisiae CNCM I-3856 in a murine model infected with Salmonella Typhimurium (ST). This yeast species has previously demonstrated the potential to support immune function and reduce inflammation and the ability to exert antimicrobial activity, which is important considering the increasing prevalence of antibiotic-resistant bacteria. Our findings revealed that mice infected with ST and only treated with sterile saline exhibited a higher mortality rate and body weight loss. In contrast, mice treated with I-3856 showed a notable reduction in these adverse outcomes. The yeast demonstrated a high capacity for co-aggregation with the pathogen. Furthermore, the significant amounts of yeast found in the feces of treated mice suggest that intestinal colonization was effective, which was associated with several beneficial effects, including reduced intestinal permeability, which likely limits bacterial translocation to extraintestinal organs. Additionally, the administration of I-3856 reduced levels of sIgA and resulted in a decrease in the recruitment of neutrophils and eosinophils to infection sites, indicating a modulation of the inflammatory response. Histological analyses showed attenuated liver and intestinal lesions in the yeast-treated mice, corroborating the protective effects of the yeast. In conclusion, the results suggest that S. cerevisiae CNCM I-3856 has the potential to control the inflammatory response experimentally induced by S. Typhimurium when administered to mice.
Collapse
Affiliation(s)
- Lara L Campos
- Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Samantha R M Oliveira
- Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Maisa N S Amaral
- Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Bruno Gallotti
- Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Aline F Oliveira
- Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Rosa M E Arantes
- Departamento de Patologia Geral, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Samantha Ribeiro-Souza
- Departamento de Ciências Biológicas, Universidade Federal de Ouro Preto, Ouro Preto, MG, Brazil
| | - Katia D Vital
- Departamento de Análises Clínicas E Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Simone O A Fernandes
- Departamento de Análises Clínicas E Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Valbert N Cardoso
- Departamento de Análises Clínicas E Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Jacques R Nicoli
- Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Flaviano S Martins
- Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil.
| |
Collapse
|
13
|
Stefanaki C, Rozou P, Efthymiou V, Xinias I, Mastorakos G, Bacopoulou F, Papagianni M. Impact of Probiotics on the Glycemic Control of Pediatric and Adolescent Individuals with Type 1 Diabetes: A Systematic Review and Meta-Analysis. Nutrients 2024; 16:2629. [PMID: 39203766 PMCID: PMC11357215 DOI: 10.3390/nu16162629] [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: 07/07/2024] [Revised: 08/03/2024] [Accepted: 08/05/2024] [Indexed: 09/03/2024] Open
Abstract
AIMS Human recombinant insulin is currently the only therapy for children and adolescents with type 1 diabetes (T1D), although not always efficient for the glycemic control of these individuals. The interrelation between the gut microbiome and the glycemic control of apparently healthy populations, as well as various populations with diabetes, is undeniable. Probiotics are biotherapeutics that deliver active components to various targets, primarily the gastrointestinal tract. This systematic review and meta-analysis examined the effect of the administration of probiotics on the glycemic control of pediatric and adolescent individuals with T1D. MATERIALS AND METHODS Randomized controlled trials employing the administration of probiotics in children and adolescents with T1D (with ≥10 individuals per treatment arm), written in English, providing parameters of glycemic control, such as mean glucose concentrations and glycosylated hemoglobin (HbA1c), were deemed eligible. RESULTS The search strategy resulted in six papers with contradictory findings. Ultimately, five studies of acceptable quality, comprising 388 children and adolescents with T1D, were included in the meta-analysis. Employing a random and fixed effects model revealed statistically significant negative effect sizes of probiotics on the glycemic control of those individuals, i.e., higher concentrations of glucose and HbA1c than controls. CONCLUSIONS Children and adolescents with T1D who received probiotics demonstrated worse glycemic control than controls after the intervention. Adequately powered studies, with extended follow-up periods, along with monitoring of compliance and employing the proper strains, are required to unravel the mechanisms of action and the relative effects of probiotics, particularly concerning diabetes-related complications and metabolic outcomes.
Collapse
Affiliation(s)
- Charikleia Stefanaki
- Department of Pharmacology, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
- Center for Adolescent Medicine and UNESCO Chair in Adolescent Health Care, First Department of Pediatrics, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Paraskevi Rozou
- Hygiene, Social & Preventive Medicine and Medical Statistics Laboratory, Medical School, Aristotle University of Thessaloniki, 54642 Thessaloniki, Greece
- Department of Pediatrics, General Hospital of Larisa, 38221 Larissa, Greece
| | - Vasiliki Efthymiou
- Center for Adolescent Medicine and UNESCO Chair in Adolescent Health Care, First Department of Pediatrics, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Ioannis Xinias
- Third Pediatric Department, Hippokration Hospital, 54642 Thessaloniki, Greece
| | - George Mastorakos
- Unit of Endocrinology, Diabetes Mellitus and Metabolism, Aretaieion Hospital, Athens Medical School, National and Kapodistrian University of Athens, 11528 Athens, Greece
| | - Flora Bacopoulou
- Center for Adolescent Medicine and UNESCO Chair in Adolescent Health Care, First Department of Pediatrics, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Maria Papagianni
- Endocrine Unit, 3rd Department of Pediatrics, Hippokration Hospital of Thessaloniki, Aristotle University of Thessaloniki, 54642 Thessaloniki, Greece
| |
Collapse
|
14
|
Hasnain MA, Kang D, Moon GS. Research trends of next generation probiotics. Food Sci Biotechnol 2024; 33:2111-2121. [PMID: 39130671 PMCID: PMC11315851 DOI: 10.1007/s10068-024-01626-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 05/21/2024] [Accepted: 05/29/2024] [Indexed: 08/13/2024] Open
Abstract
Gut represents one of the largest interfaces for the interaction of host factors and the environmental ones. Gut microbiota, largely dominated by bacterial community, plays a significant role in the health status of the host. The healthy gut microbiota fulfills several vital functions such as energy metabolism, disease protection, and immune modulation. Dysbiosis, characterized by microbial imbalance, can contribute to the development of various disorders, including intestinal, systemic, metabolic, and neurodegenerative conditions. Probiotics offer the potential to address dysbiosis and improve overall health. Advancements in high-throughput sequencing, bioinformatics, and omics have enabled mechanistic studies for the development of bespoke probiotics, referred to as next generation probiotics. These tailor-made probiotics have the potential to ameliorate specific disease conditions and thus fulfill the specific consumer needs. This review discusses recent updates on the most promising next generation probiotics, along with the challenges that must be addressed to translate this concept into reality.
Collapse
Affiliation(s)
- Muhammad Adeel Hasnain
- Major in IT·Biohealth Convergence, Department of IT·Energy Convergence, Graduate School, Korea National University of Transportation, Chungju, 27469 Republic of Korea
| | - Dae‑Kyung Kang
- Department of Animal Resources Science, Dankook University, Cheonan, 31116 Republic of Korea
| | - Gi-Seong Moon
- Major in IT·Biohealth Convergence, Department of IT·Energy Convergence, Graduate School, Korea National University of Transportation, Chungju, 27469 Republic of Korea
- Major in Biotechnology, Korea National University of Transportation, Jeungpyeong, 27909 Republic of Korea
- 4D Convergence Technology Institute, Korea National University of Transportation, Jeungpyeong, 27909 Republic of Korea
| |
Collapse
|
15
|
Kerek Á, Román IL, Szabó Á, Papp M, Bányai K, Kardos G, Kaszab E, Bali K, Makrai L, Jerzsele Á. Comprehensive Metagenomic Analysis of Veterinary Probiotics in Broiler Chickens. Animals (Basel) 2024; 14:1927. [PMID: 38998039 PMCID: PMC11240415 DOI: 10.3390/ani14131927] [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: 05/26/2024] [Revised: 06/21/2024] [Accepted: 06/28/2024] [Indexed: 07/14/2024] Open
Abstract
Probiotics are widely used in broiler chickens to support the gut microbiome, gut health, and to reduce the amount of antibiotics used. Despite their benefits, there is concern over their ability to carry and spread antimicrobial resistance genes (ARGs), posing a significant public health risk. This study utilized next-generation sequencing to investigate ARGs in probiotics approved for poultry, focusing on their potential to be transferred via mobile genetic elements such as plasmids and phages. We examined the gut microbiome and resistome changes in 60 broiler chickens over their rearing period, correlating these changes with different probiotic treatments. Specific resistance mechanisms against critically important antibiotics were identified, including genes related to fluoroquinolone resistance and peptide antibiotic resistance. We also found genes with significant relevance to public health (aadK, AAC(6')-Ii) and multiple drug-resistance genes (vmlR, ykkC, ykkD, msrC, clbA, eatAv). Only one phage-encoded gene (dfrA43) was detected, with no evidence of plasmid or mobile genetic element transmission. Additionally, metagenomic analysis of fecal samples showed no significant changes corresponding to time or diet across groups. Our findings highlight the potential risks associated with the use of probiotics in poultry, particularly regarding the carriage of ARGs. It is crucial to conduct further research into the molecular genetics of probiotics to develop strategies that mitigate the risk of resistance gene transfer in agriculture, ensuring the safe and effective use of probiotics in animal husbandry.
Collapse
Affiliation(s)
- Ádám Kerek
- Department of Pharmacology and Toxicology, University of Veterinary Medicine, István utca 2, H-1078 Budapest, Hungary; (I.L.R.); (Á.S.); (K.B.); (Á.J.)
- National Laboratory of Infectious Animal Diseases, Antimicrobial Resistance, Veterinary Public Health and Food Chain Safety, University of Veterinary Medicine Budapest, H-1078 Budapest, Hungary; (M.P.); (G.K.); (E.K.); (K.B.)
| | - István László Román
- Department of Pharmacology and Toxicology, University of Veterinary Medicine, István utca 2, H-1078 Budapest, Hungary; (I.L.R.); (Á.S.); (K.B.); (Á.J.)
| | - Ábel Szabó
- Department of Pharmacology and Toxicology, University of Veterinary Medicine, István utca 2, H-1078 Budapest, Hungary; (I.L.R.); (Á.S.); (K.B.); (Á.J.)
| | - Márton Papp
- National Laboratory of Infectious Animal Diseases, Antimicrobial Resistance, Veterinary Public Health and Food Chain Safety, University of Veterinary Medicine Budapest, H-1078 Budapest, Hungary; (M.P.); (G.K.); (E.K.); (K.B.)
- Centre for Bioinformatics, University of Veterinary Medicine, István utca 2, H-1078 Budapest, Hungary
| | - Krisztián Bányai
- Department of Pharmacology and Toxicology, University of Veterinary Medicine, István utca 2, H-1078 Budapest, Hungary; (I.L.R.); (Á.S.); (K.B.); (Á.J.)
- National Laboratory of Infectious Animal Diseases, Antimicrobial Resistance, Veterinary Public Health and Food Chain Safety, University of Veterinary Medicine Budapest, H-1078 Budapest, Hungary; (M.P.); (G.K.); (E.K.); (K.B.)
- Veterinary Medical Research Institute, Hungária krt. 21, H-1143 Budapest, Hungary
| | - Gábor Kardos
- National Laboratory of Infectious Animal Diseases, Antimicrobial Resistance, Veterinary Public Health and Food Chain Safety, University of Veterinary Medicine Budapest, H-1078 Budapest, Hungary; (M.P.); (G.K.); (E.K.); (K.B.)
- One Health Institute, University of Debrecen, Nagyerdei krt. 98, H-4032 Debrecen, Hungary
- National Public Health Center, Albert Flórián út 2-6, H-1097 Budapest, Hungary
- Department of Gerontology, Faculty of Health Sciences, University of Debrecen, Sóstói út 2-4, H-4400 Nyíregyháza, Hungary
| | - Eszter Kaszab
- National Laboratory of Infectious Animal Diseases, Antimicrobial Resistance, Veterinary Public Health and Food Chain Safety, University of Veterinary Medicine Budapest, H-1078 Budapest, Hungary; (M.P.); (G.K.); (E.K.); (K.B.)
- One Health Institute, University of Debrecen, Nagyerdei krt. 98, H-4032 Debrecen, Hungary
- Department of Microbiology and Infectious Diseases, University of Veterinary Medicine, István utca 2, H-1078 Budapest, Hungary
| | - Krisztina Bali
- National Laboratory of Infectious Animal Diseases, Antimicrobial Resistance, Veterinary Public Health and Food Chain Safety, University of Veterinary Medicine Budapest, H-1078 Budapest, Hungary; (M.P.); (G.K.); (E.K.); (K.B.)
- Department of Microbiology and Infectious Diseases, University of Veterinary Medicine, István utca 2, H-1078 Budapest, Hungary
| | - László Makrai
- Autovakcina Kft., Szabadság sgrt. 57, H-1171 Budapest, Hungary;
| | - Ákos Jerzsele
- Department of Pharmacology and Toxicology, University of Veterinary Medicine, István utca 2, H-1078 Budapest, Hungary; (I.L.R.); (Á.S.); (K.B.); (Á.J.)
- National Laboratory of Infectious Animal Diseases, Antimicrobial Resistance, Veterinary Public Health and Food Chain Safety, University of Veterinary Medicine Budapest, H-1078 Budapest, Hungary; (M.P.); (G.K.); (E.K.); (K.B.)
| |
Collapse
|
16
|
Kango N, Nath S. Prebiotics, Probiotics and Postbiotics: The Changing Paradigm of Functional Foods. J Diet Suppl 2024; 21:709-735. [PMID: 38881201 DOI: 10.1080/19390211.2024.2363199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/18/2024]
Abstract
The rampant use of antibiotics has led to the emergence of multidrug resistance and is often coupled with gut dysbiosis. To circumvent the harmful impact of antibiotics, probiotics have emerged as an effective intervention. However, while the new probiotics are being added to the list, more recently, the nature and role of their counterparts, viz. prebiotics, postbiotics and parabiotics have also drawn considerable attention. As such, intricate relationships among these gut-biotics vis-à-vis their role in imparting health benefits is to be delineated in a holistic manner. Prebiotic dietary fibers are selectively fermented by probiotics and promote their colonization in the gut. The proliferation of probiotics leads to production of fermentation by-products (postbiotics) which affect the growth of enteropathogens by lowering the pH and producing inhibitory bacteriocins. After completing life-cycle, their dead remnants (parabiotics e.g. exopolysaccharides and cell wall glycoproteins) also inhibit adhesion and biofilm formation of pathogens on the gut epithelium. These beneficial effects are not just endemic to gut but a systemic response is witnessed at different gut-organ axes. Thus, to decipher the role of probiotics, it is imperative to unravel the interdependence between these components. This review elaborates on the recent advancements on various aspects of these gut-biotics and the mechanism of potential attributes like anti-oxidant, anti-inflammatory, anti-neoplastic, anti-lipidemic and anti-hyperglycemic benefits.
Collapse
Affiliation(s)
- Naveen Kango
- Department of Microbiology, Dr. Harisingh Gour Vishwavidyalaya (A Central University), Sagar, India
| | - Suresh Nath
- Department of Microbiology, Dr. Harisingh Gour Vishwavidyalaya (A Central University), Sagar, India
| |
Collapse
|
17
|
Chen K, Jin S, Ma Y, Cai L, Xu P, Nie Y, Luo L, Yu Q, Shen Y, Zhou Z, Liu C. Adjudicative efficacy of Bifidobacterium animalis subsp. lactis BLa80 in treating acute diarrhea in children: a randomized, double-blinded, placebo-controlled study. Eur J Clin Nutr 2024; 78:501-508. [PMID: 38467857 PMCID: PMC11182741 DOI: 10.1038/s41430-024-01428-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Revised: 02/28/2024] [Accepted: 03/01/2024] [Indexed: 03/13/2024]
Abstract
The goal of this study is to assess the efficacy and safety of Bifidobacterium animalis subsp. lactis BLa80, as an adjunct treatment for diarrhea in children with a randomized, double-blinded, placebo-controlled study design. Eligible diarrheal children, aged 0-3 years without the need for antibiotic treatment based on clinical diagnosis when recruited, were randomized into the intervention group (IG, n = 58, with probiotic) or the control group (CG, n = 53, placebo). The primary assessment was the duration of diarrhea. Fecal samples were collected for biochemical index measurement, analysis of gut microbiome composition, and prediction of gene family abundances. The total duration of diarrhea in the IG (122.6 ± 13.1 h) was significantly shorter than in the CG (148.4 ± 17.6 h, p < 0.001). More children in the IG showed improvements in diarrhea compared to the CG, both in intention-to-treat analysis (81.7% vs. 40.0%, p < 0.001) and per protocol analysis (84.4% vs 45.3%, p < 0.001). Cathelicidin level in the IG was significantly higher than that in the CG after the intervention (4415.00 ± 1036.93 pg/g vs. 3679.49 ± 871.18 pg/g, p = 0.0175). The intervention led to an increased abundance of Bifidobacterium breve and Collinsella aerofaciens species, higher alpha-diversity (p < 0.05), and enrichment of functional genes in the gut microbiota related to immunity regulation. Administration of BLa80 at a dose of 5 × 109 CFU/day resulted in a shorter duration of diarrhea and alterations in gut microbiome composition and gene functions.
Collapse
Affiliation(s)
- Ke Chen
- Department of Nutrition, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China.
| | - Shanshan Jin
- Department of Nutrition, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Yu Ma
- Department of Neonatology, Dayi Maternal and Child Health Care Hospital, Chengdu, China
| | - Limei Cai
- Department of Neonatology, Qingbaijiang Maternal and Child Health Care Hospital, Chengdu, China
| | - Ping Xu
- Department of Child Health Care, Qingbaijiang Maternal and Child Health Care Hospital, Chengdu, China
| | - Yang Nie
- Department of Child Health Care, Chongzhou Maternal and Child Health Care Hospital, Chengdu, China
| | - Li Luo
- Department of Pediatrics, Dayi Maternal and Child Health Care Hospital, Chengdu, China
| | - Qinghua Yu
- Laboratory of Microbiology, Immunology and Metabolism, Diprobio (Shanghai) Co., Limited, Shanghai, China
| | - Yang Shen
- Laboratory of Microbiology, Immunology and Metabolism, Diprobio (Shanghai) Co., Limited, Shanghai, China
| | - Zengyuan Zhou
- Department of Nutrition, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Changqi Liu
- School of Exercise and Nutritional Sciences, San Diego State University, San Diego, CA, USA
| |
Collapse
|
18
|
Chuang YF, Fan KC, Su YY, Wu MF, Chiu YL, Liu YC, Lin CC. Precision probiotics supplement strategy in aging population based on gut microbiome composition. Brief Bioinform 2024; 25:bbae351. [PMID: 39038938 PMCID: PMC11262831 DOI: 10.1093/bib/bbae351] [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: 05/02/2024] [Revised: 06/12/2024] [Accepted: 06/24/2024] [Indexed: 07/24/2024] Open
Abstract
With the increasing prevalence of age-related chronic diseases burdening healthcare systems, there is a pressing need for innovative management strategies. Our study focuses on the gut microbiota, essential for metabolic, nutritional, and immune functions, which undergoes significant changes with aging. These changes can impair intestinal function, leading to altered microbial diversity and composition that potentially influence health outcomes and disease progression. Using advanced metagenomic sequencing, we explore the potential of personalized probiotic supplements in 297 older adults by analyzing their gut microbiota. We identified distinctive Lactobacillus and Bifidobacterium signatures in the gut microbiota of older adults, revealing probiotic patterns associated with various population characteristics, microbial compositions, cognitive functions, and neuroimaging results. These insights suggest that tailored probiotic supplements, designed to match individual probiotic profile, could offer an innovative method for addressing age-related diseases and functional declines. Our findings enhance the existing evidence base for probiotic use among older adults, highlighting the opportunity to create more targeted and effective probiotic strategies. However, additional research is required to validate our results and further assess the impact of precision probiotics on aging populations. Future studies should employ longitudinal designs and larger cohorts to conclusively demonstrate the benefits of tailored probiotic treatments.
Collapse
Affiliation(s)
- Yi-Fang Chuang
- School of Medicine, National Yang Ming Chiao Tung University, No. 155, Sec. 2, Linong St. Beitou Dist., Taipei City 112304, Taiwan
- Institute of Public Health, National Yang Ming Chiao Tung University, No. 155, Sec. 2, Linong St. Beitou Dist., Taipei City 112304, Taiwan
- Department of Psychiatry, Far Eastern Memorial Hospital, No. 21, Sec. 2, Nanya S. Rd., Banqiao Dist., New Taipei City 220216, Taiwan
| | - Kang-Chen Fan
- School of Medicine, National Yang Ming Chiao Tung University, No. 155, Sec. 2, Linong St. Beitou Dist., Taipei City 112304, Taiwan
| | - Yin-Yuan Su
- Institute of Biomedical Informatics, National Yang Ming Chiao Tung University, No. 155, Sec. 2, Linong St. Beitou Dist., Taipei City 112304, Taiwan
| | - Ming-Fong Wu
- Institute of Biomedical Informatics, National Yang Ming Chiao Tung University, No. 155, Sec. 2, Linong St. Beitou Dist., Taipei City 112304, Taiwan
| | - Yen-Ling Chiu
- Department of Medical Research, Far Eastern Memorial Hospital, No. 21, Sec. 2, Nanya S. Rd., Banqiao Dist., New Taipei City 220216, Taiwan
- Graduate Program in Biomedical Informatics and Graduate Institute of Medicine, Yuan Ze University, No. 135, Yuandong Rd., Zhongli Dist., Taoyuan City 320315, Taiwan
- Graduate Institute of Clinical Medicine, National Taiwan University, No. 1, Sec. 4, Roosevelt Rd., Taipei City 106319, Taiwan
| | - Yi-Chien Liu
- Department of Neurology, Cardinal Tien Hospital, No. 362, Zhongzheng Rd., Xindian Dist., New Taipei City 231009, Taiwan
| | - Chen-Ching Lin
- Institute of Biomedical Informatics, National Yang Ming Chiao Tung University, No. 155, Sec. 2, Linong St. Beitou Dist., Taipei City 112304, Taiwan
| |
Collapse
|
19
|
Deady C, McCarthy FP, Barron A, McCarthy CM, O’Keeffe GW, O’Mahony SM. An altered gut microbiome in pre-eclampsia: cause or consequence. Front Cell Infect Microbiol 2024; 14:1352267. [PMID: 38774629 PMCID: PMC11106424 DOI: 10.3389/fcimb.2024.1352267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 04/22/2024] [Indexed: 05/24/2024] Open
Abstract
Hypertensive disorders of pregnancy, including pre-eclampsia, are a leading cause of serious and debilitating complications that affect both the mother and the fetus. Despite the occurrence and the health implications of these disorders there is still relatively limited evidence on the molecular underpinnings of the pathophysiology. An area that has come to the fore with regard to its influence on health and disease is the microbiome. While there are several microbiome niches on and within the body, the distal end of the gut harbors the largest of these impacting on many different systems of the body including the central nervous system, the immune system, and the reproductive system. While the role of the microbiome in hypertensive disorders, including pre-eclampsia, has not been fully elucidated some studies have indicated that several of the symptoms of these disorders are linked to an altered gut microbiome. In this review, we examine both pre-eclampsia and microbiome literature to summarize the current knowledge on whether the microbiome drives the symptoms of pre-eclampsia or if the aberrant microbiome is a consequence of this condition. Despite the paucity of studies, obvious gut microbiome changes have been noted in women with pre-eclampsia and the individual symptoms associated with the condition. Yet further research is required to fully elucidate the role of the microbiome and the significance it plays in the development of the symptoms. Regardless of this, the literature highlights the potential for a microbiome targeted intervention such as dietary changes or prebiotic and probiotics to reduce the impact of some aspects of these disorders.
Collapse
Affiliation(s)
- Clara Deady
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Fergus P. McCarthy
- Department of Obstetrics and Gynecology, University College Cork, Cork, Ireland
- The Infant Research Centre, University College Cork, Cork, Ireland
| | - Aaron Barron
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
- Department of Pharmacology and Therapeutics, University College Cork, Cork, Ireland
| | - Cathal M. McCarthy
- Department of Pharmacology and Therapeutics, University College Cork, Cork, Ireland
| | - Gerard W. O’Keeffe
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- Cork Neuroscience Centre, University College Cork, Cork, Ireland
| | - Siobhain M. O’Mahony
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| |
Collapse
|
20
|
Meng J, Liu S, Wu X. Engineered probiotics as live biotherapeutics for diagnosis and treatment of human diseases. Crit Rev Microbiol 2024; 50:300-314. [PMID: 36946080 DOI: 10.1080/1040841x.2023.2190392] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 01/09/2023] [Indexed: 03/23/2023]
Abstract
The use of probiotics to regulate the intestinal microbiota to prevent and treat a large number of disorders and diseases has been an international research hotspot. Although conventional probiotics have a certain regulatory role in nutrient metabolism, inhibiting pathogens, inducing immune regulation, and maintaining intestinal epithelial barrier function, they are unable to treat certain diseases. In recent years, aided by the continuous development of synthetic biology, engineering probiotics with desired characteristics and functionalities to benefit human health has made significant progress. In this article, we summarise the mechanism of action of conventional probiotics and their limitations and highlight the latest developments in the design and construction of probiotics as living diagnostics and therapeutics for the detection and treatment of a series of diseases, including pathogen infections, cancer, intestinal inflammation, metabolic disorders, vaccine delivery, cognitive health, and fatty liver. Besides we discuss the concerns regarding engineered probiotics and corresponding countermeasures and outline the desired features in the future development of engineered live biotherapeutics.
Collapse
Affiliation(s)
- Jiao Meng
- Laboratory of Nutrient Resources and Synthetic Biology, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Science, Tianjin, China
| | - Shufan Liu
- Laboratory of Nutrient Resources and Synthetic Biology, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Science, Tianjin, China
- Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education, Tianjin University of Science and Technology; College of Bioengineering, Tianjin University of Science and Technology, Tianjin, China
| | - Xin Wu
- Laboratory of Nutrient Resources and Synthetic Biology, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Science, Tianjin, China
- CAS Key Laboratory of Agro-Ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, China
| |
Collapse
|
21
|
Porcari S, Fusco W, Spivak I, Fiorani M, Gasbarrini A, Elinav E, Cammarota G, Ianiro G. Fine-tuning the gut ecosystem: the current landscape and outlook of artificial microbiome therapeutics. Lancet Gastroenterol Hepatol 2024; 9:460-475. [PMID: 38604200 DOI: 10.1016/s2468-1253(23)00357-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 10/04/2023] [Accepted: 10/10/2023] [Indexed: 04/13/2024]
Abstract
The gut microbiome is acknowledged as a key determinant of human health, and technological progress in the past two decades has enabled the deciphering of its composition and functions and its role in human disorders. Therefore, manipulation of the gut microbiome has emerged as a promising therapeutic option for communicable and non-communicable disorders. Full exploitation of current therapeutic microbiome modulators (including probiotics, prebiotics, and faecal microbiota transplantation) is hindered by several factors, including poor precision, regulatory and safety issues, and the impossibility of providing reproducible and targeted treatments. Artificial microbiota therapeutics (which include a wide range of products, such as microbiota consortia, bacteriophages, bacterial metabolites, and engineered probiotics) have appeared as an evolution of current microbiota modulators, as they promise safe and reproducible effects, with variable levels of precision via different pathways. We describe the landscape of artificial microbiome therapeutics, from those already on the market to those still in the pipeline, and outline the major challenges for positioning these therapeutics in clinical practice.
Collapse
Affiliation(s)
- Serena Porcari
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy; UOC Gastroenterologia and UOC CEMAD Medicina Interna e Gastroenterologia, Department of Medical and Surgical Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - William Fusco
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy; UOC Gastroenterologia and UOC CEMAD Medicina Interna e Gastroenterologia, Department of Medical and Surgical Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Igor Spivak
- Systems Immunology Department, Weizmann Institute of Science, Rehovot, Israel; Medical Clinic III, University Hospital Aachen, Aachen, Germany
| | - Marcello Fiorani
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy; UOC Gastroenterologia and UOC CEMAD Medicina Interna e Gastroenterologia, Department of Medical and Surgical Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Antonio Gasbarrini
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy; UOC Gastroenterologia and UOC CEMAD Medicina Interna e Gastroenterologia, Department of Medical and Surgical Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Eran Elinav
- Systems Immunology Department, Weizmann Institute of Science, Rehovot, Israel; Microbiome and Cancer Division, DKFZ, Heidelberg, Germany
| | - Giovanni Cammarota
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy; UOC Gastroenterologia and UOC CEMAD Medicina Interna e Gastroenterologia, Department of Medical and Surgical Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Gianluca Ianiro
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy; UOC Gastroenterologia and UOC CEMAD Medicina Interna e Gastroenterologia, Department of Medical and Surgical Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy.
| |
Collapse
|
22
|
Martins E, Nascimento da Silva L, Carmo M. Probiotics, prebiotics, and synbiotics in childhood diarrhea. Braz J Med Biol Res 2024; 57:e13205. [PMID: 38656071 PMCID: PMC11027179 DOI: 10.1590/1414-431x2024e13205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 02/08/2024] [Indexed: 04/26/2024] Open
Abstract
Acute diarrhea is the second leading cause of morbidity and mortality attributed to infections in children under five years of age worldwide, with 1.7 million annual estimated cases and more than 500,000 deaths. Although hydroelectrolytic replacement is the gold standard in treating diarrhea, it does not interfere with the restoration of the intestinal microbiota. Several studies have searched for an adequate alternative in restructuring intestinal homeostasis, finding that treatments based on probiotics, prebiotics, and synbiotics are effective, which made such treatments increasingly present in clinical practice by reducing illness duration with minimal side effects. However, there are still controversies regarding some unwanted reactions in patients. The diversity of strains and the peculiarities of the pathogens that cause diarrhea require further studies to develop effective protocols for prevention and treatment. Here, we provide a descriptive review of childhood diarrhea, emphasizing treatment with probiotics, prebiotics, and synbiotics.
Collapse
Affiliation(s)
- E.M.S. Martins
- Laboratório de Patogenicidade Microbiana, Universidade Ceuma, São Luís, MA, Brasil
| | | | - M.S. Carmo
- Laboratório de Patogenicidade Microbiana, Universidade Ceuma, São Luís, MA, Brasil
- Centro Universitário Dom Bosco, São Luís, MA, Brasil
| |
Collapse
|
23
|
Masset Z, Gunaratnam S, Millette M, McFarland LV, Lacroix M. Environmental and Nutritional Parameters Modulating Genetic Expression for Virulence Factors of Clostridioides difficile. Antibiotics (Basel) 2024; 13:365. [PMID: 38667041 PMCID: PMC11047382 DOI: 10.3390/antibiotics13040365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 04/09/2024] [Accepted: 04/11/2024] [Indexed: 04/29/2024] Open
Abstract
Clostridioides difficile infections (CDIs) continue to be a persistent healthcare concern despite newer antibiotic treatments, enhanced infection control practices, and preventive strategies focused on restoring the protective intestinal microbial barrier. Recent strides in gene sequencing research have identified many genes regulating diverse virulence factors for CDIs. These genes may be over- or under-expressed when triggered by various environmental and nutritional factors. The aims of this paper are to review the important genes involved in C. difficile pathogenesis and to identify modifiable environmental, nutritional, and other factors that may trigger the expression of these genes and thus offer new strategies to prevent CDIs.
Collapse
Affiliation(s)
- Zoe Masset
- INRS Armand-Frappier Health Biotechnology Research Centre, Research Laboratories in Sciences, 531 des Prairies Blvd, Laval, QC H7V 1B7, Canada; (Z.M.); (M.L.)
| | - Sathursha Gunaratnam
- Bio-K+, a Kerry Company, Preclinical Research Division, 495 Armand-Frappier Blvd, Laval, QC H7V 4B3, Canada; (S.G.); (M.M.)
| | - Mathieu Millette
- Bio-K+, a Kerry Company, Preclinical Research Division, 495 Armand-Frappier Blvd, Laval, QC H7V 4B3, Canada; (S.G.); (M.M.)
| | - Lynne V. McFarland
- Public Health Reserves Corps, Seattle, WA 98115, USA
- McFarland Consulting, Seattle, WA 98115, USA
| | - Monique Lacroix
- INRS Armand-Frappier Health Biotechnology Research Centre, Research Laboratories in Sciences, 531 des Prairies Blvd, Laval, QC H7V 1B7, Canada; (Z.M.); (M.L.)
| |
Collapse
|
24
|
Laguna JG, Freitas ADS, Barroso FAL, De Jesus LCL, De Vasconcelos OAGG, Quaresma LS, Américo MF, Campos GM, Glória RDA, Dutra JDCF, Da Silva TF, Vital KD, Fernandes SO, Souza RO, Martins FDS, Ferreira E, Santos TM, Birbrair A, De Oliveira MFA, Faria AMC, Carvalho RDDO, Venanzi FM, Le Loir Y, Jan G, Guédon É, Azevedo VADC. Recombinant probiotic Lactococcus lactis delivering P62 mitigates moderate colitis in mice. Front Microbiol 2024; 15:1309160. [PMID: 38680913 PMCID: PMC11047439 DOI: 10.3389/fmicb.2024.1309160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Accepted: 02/21/2024] [Indexed: 05/01/2024] Open
Abstract
Introduction and objective p62 is a human multifunctional adaptor protein involved in key cellular processes such as tissue homeostasis, inflammation, and cancer. It acts as a negative regulator of inflammasome complexes. It may thus be considered a good candidate for therapeutic use in inflammatory bowel diseases (IBD), such as colitis. Probiotics, including recombinant probiotic strains producing or delivering therapeutic biomolecules to the host mucosal surfaces, could help prevent and mitigate chronic intestinal inflammation. The objective of the present study was to combine the intrinsic immunomodulatory properties of the probiotic Lactococcus lactis NCDO2118 with its ability to deliver health-promoting molecules to enhance its protective and preventive effects in the context of ulcerative colitis (UC). Material and methods This study was realized in vivo in which mice were supplemented with the recombinant strain. The intestinal barrier function was analyzed by monitoring permeability, secretory IgA total levels, mucin expression, and tight junction genes. Its integrity was evaluated by histological analyses. Regarding inflammation, colonic cytokine levels, myeloperoxidase (MPO), and expression of key genes were monitored. The intestinal microbiota composition was investigated using 16S rRNA Gene Sequencing. Results and discussion No protective effect of L. lactis NCDO2118 pExu:p62 was observed regarding mice clinical parameters compared to the L. lactis NCDO2118 pExu: empty. However, the recombinant strain, expressing p62, increased the goblet cell counts, upregulated Muc2 gene expression in the colon, and downregulated pro-inflammatory cytokines Tnf and Ifng when compared to L. lactis NCDO2118 pExu: empty and inflamed groups. This recombinant strain also decreased colonic MPO activity. No difference in the intestinal microbiota was observed between all treatments. Altogether, our results show that recombinant L. lactis NCDO2118 delivering p62 protein protected the intestinal mucosa and mitigated inflammatory damages caused by dextran sodium sulfate (DSS). We thus suggest that p62 may constitute part of a therapeutic approach targeting inflammation.
Collapse
Affiliation(s)
- Juliana Guimarães Laguna
- Department of Genetics, Ecology, and Evolution, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Andria dos Santos Freitas
- Department of Genetics, Ecology, and Evolution, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | | | - Luís Cláudio Lima De Jesus
- Department of Genetics, Ecology, and Evolution, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | | | - Ludmila Silva Quaresma
- Department of Genetics, Ecology, and Evolution, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Monique Ferrary Américo
- Department of Genetics, Ecology, and Evolution, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Gabriela Munis Campos
- Department of Genetics, Ecology, and Evolution, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Rafael de Assis Glória
- Department of Genetics, Ecology, and Evolution, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Joyce da Cruz Ferraz Dutra
- Department of Genetics, Ecology, and Evolution, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Tales Fernando Da Silva
- Department of Genetics, Ecology, and Evolution, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Kátia Duarte Vital
- Department of Clinical Analysis and Toxicology, Federal University of Minas Gerais Belo Horizonte, Minas Gerais, Brazil
| | - Simone O. Fernandes
- Department of Clinical Analysis and Toxicology, Federal University of Minas Gerais Belo Horizonte, Minas Gerais, Brazil
| | - Ramon O. Souza
- Department of Microbiology, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | | | - Enio Ferreira
- Department of General Pathology, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Túlio Marcos Santos
- Department of Genetics, Ecology, and Evolution, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Alexander Birbrair
- Department of General Pathology, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
- Department of Dermatology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, United States
| | | | - Ana Maria Caetano Faria
- Department of Biochemistry and Immunology, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | | | - Franco Maria Venanzi
- School of Biosciences and Veterinary Medicine, University of Camerino, Matelica, Italy
| | | | | | | | | |
Collapse
|
25
|
Kumar S, Pattanaik AK, Jadhav SE, Jangir BL. Lactobacillus johnsonii CPN23 vis-à-vis Lactobacillus acidophilus NCDC15 Improves Gut Health, Intestinal Morphometry, and Histology in Weaned Wistar Rats. Probiotics Antimicrob Proteins 2024; 16:474-489. [PMID: 36976517 DOI: 10.1007/s12602-023-10063-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/10/2023] [Indexed: 03/29/2023]
Abstract
The present investigation was carried out with the aim to establish the comparative efficacy of a canine-sourced probiotic meant for canine feeding and a conventional dairy-sourced probiotic. For this purpose, canine-origin Lactobacillus johnsonii CPN23 and dairy-origin Lactobacillus acidophilus NCDC15 were evaluated for potential probiotics health benefits in the rat model. Forty-eight weaned Wistar rats enrolled in this experiment of 8 weeks were fed a basal diet and divided into three dietary treatments. Rats of group I enrolled as control (CON) were given MRS placebo at 1 mL/head/day, while rats of group II (LAJ) and III (LAC) were administered with overnight MRS broth grown-culture of L. johnsonii CPN23 and L. acidophilus NCDC15, respectively, at 1 mL/head/day (108 cfu/mL). The average daily gain and net gain in body weight were significantly higher (p < 0.05) in LAJ and LAC than in CON. Fecal and digesta biochemical attributes altered (p < 0.05) positively in response to both probiotics. Total fecal and pooled digesta SCFAs were higher (p < 0.05) in both LAJ and LAC than in CON. The microbial population in cecal and colonic digesta responded (p < 0.05) positively to both probiotics. The diameter of intestinal segments was higher (p < 005) in LAJ as compared to CON. The number and height of villi in jejunum tended to be higher in LAJ as compared to CON. The humoral immune response to sheep erythrocytes as well as chicken egg-white lysozyme was higher in LAJ as compared to CON. Overall, the results of the study have demonstrated the effectiveness of the canine-sourced L. johnsonii CPN23 as a potential probiotic, with a comparatively better response than the dairy-sourced L. acidophilus NCDC15. It could thus be recommended for use in feeding dogs to help augment their health.
Collapse
Affiliation(s)
- Sachin Kumar
- Clinical and Pet Nutrition Laboratory, Division of Animal Nutrition, ICAR-Indian Veterinary Research Institute, Izatnagar, 243 122, India
- Animal Nutrition Division, ICAR-National Dairy Research Institute, Karnal, 132 001, India
| | - Ashok Kumar Pattanaik
- Clinical and Pet Nutrition Laboratory, Division of Animal Nutrition, ICAR-Indian Veterinary Research Institute, Izatnagar, 243 122, India.
| | - Sunil Ekanath Jadhav
- Clinical and Pet Nutrition Laboratory, Division of Animal Nutrition, ICAR-Indian Veterinary Research Institute, Izatnagar, 243 122, India
| | - Babu Lal Jangir
- Clinical and Pet Nutrition Laboratory, Division of Animal Nutrition, ICAR-Indian Veterinary Research Institute, Izatnagar, 243 122, India
- Department of Veterinary Pathology, College of Veterinary Sciences, Lala Lajpat Rai University of Veterinary and Animal Sciences, Hisar, 125 004, India
| |
Collapse
|
26
|
Wang Y, Liu Z, Chen T. Vaginal microbiota: Potential targets for vulvovaginal candidiasis infection. Heliyon 2024; 10:e27239. [PMID: 38463778 PMCID: PMC10923723 DOI: 10.1016/j.heliyon.2024.e27239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 01/28/2024] [Accepted: 02/27/2024] [Indexed: 03/12/2024] Open
Abstract
Vulvovaginal candidiasis (VVC) is the second most common cause of vaginal infection globally after bacterial vaginosis (BV) and associated with adverse reproductive and obstetric outcomes, including preterm delivery, sexually transmitted infections and pelvic inflammatory disease. Although effective control of VVC is achievable with the use of traditional treatment strategies (i.e., antifungals), the possibility of drug intolerance, treatment failure and recurrence, as well as the appearance of antifungal-resistant Candida species remain critical challenges. Therefore, alternative therapeutic strategies against VVC are urgently required. In recent years, an improved understanding of the dysbiotic vaginal microbiota (VMB) during VVC has prompted the consideration of administering -biotics to restore the balance of the VMB within the context of VVC prevention and treatment. Here, we aim to summarize the current evidence of the anti-Candida effects of probiotics, postbiotics and synbiotics and their potential use as an alternative/complementary therapy against VVC. Additionally, this review discusses advantages and challenges associated with the application of -biotics in VVC to provide guidance for their later use. We also review new developments in VVC therapy, i.e., vaginal microbiota transplantation (VMT) as an emerging live biotherapeutic therapy against VVC and discuss existing shortcomings associated with this nascent field, expecting to stimulate further investigations for introduction of new therapies against VVC.
Collapse
Affiliation(s)
- Yufei Wang
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Nanchang University Jiangxi Medical College, No.1299, Xuefu Avenue, Honggutan District, Nanchang City, Jiangxi Province, China
- Queen Mary School, Jiangxi Medical College, Nanchang University, No.1299, Xuefu Avenue, Honggutan District, Nanchang City, Jiangxi Province, China
| | - Zhaoxia Liu
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Nanchang University Jiangxi Medical College, No.1299, Xuefu Avenue, Honggutan District, Nanchang City, Jiangxi Province, China
| | - Tingtao Chen
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Nanchang University Jiangxi Medical College, No.1299, Xuefu Avenue, Honggutan District, Nanchang City, Jiangxi Province, China
- School of Pharmacy, National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Jiangxi Medical College, Nanchang University, No.1299, Xuefu Avenue, Honggutan District, Nanchang City, Jiangxi Province, China
| |
Collapse
|
27
|
Mitsou EK, Katsagoni CN, Janiszewska K. Knowledge of Dietitians on Gut Microbiota in Health-An Online Survey of the European Federation of the Associations of Dietitians (EFAD). Nutrients 2024; 16:621. [PMID: 38474750 DOI: 10.3390/nu16050621] [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: 01/22/2024] [Revised: 02/16/2024] [Accepted: 02/21/2024] [Indexed: 03/14/2024] Open
Abstract
Explorations of current knowledge of dietitians about gut-health interconnection and the role of diet in gut microbiota manipulation are rather scarce in the literature. In this online survey we assessed the perceived and current knowledge of dietitians across Europe about gut microbiota and systemic health, nutrition as a modulator of the gut ecosystem, and the role of probiotics and prebiotics. Pre-graduate dietetic students and other professionals were also invited to participate. A total of 179 full responses were recorded (dietitians, n = 155), mainly from Southern and Western regions. Most participants (>78.0%) reported an average to good level of perceived knowledge, with significant positive correlations between perceived and current knowledge in all sections and overall (p for all <0.05). Nevertheless, a rather low current knowledge scoring of participants about probiotics and prebiotics was observed. Features such as being a dietitian, having a higher educational level as dietitian and working in an academic/research setting were usually associated with higher current knowledge. Further analysis revealed that dietitians had a trend for higher scoring about probiotics and prebiotics compared to pre-graduate students or other professionals. Moreover, for dietitians, working in an academic or research setting was an independent factor for scoring in the highest quartile in all tested sections and overall (p for all <0.05). In conclusion, this online survey shed some light on the current knowledge of dietitians across Europe about gut microbiota parameters, including dietary modulation, highlighting in parallel possible knowledge determinants. Potential areas for future educational efforts in this rather unexplored field were indicated.
Collapse
Affiliation(s)
- Evdokia K Mitsou
- Department of Nutrition and Dietetics, School of Health Science and Education, Harokopio University, 17671 Athens, Greece
| | - Christina N Katsagoni
- Department of Clinical Nutrition, Agia Sofia Children's Hospital, 11527 Athens, Greece
- The European Federation of the Associations of Dietitians (EFAD), Gooimeer 4-15, 1411 DC Naarden, The Netherlands
| | - Katarzyna Janiszewska
- The European Federation of the Associations of Dietitians (EFAD), Gooimeer 4-15, 1411 DC Naarden, The Netherlands
| |
Collapse
|
28
|
Kallur RK, Madapati S, Mathur A, Bhattacharya S. The role of Weizmannia (Bacillus) coagulans LMG S-31876 in treating IBS-diarrhea. Front Nutr 2024; 10:1310462. [PMID: 38375355 PMCID: PMC10875997 DOI: 10.3389/fnut.2023.1310462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Accepted: 12/15/2023] [Indexed: 02/21/2024] Open
Abstract
Introduction Irritable bowel syndrome (IBS) is a common gastrointestinal condition. Some studies have shown the efficacy of probiotics in the treatment of irritable bowel syndrome (IBS). Weizmannia (Bacillus) coagulans LMG S-31876 has been marketed as a dietary ingredient, but to date, its efficacy in diarrhea-predominant irritable bowel syndrome (IBS) condition has not been clinically elucidated. Thus, a double-blind placebo-controlled multi-centered trial was planned to evaluate the safety and efficacy of Weizmannia (Bacillus) coagulans LMG S-31876 in diarrhea-predominant IBS patients. Experimental design Study participants (n = 50) diagnosed with IBS prominent symptoms that include abdominal pain and other GI-related symptoms were treated with ProBC Plus (2 billion CFU) along with a placebo capsule once daily for approximately 8 weeks. Study participants were evaluated for the treatment success determined by the differences in stool consistency and frequency per day between the intervention and placebo groups over the study period. Results The vital signs and the biochemistry parameters were under the normal range; the other parameters showed a significant result as compared to the placebo during the study period. Conclusion This study depicts a significant decline in the clinical symptoms such as abdominal pain, bloating, diarrhea, and frequency of the stool as compared to the placebo. All the parameters such as hematology, lipid profile, and vital signs were in the normal range during the supplementation of ProBC Plus for a period of 8 weeks. Furthermore, the study verified that Weizmannia (Bacillus) coagulans LMG S-31876 and its probiotic product ProBC Plus at a dose of 2 billion/CFU/day has a prominent action in the relief from the clinical symptoms of IBS-D. Therefore, the product is intended safe to utilize for IBS-related symptoms.Clinical trial registration: The clinical study has been registered with CTRI/2023/01/048644 with https://ctri.nic.in/Clinicaltrials/pmaindet2.php?trialid=77708&EncHid=24313.96864&userName=CTRI/2023/01/048644 [CTRI/2023/01/048644].
Collapse
Affiliation(s)
| | | | | | - Sourish Bhattacharya
- Process Design and Engineering Cell, CSIR-Central Salt and Marine Chemicals Research Institute, Bhavnagar, India
| |
Collapse
|
29
|
Mantel M, Durand T, Bessard A, Pernet S, Beaudeau J, Guimaraes-Laguna J, Maillard MB, Guédon E, Neunlist M, Le Loir Y, Jan G, Rolli-Derkinderen M. Propionibacterium freudenreichii CIRM-BIA 129 mitigates colitis through S layer protein B-dependent epithelial strengthening. Am J Physiol Gastrointest Liver Physiol 2024; 326:G163-G175. [PMID: 37988603 DOI: 10.1152/ajpgi.00198.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 11/10/2023] [Accepted: 11/20/2023] [Indexed: 11/23/2023]
Abstract
The growing incidence of human diseases involving inflammation and increased gut permeability makes the quest for protective functional foods more crucial than ever. Propionibacterium freudenreichii (P. freudenreichii) is a beneficial bacterium used in the dairy and probiotic industries. Selected strains exert anti-inflammatory effects, and the present work addresses whether the P. freudenreichii CIRM-BIA129, consumed daily in a preventive way, could protect mice from acute colitis induced by dextran sodium sulfate (DSS), and more precisely, whether it could protect from intestinal epithelial breakdown induced by inflammation. P. freudenreichii CIRM-BIA129 mitigated colitis severity and inhibited DSS-induced permeability. It limited crypt length reduction and promoted the expression of zonula occludens-1 (ZO-1), without reducing interleukin-1β mRNA (il-1β) expression. In vitro, P. freudenreichii CIRM-BIA129 prevented the disruption of a Caco-2 monolayer induced by proinflammatory cytokines. It increased transepithelial electrical resistance (TEER) and inhibited permeability induced by inflammation, along with an increased ZO-1 expression. Extracellular vesicles (EVs) from P. freudenreichii CIRM-BIA129, carrying the surface layer protein (SlpB), reproduced the protective effect of P. freudenreichii CIRM-BIA129. A mutant strain deleted for slpB (ΔslpB), or EVs from this mutant strain, had lost their protective effects and worsened both DSS-induced colitis and inflammation in vivo. These results shown that P. freudenreichii CIRM-BIA129 daily consumption has the potential to greatly alleviate colitis symptoms and, particularly, to counter intestinal epithelial permeability induced by inflammation by restoring ZO-1 expression through mechanisms involving S-layer protein B. They open new avenues for the use of probiotic dairy propionibacteria and/or postbiotic fractions thereof, in the context of gut permeability.NEW & NOTEWORTHY Propionibacterium freudenreichii reduces dextran sodium sulfate (DSS)-induced intestinal permeability in vivo. P. freudenreichii does not inhibit inflammation but damages linked to inflammation. P. freudenreichii inhibits intestinal epithelial breakdown through S-layer protein B. The protective effects of P. freudenreichii depend on S-layer protein B. Extracellular vesicles from P. freudenreichii CB 129 mimic the protective effect of the probiotic.
Collapse
Affiliation(s)
- Marine Mantel
- The Enteric Nervous System In Gut And Brain Disorders, IMAD, Institut National de la Santé et de la Recherche Médicale, Nantes Université, Nantes, France
- Institut National de Recherche pour l'Agriculture, l'Alimentation et l'Environnement, Unité Mixte de Recherche, L'Institut Agro, Rennes, France
| | - Tony Durand
- The Enteric Nervous System In Gut And Brain Disorders, IMAD, Institut National de la Santé et de la Recherche Médicale, Nantes Université, Nantes, France
| | - Anne Bessard
- The Enteric Nervous System In Gut And Brain Disorders, IMAD, Institut National de la Santé et de la Recherche Médicale, Nantes Université, Nantes, France
| | - Ségolène Pernet
- The Enteric Nervous System In Gut And Brain Disorders, IMAD, Institut National de la Santé et de la Recherche Médicale, Nantes Université, Nantes, France
| | - Julie Beaudeau
- The Enteric Nervous System In Gut And Brain Disorders, IMAD, Institut National de la Santé et de la Recherche Médicale, Nantes Université, Nantes, France
- Centres de Recherche en Nutrition Humaine-Ouest, Nantes, France
| | - Juliana Guimaraes-Laguna
- Institut National de Recherche pour l'Agriculture, l'Alimentation et l'Environnement, Unité Mixte de Recherche, L'Institut Agro, Rennes, France
| | - Marie-Bernadette Maillard
- Institut National de Recherche pour l'Agriculture, l'Alimentation et l'Environnement, Unité Mixte de Recherche, L'Institut Agro, Rennes, France
| | - Eric Guédon
- Institut National de Recherche pour l'Agriculture, l'Alimentation et l'Environnement, Unité Mixte de Recherche, L'Institut Agro, Rennes, France
| | - Michel Neunlist
- The Enteric Nervous System In Gut And Brain Disorders, IMAD, Institut National de la Santé et de la Recherche Médicale, Nantes Université, Nantes, France
| | - Yves Le Loir
- Institut National de Recherche pour l'Agriculture, l'Alimentation et l'Environnement, Unité Mixte de Recherche, L'Institut Agro, Rennes, France
| | - Gwénaël Jan
- Institut National de Recherche pour l'Agriculture, l'Alimentation et l'Environnement, Unité Mixte de Recherche, L'Institut Agro, Rennes, France
| | - Malvyne Rolli-Derkinderen
- The Enteric Nervous System In Gut And Brain Disorders, IMAD, Institut National de la Santé et de la Recherche Médicale, Nantes Université, Nantes, France
| |
Collapse
|
30
|
Souza RO, Miranda VC, Quintanilha MF, Gallotti B, Oliveira SRM, Silva JL, Alvarez-Leite JI, Jesus LCL, Azevedo V, Vital KD, Fernandes SOA, Cardoso VN, Ferreira E, Nicoli JR, Martins FS. Evaluation of the Treatment with Akkermansia muciniphila BAA-835 of Chemotherapy-induced Mucositis in Mice. Probiotics Antimicrob Proteins 2024; 16:275-292. [PMID: 36652108 DOI: 10.1007/s12602-023-10040-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/05/2023] [Indexed: 01/19/2023]
Abstract
Mucositis is a high-incidence side effect in cancer patients undergoing chemotherapy. Next-generation probiotics are emerging as new therapeutic tools for managing various disorders. Studies have demonstrated the potential of Akkermansia muciniphila to increase the efficiency of anticancer treatment and to mitigate mucositis. Due to the beneficial effect of A. muciniphila on the host, we evaluated the dose-response, the microorganism viability, and the treatment protocol of A. muciniphila BAA-835 in a murine model of chemotherapy-induced mucositis. Female Balb/c mice were divided into groups that received either sterile 0.9% saline or A. muciniphila by gavage. Mucositis was induced using a single intraperitoneal injection of 5-fluorouracil. The animals were euthanized three days after the induction of mucositis, and tissue and blood were collected for analysis. Prevention of weight loss and small intestine shortening and reduction of neutrophil and eosinophil influx were observed when animals were pretreated with viable A. muciniphila at 1010 colony-forming units per mL (CFU/mL). The A. muciniphila improved mucosal damage by preserving tissue architecture and increasing villus height and goblet cell number. It also improved the integrity of the epithelial barrier, decreasing intestinal permeability and bacterial translocation. In addition, the treatment prevented the expansion of Enterobacteriaceae. The immunological parameters were also improved by decreasing the expression of pro-inflammatory cytokines (IL6, IL1β, and TNF) and increasing IL10. In conclusion, pretreatment with 1010 CFU/mL of viable A. muciniphila effectively controlled inflammation, protected the intestinal mucosa and the epithelial barrier, and prevented Enterobacteriaceae expansion in treated mice.
Collapse
Affiliation(s)
- Ramon O Souza
- Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Vivian C Miranda
- Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Mônica F Quintanilha
- Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Bruno Gallotti
- Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Samantha R M Oliveira
- Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Janayne L Silva
- Departamento de Bioquímica E Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Jacqueline I Alvarez-Leite
- Departamento de Bioquímica E Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Luís C L Jesus
- Departamento de Genética, Ecologia E Evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Vasco Azevedo
- Departamento de Genética, Ecologia E Evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Kátia D Vital
- Departamento de Análises Clínicas E Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Simone O A Fernandes
- Departamento de Análises Clínicas E Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Valbert N Cardoso
- Departamento de Análises Clínicas E Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Enio Ferreira
- Departamento de Patologia Geral, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Jacques R Nicoli
- Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Flaviano S Martins
- Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil.
| |
Collapse
|
31
|
Maftei NM, Raileanu CR, Balta AA, Ambrose L, Boev M, Marin DB, Lisa EL. The Potential Impact of Probiotics on Human Health: An Update on Their Health-Promoting Properties. Microorganisms 2024; 12:234. [PMID: 38399637 PMCID: PMC10891645 DOI: 10.3390/microorganisms12020234] [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: 12/17/2023] [Revised: 01/17/2024] [Accepted: 01/17/2024] [Indexed: 02/25/2024] Open
Abstract
Probiotics, known to be live microorganisms, have been shown to improve or restore the gut microbiota, which in turn has been linked to improved health. It is believed that probiotics are the modern equivalent of a panacea, with claims that they may treat or prevent different diseases both in children and adults (e.g., from colic in babies to cardiovascular disease, respiratory infection, and cancer in adults). Ever since the early 2000s, probiotic-based fermented foods have had a resurgence in popularity, mostly due to claims made regarding their health benefits. Fermented foods have been associated with the prevention of irritable bowel syndrome, lactose intolerance, gastroenteritis, and obesity, but also other conditions such as chronic diarrhea, allergies, dermatitis, and bacterial and viral infections, all of which are closely related to an unhealthy lifestyle. Recent and ongoing developments in microbiome/microbiota science have given us new research directions for probiotics. The new types, mechanisms, and applications studied so far, and those currently under study, have a great potential to change scientific understanding of probiotics' nutritional applications and human health care. The expansion of fields related to the study of the microbiome and the involvement of probiotics in its improvement foreshadow an era of significant changes. An expanding range of candidate probiotic species is emerging that can address newly elucidated data-driven microbial niches and host targets. In the probiotic field, new variants of microbiome-modulating interventions are being developed, including prebiotics, symbiotics, postbiotics, microbial consortia, live biotherapeutic products, and genetically modified organisms, with renewed interest in polyphenols, fibers, and fermented foods to ensure human health. This manuscript aims to analyze recent, emerging, and anticipated trends in probiotics (sources, doses, mechanism of action, diseases for which probiotics are administered, side effects, and risks) and create a vision for the development of related areas of influence in the field.
Collapse
Affiliation(s)
- Nicoleta-Maricica Maftei
- Department of Pharmaceutical Sciences, Faculty of Medicine, and Pharmacy, “Dunărea de Jos” University, 800010 Galati, Romania; (N.-M.M.); (E.L.L.)
- Clinic Laboratory Department, Clinical Hospital of Children Hospital “Sf. Ioan”, 800487 Galati, Romania
- Research Centre in the Medical-Pharmaceutical Field, “Dunarea de Jos” University of Galati, 800010 Galati, Romania
| | - Cosmin Raducu Raileanu
- Department of Morphological and Functional Sciences, Faculty of Medicine, and Pharmacy, “Dunărea de Jos” University, 800010 Galati, Romania; (C.R.R.); (L.A.)
| | - Alexia Anastasia Balta
- Medical Department Faculty of Medicine and Pharmacy, “Dunărea de Jos” University, 800010 Galati, Romania;
| | - Lenuta Ambrose
- Department of Morphological and Functional Sciences, Faculty of Medicine, and Pharmacy, “Dunărea de Jos” University, 800010 Galati, Romania; (C.R.R.); (L.A.)
| | - Monica Boev
- Department of Pharmaceutical Sciences, Faculty of Medicine, and Pharmacy, “Dunărea de Jos” University, 800010 Galati, Romania; (N.-M.M.); (E.L.L.)
- Research Centre in the Medical-Pharmaceutical Field, “Dunarea de Jos” University of Galati, 800010 Galati, Romania
| | - Denisa Batîr Marin
- Department of Pharmaceutical Sciences, Faculty of Medicine, and Pharmacy, “Dunărea de Jos” University, 800010 Galati, Romania; (N.-M.M.); (E.L.L.)
- Research Centre in the Medical-Pharmaceutical Field, “Dunarea de Jos” University of Galati, 800010 Galati, Romania
| | - Elena Lacramioara Lisa
- Department of Pharmaceutical Sciences, Faculty of Medicine, and Pharmacy, “Dunărea de Jos” University, 800010 Galati, Romania; (N.-M.M.); (E.L.L.)
- Research Centre in the Medical-Pharmaceutical Field, “Dunarea de Jos” University of Galati, 800010 Galati, Romania
| |
Collapse
|
32
|
Hassan WNM, Al-Kaabi MM, Akram NN, Kassim MAK, Pantazi AC. Probiotics for Inflammatory Bowel Disease; A Deep Dive into their Impact on Disease Course and Associated Health Risks. Curr Med Chem 2024; 31:4807-4825. [PMID: 38693730 DOI: 10.2174/0109298673314861240429072352] [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: 03/06/2024] [Revised: 04/05/2024] [Accepted: 04/16/2024] [Indexed: 05/03/2024]
Abstract
To date, the underlying pathology of inflammatory bowel disease (IBD) is undetermined. Disturbance of intestinal gut microbiota was implicated in many health diseases, including IBD. Increasing evidence suggests that probiotics play a beneficial role in restoring the balance of the gut ecosystem. This review searched multiple databases for relevant works that examined probiotics' possible benefits in adults with IBD. Probiotic mode of action in ulcerative colitis patients and Crohn's disease were examined with respect to probiotic strain, their benefits, and their advantages in adult cases. Eligible studies for inclusion were assessed and analyzed. They were effective in reducing IBD disease course, inducing and maintaining remission, particularly for ulcerative colitis patients, with good efficacy and safety profile. However, the evidence for Crohn's disease was lacking. Probiotics positively affect IBD-related risks, reducing the risk of gastrointestinal malignancy and optimizing treating them. Additionally, they improved reduced fertility odds for both genders. The osteoporosis risk among IBD patients was also reduced, although the duration of use and dose were still not established. There was an encouraging role for them in reducing IBD -cardiovascular risks among cases with acute myocardial infarction and those with chronic heart failure. Finally, they had novel use in reducing IBD-related depression and improved overall mental health. In conclusion, we recommend probiotics as an adjuvant therapeutic option for IBD therapy for ulcerative colitis; however, their role in Crohn's disease needs further research.
Collapse
|
33
|
Pal AD, Pal A. Probiotics: beneficial microbes for health and the food industry. MICROBIAL ESSENTIALISM 2024:47-86. [DOI: 10.1016/b978-0-443-13932-1.00026-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
34
|
Rathi A, Pagare R. Efficacy and Safety of Bacillus coagulans LBSC in Drug Induced Constipation Associated With Functional Gastrointestinal Disorder: A Double-Blind, Randomized, Interventional, Parallel, Controlled Trial a Clinical Study on Bacillus coagulans LBSC for Drug Induced Constipation Associated With FGIDs. GLOBAL ADVANCES IN INTEGRATIVE MEDICINE AND HEALTH 2024; 13:27536130241286511. [PMID: 39295947 PMCID: PMC11409293 DOI: 10.1177/27536130241286511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 09/02/2024] [Accepted: 09/08/2024] [Indexed: 09/21/2024]
Abstract
Background Active drugs and nutraceutical supplements commonly induce various gastrointestinal illnesses, and constipation is a major gastrointestinal symptom accompanied with functional gastrointestinal disorders. Drug-induced imbalance in gut microbiota may play critical role in such physiological disturbances. Probiotics have been known for resuming normal and healthy gut microbiome. Objective To investigate the clinical efficacy and safety of Bacillus coagulans LBSC in the treatment of drug induced constipation associated with functional gastrointestinal disorder (FGID) symptoms. Methods A prospective, interventional, randomized, double-blind, parallel, multi-arm, controlled trial with 168 patients experiencing drug induced constipation associated with FGID symptoms (DICAWFGID) screened through Rome IV criteria were randomized into 2 arms, i.e. placebo arm (n = 28) and atorvastatin, atenolol, metformin, amitriptyline, and calcium in test arm (n = 28/arm). Patients in both arms received similar dosages (1 g sachet, 3 times a day) for 35 days. The occurrence of constipation using Bristol Stool Form Scale, assessment of degree of constipation on 4-point Likert scale, occurrence of hard stool and degree of stool expulsion on 3-point scale, and defecation frequency were primary endpoints. While, secondary outcomes consisted of the changes in severity of FGID symptoms, visual analogue scale and tolerance to IP, along with reports of adverse events (AEs) and severe adverse events (SAEs). Results There was a significant reduction in occurrence of constipation (≥98.6% and P-value <0.05) in test arm over the placebo arm. Assessment of co-primary endpoints showed significant improvements in degree of stool consistency (P-value 0.0232; CI: 0.1870, 1.1629), borderline significantly superior in degree of stool expulsion (P-value 0.0553; CI: 0.0378, -0.4939), while the other co-primary efficacy endpoints displayed considerably improved advancement (non-significant, P-value ≥0.05). The intra group analysis of symptoms at start of treatment (SOT) and end of treatment (EOT) revealed a significant reduction in scores for occurrence of constipation and degree of constipation, whereas significant improvement in the scores for degree of stool consistency and degree of stool expulsion (P-value <0.001) after the intervention period. In secondary endpoints, the processed responses clearly signified a considerable positive improvement (non-significant, P-value ≥0.05) in other symptoms of constipation associated with FGIDs as determined by the changes in the EOT-SOT score. The study data also highlighted the safety of Bacillus coagulans LBSC at the studied dose. No AEs and/or SAEs were documented during the investigation. Conclusion At the studied dose, Bacillus coagulans LBSC was safe for oral consumption and effective in the management of the drug induced constipation associated with FGIDs symptoms.
Collapse
Affiliation(s)
- Ankit Rathi
- Department of Biological Sciences, School of Science, Sandip University, Nashik, Maharashtra, India
| | - Ravikiran Pagare
- Department of Biological Sciences, School of Science, Sandip University, Nashik, Maharashtra, India
| |
Collapse
|
35
|
Goff DA, McFarland LV, Johnson S, Goff DW. The dual role for probiotics use in dental practices. FRONTIERS IN ORAL HEALTH 2023; 4:1336565. [PMID: 38179437 PMCID: PMC10764435 DOI: 10.3389/froh.2023.1336565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 12/05/2023] [Indexed: 01/06/2024] Open
Affiliation(s)
- Debra A. Goff
- Global Antibiotic Stewardship, The Ohio State University Wexner Medical Center, Columbus, OH, United States
- Department of Pharmacy, The Ohio State University College of Pharmacy, Columbus, OH, United States
| | | | - Stuart Johnson
- Infectious Diseases Division, Loyola University Medical Center, Chicago, IL, United States
- Departments of Medicine and Research, Edward Hines Jr. VA Hospital, Hines, IL, United States
| | - Douglas W. Goff
- Gilbert and Goff Prosthodontists, Upper Arlington, Columbus, OH, United States
| |
Collapse
|
36
|
McFarland LV, Hecht G, Sanders ME, Goff DA, Goldstein EJC, Hill C, Johnson S, Kashi MR, Kullar R, Marco ML, Merenstein DJ, Millette M, Preidis GA, Quigley EMM, Reid G, Salminen S, Sniffen JC, Sokol H, Szajewska H, Tancredi DJ, Woolard K. Recommendations to Improve Quality of Probiotic Systematic Reviews With Meta-Analyses. JAMA Netw Open 2023; 6:e2346872. [PMID: 38064222 DOI: 10.1001/jamanetworkopen.2023.46872] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2023] Open
Abstract
Importance Systematic reviews and meta-analyses often report conflicting results when assessing evidence for probiotic efficacy, partially because of the lack of understanding of the unique features of probiotic trials. As a consequence, clinical decisions on the use of probiotics have been confusing. Objective To provide recommendations to improve the quality and consistency of systematic reviews with meta-analyses on probiotics, so evidence-based clinical decisions can be made with more clarity. Evidence Review For this consensus statement, an updated literature review was conducted (January 1, 2020, to June 30, 2022) to supplement a previously published 2018 literature search to identify areas where probiotic systematic reviews with meta-analyses might be improved. An expert panel of 21 scientists and physicians with experience on writing and reviewing probiotic reviews and meta-analyses was convened and used a modified Delphi method to develop recommendations for future probiotic reviews. Findings A total of 206 systematic reviews with meta-analysis components on probiotics were screened and representative examples discussed to determine areas for improvement. The expert panel initially identified 36 items that were inconsistently reported or were considered important to consider in probiotic meta-analyses. Of these, a consensus was reached for 9 recommendations to improve the quality of future probiotic meta-analyses. Conclusions and Relevance In this study, the expert panel reached a consensus on 9 recommendations that should promote improved reporting of probiotic systematic reviews with meta-analyses and, thereby, assist in clinical decisions regarding the use of probiotics.
Collapse
Affiliation(s)
- Lynne V McFarland
- McFarland Consulting, Seattle, Washington
- Public Health Reserve Corp, Seattle Washington
| | - Gail Hecht
- Division of Gastroenterology and Nutrition, Loyola University Chicago, Maywood, Illinois
| | - Mary E Sanders
- International Scientific Association for Probiotics and Prebiotics, Centennial, Colorado
| | - Debra A Goff
- Ohio State University Wexner Medical Center, Ohio State University College of Pharmacy, Columbus
| | | | - Colin Hill
- International Scientific Association for Probiotics and Prebiotics, University College Cork, Ireland
| | - Stuart Johnson
- Stritch School of Medicine, Loyola University Medical Center, Chicago, Illinois
- Departments of Medicine and Research, Edward Hines Jr Veterans Affairs Hospital, Hines, Illinois
| | - Maryam R Kashi
- Department of Gastroenterology, AdventHealth Medical Group, Orlando, Florida
| | | | - Maria L Marco
- Department of Food Science and Technology, University of California, Davis
| | - Daniel J Merenstein
- Research Programs Family Medicine, Department of Human Science, Georgetown University School of Health, Washington, DC
| | - Mathieu Millette
- Bio-K Plus, a Kerry Company, Laval, Quebec, Canada
- INRS-Centre Armand-Frappier Santé Biotechnologie, Laval, Quebec, Canada
| | - Geoffrey A Preidis
- Department of Pediatrics, Division of Gastroenterology, Hepatology, and Nutrition, Baylor College of Medicine and Texas Children's Hospital, Houston
| | - Eamonn M M Quigley
- Lynda K and David M. Underwood Center for Digestive Disorders, Houston Methodist Hospital and Weill Cornell Medical College, Houston, Texas
| | - Gregor Reid
- St Joseph's Hospital, Lawson Health Research Institute, London, Ontario, Canada
| | - Seppo Salminen
- Functional Foods Forum, Faculty of Medicine, University of Turku, Turku, Finland
| | - Jason C Sniffen
- Infectious Disease Consultants, Altamonte Springs, Florida
- Department of Internal Medicine, Infectious Diseases and Tropical Medicine Section, University of South Florida, Tampa
| | - Harry Sokol
- Gastroenterology Department, Centre de Recherche Saint-Antoine, Assistance Publique-Hôpitaux de Paris, Sorbonne University, INSERM, Paris, France
- Paris Centre for Microbiome Medicine FHU, Paris, France
- Institut National de la Recherche Agronomique, Unité Mixte de Recherche, Micalis & AgroParisTech, Jouy en Josas, France
| | - Hania Szajewska
- Department of Paediatrics, Medical University of Warsaw, Warsaw, Poland
| | - Daniel J Tancredi
- Department of Pediatrics, University of California, Davis School of Medicine, Sacramento
| | | |
Collapse
|
37
|
DI Pierro F, Guasti L, Zerbinati N, Bertuccioli A, Risso P, DE Palma AA, Calloni AG, Lukezic M, Baggi E, Colombo M. Role of S. salivarius K12 in the prevention of URTI and AGE in nursery-aged children. Minerva Med 2023; 114:878-880. [PMID: 37768683 DOI: 10.23736/s0026-4806.23.08920-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/29/2023]
Affiliation(s)
- Francesco DI Pierro
- Scientific and Research Department, Velleja Research, Milan, Italy -
- Department of Medicine and Surgery, University of Insubria, Varese, Italy -
| | - Luigina Guasti
- Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Nicola Zerbinati
- Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | | | - Paolo Risso
- Department of Health Sciences, University of Genoa, Genoa, Italy
| | | | | | | | | | | |
Collapse
|
38
|
da Silva TF, Glória RDA, de Sousa TJ, Americo MF, Freitas ADS, Viana MVC, de Jesus LCL, da Silva Prado LC, Daniel N, Ménard O, Cochet MF, Dupont D, Jardin J, Borges AD, Fernandes SOA, Cardoso VN, Brenig B, Ferreira E, Profeta R, Aburjaile FF, de Carvalho RDO, Langella P, Le Loir Y, Cherbuy C, Jan G, Azevedo V, Guédon É. Comprehensive probiogenomics analysis of the commensal Escherichia coli CEC15 as a potential probiotic strain. BMC Microbiol 2023; 23:364. [PMID: 38008714 PMCID: PMC10680302 DOI: 10.1186/s12866-023-03112-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 11/06/2023] [Indexed: 11/28/2023] Open
Abstract
BACKGROUND Probiotics have gained attention for their potential maintaining gut and immune homeostasis. They have been found to confer protection against pathogen colonization, possess immunomodulatory effects, enhance gut barrier functionality, and mitigate inflammation. However, a thorough understanding of the unique mechanisms of effects triggered by individual strains is necessary to optimize their therapeutic efficacy. Probiogenomics, involving high-throughput techniques, can help identify uncharacterized strains and aid in the rational selection of new probiotics. This study evaluates the potential of the Escherichia coli CEC15 strain as a probiotic through in silico, in vitro, and in vivo analyses, comparing it to the well-known probiotic reference E. coli Nissle 1917. Genomic analysis was conducted to identify traits with potential beneficial activity and to assess the safety of each strain (genomic islands, bacteriocin production, antibiotic resistance, production of proteins involved in host homeostasis, and proteins with adhesive properties). In vitro studies assessed survival in gastrointestinal simulated conditions and adhesion to cultured human intestinal cells. Safety was evaluated in BALB/c mice, monitoring the impact of E. coli consumption on clinical signs, intestinal architecture, intestinal permeability, and fecal microbiota. Additionally, the protective effects of both strains were assessed in a murine model of 5-FU-induced mucositis. RESULTS CEC15 mitigates inflammation, reinforces intestinal barrier, and modulates intestinal microbiota. In silico analysis revealed fewer pathogenicity-related traits in CEC15, when compared to Nissle 1917, with fewer toxin-associated genes and no gene suggesting the production of colibactin (a genotoxic agent). Most predicted antibiotic-resistance genes were neither associated with actual resistance, nor with transposable elements. The genome of CEC15 strain encodes proteins related to stress tolerance and to adhesion, in line with its better survival during digestion and higher adhesion to intestinal cells, when compared to Nissle 1917. Moreover, CEC15 exhibited beneficial effects on mice and their intestinal microbiota, both in healthy animals and against 5FU-induced intestinal mucositis. CONCLUSIONS These findings suggest that the CEC15 strain holds promise as a probiotic, as it could modulate the intestinal microbiota, providing immunomodulatory and anti-inflammatory effects, and reinforcing the intestinal barrier. These findings may have implications for the treatment of gastrointestinal disorders, particularly some forms of diarrhea.
Collapse
Affiliation(s)
- Tales Fernando da Silva
- 1INRAE, Institut Agro, STLO, UMR1253, 65 rue de Saint Brieuc, 35042, Rennes, Cedex, France
- Department of Genetics, Ecology, and Evolution, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Rafael de Assis Glória
- Department of Genetics, Ecology, and Evolution, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Thiago Jesus de Sousa
- Department of Genetics, Ecology, and Evolution, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Monique Ferrary Americo
- Department of Genetics, Ecology, and Evolution, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Andria Dos Santos Freitas
- 1INRAE, Institut Agro, STLO, UMR1253, 65 rue de Saint Brieuc, 35042, Rennes, Cedex, France
- Department of Genetics, Ecology, and Evolution, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Marcus Vinicius Canário Viana
- Department of Genetics, Ecology, and Evolution, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Luís Cláudio Lima de Jesus
- Department of Genetics, Ecology, and Evolution, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | | | - Nathalie Daniel
- 1INRAE, Institut Agro, STLO, UMR1253, 65 rue de Saint Brieuc, 35042, Rennes, Cedex, France
| | - Olivia Ménard
- 1INRAE, Institut Agro, STLO, UMR1253, 65 rue de Saint Brieuc, 35042, Rennes, Cedex, France
| | - Marie-Françoise Cochet
- 1INRAE, Institut Agro, STLO, UMR1253, 65 rue de Saint Brieuc, 35042, Rennes, Cedex, France
| | - Didier Dupont
- 1INRAE, Institut Agro, STLO, UMR1253, 65 rue de Saint Brieuc, 35042, Rennes, Cedex, France
| | - Julien Jardin
- 1INRAE, Institut Agro, STLO, UMR1253, 65 rue de Saint Brieuc, 35042, Rennes, Cedex, France
| | - Amanda Dias Borges
- Department of clinical and toxicological analysis, Faculty of Pharmacy, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Simone Odília Antunes Fernandes
- Department of clinical and toxicological analysis, Faculty of Pharmacy, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Valbert Nascimento Cardoso
- Department of clinical and toxicological analysis, Faculty of Pharmacy, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Bertram Brenig
- Department of Molecular Biology of Livestock, Institute of Veterinary Medicine, Georg-August Universität Göttingen, Göttingen, Germany
| | - Enio Ferreira
- Department of general pathology, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Rodrigo Profeta
- Department of Genetics, Ecology, and Evolution, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Flavia Figueira Aburjaile
- Department of Genetics, Ecology, and Evolution, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil
- Veterinary school, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | | | - Philippe Langella
- Université Paris Saclay, INRAE, AgroParisTech, UMR1319, MICALIS, Jouy-en-Josas, France
| | - Yves Le Loir
- 1INRAE, Institut Agro, STLO, UMR1253, 65 rue de Saint Brieuc, 35042, Rennes, Cedex, France
| | - Claire Cherbuy
- Université Paris Saclay, INRAE, AgroParisTech, UMR1319, MICALIS, Jouy-en-Josas, France
| | - Gwénaël Jan
- 1INRAE, Institut Agro, STLO, UMR1253, 65 rue de Saint Brieuc, 35042, Rennes, Cedex, France
| | - Vasco Azevedo
- Department of Genetics, Ecology, and Evolution, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Éric Guédon
- 1INRAE, Institut Agro, STLO, UMR1253, 65 rue de Saint Brieuc, 35042, Rennes, Cedex, France.
| |
Collapse
|
39
|
Nongkhai SN, Piemthongkham P, Bankeeree W, Punnapayak H, Lotrakul P, Prasongsuk S. Xylooligosaccharides produced from sugarcane leaf arabinoxylan using xylanase from Aureobasidium pullulans NRRL 58523 and its prebiotic activity toward Lactobacillus spp. Heliyon 2023; 9:e22107. [PMID: 38034795 PMCID: PMC10682688 DOI: 10.1016/j.heliyon.2023.e22107] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 09/07/2023] [Accepted: 11/04/2023] [Indexed: 12/02/2023] Open
Abstract
In an attempt to enhance the value of sugarcane leaf, xylan was extracted and used for xylooligosaccharide (XO) production via enzymatic hydrolysis using xylanase from the black yeast Aureobasidium pullulans. The xylan was extracted from sugarcane leaf using alkali extraction according to the response surface methodology. The highest xylan yield (99.42 ± 4.05 % recovery) was obtained using 14.32 % (w/v) NaOH, 13.25:1 liquid: solid ratio, at 121 °C and 15 lb.in2 for 32 min. Sugar composition and FTIR spectrum analyses confirmed its structure as arabinoxylan. The extracted arabinoxylan had a relatively high molecular weight compared to previous studies. Crude endoxylanase from A. pullulans NRRL 58523 was selected for enzymatic hydrolysis of the xylan. The enzyme hydrolyzed well at 50 °C, pH 4.0 and was relatively stable under this condition (87.38 ± 1.26 % of the activity remained after 60 h). XOs, especially xylobiose and xylotriose, were obtained at the maximum yield of 237.51 ± 17.69 mg/g xylan via endoxylanase hydrolysis under the optimum conditions (50 °C, pH 4.0, 65.31 U/g xylan, 53 h). XOs exhibited species-specific prebiotic activity toward three strains of Lactobacillus spp. but not toward Bifidobacterium spp.
Collapse
Affiliation(s)
- Sorawit Na Nongkhai
- Program in Biotechnology, Faculty of Science, Chulalongkorn University, Bangkok 10330, Thailand
- Plant Biomass Utilization Research Unit, Department of Botany, Faculty of Science, Chulalongkorn University, Bangkok 10330, Thailand
| | - Phitchayakon Piemthongkham
- Program in Biotechnology, Faculty of Science, Chulalongkorn University, Bangkok 10330, Thailand
- Plant Biomass Utilization Research Unit, Department of Botany, Faculty of Science, Chulalongkorn University, Bangkok 10330, Thailand
| | - Wichanee Bankeeree
- Plant Biomass Utilization Research Unit, Department of Botany, Faculty of Science, Chulalongkorn University, Bangkok 10330, Thailand
| | - Hunsa Punnapayak
- Plant Biomass Utilization Research Unit, Department of Botany, Faculty of Science, Chulalongkorn University, Bangkok 10330, Thailand
- Department of Biology, Faculty Science and Technology, Airlangga University, Surabaya 60115, Indonesia
| | - Pongtharin Lotrakul
- Plant Biomass Utilization Research Unit, Department of Botany, Faculty of Science, Chulalongkorn University, Bangkok 10330, Thailand
| | - Sehanat Prasongsuk
- Plant Biomass Utilization Research Unit, Department of Botany, Faculty of Science, Chulalongkorn University, Bangkok 10330, Thailand
- Department of Biology, Faculty Science and Technology, Airlangga University, Surabaya 60115, Indonesia
| |
Collapse
|
40
|
Olson B, Ship N, Butera ML, Warm K, Oen R, Howard J. Clostridioides difficile infection in a skilled nursing facility (SNF): cost savings of an automated, standardized probiotic antimicrobial stewardship programme (ASP) policy. JAC Antimicrob Resist 2023; 5:dlad102. [PMID: 37680882 PMCID: PMC10481250 DOI: 10.1093/jacamr/dlad102] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Accepted: 08/21/2023] [Indexed: 09/09/2023] Open
Abstract
Background With multiple comorbidities and frequent exposures to antibiotics, patients in skilled nursing facilities (SNFs) are much more vulnerable to healthcare-acquired infections. We conducted a quality-improvement, retrospective analysis of all patients with Clostridioides difficile infection (CDI) from 2009 to 2021 at an SNF. Probiotics were initially added to a bundle of antimicrobial stewardship programme (ASP) CDI prevention strategies. Formulations and durations of probiotics were standardized for both oral and enteral administration. To reach all eligible patients, an ASP probiotic policy provided probiotics with every antibiotic course. Objectives To assess the value of providing probiotic therapy to SNF patients at risk for CDI. Patients and methods Patients receiving oral or enteral feeding with antibiotics ordered were eligible to receive probiotics. The incremental cost of CDI prevention, treatment and related care were calculated and compared for each phase of probiotic policy change and feeding type. ASP records for the oral probiotic and level of treatment were used in modelling the cost-effectiveness. Results From quality improvement initiatives aimed at preventing facility-onset (FO) CDI, to ASP policies, probiotic formulations and delegation of ordering authority, the days of acute care treatment required was significantly reduced over the different phases of implementation [152 to 48, OR = 0.22 (0.16-0.31) to 4, OR = 0.08 (0.03-0.23)] after reducing total CDI from 5.8 to 0.3 cases per 10 000 patient-days. The annual cost of oral probiotics increased from $6019 to $14 652 but the modelled net annual savings for the facility was $72 544-$154 085. Conclusions With optimization, the use of probiotics for CDI prevention at an SNF was safe, efficacious and cost-effective.
Collapse
Affiliation(s)
- Bridget Olson
- Department of Pharmacy, Sharp Coronado Hospital & Villa Long Term Care, Sharp HealthCare, 250 Prospect Place, Coronado, CA 92118, USA
| | - Noam Ship
- Research and Development, Bio-K Plus International Inc., 495 Armand-Frappier Boulevard, Laval, Quebec H7V4B3, Canada
| | - Michael L Butera
- Medical Staff, Sharp Coronado Hospital & Villa Long Term Care, Sharp HealthCare, 250 Prospect Place, Coronado, CA 92118, USA
| | - Kenneth Warm
- Medical Staff, Sharp Coronado Hospital & Villa Long Term Care, Sharp HealthCare, 250 Prospect Place, Coronado, CA 92118, USA
| | - Roger Oen
- Medical Staff, Sharp Coronado Hospital & Villa Long Term Care, Sharp HealthCare, 250 Prospect Place, Coronado, CA 92118, USA
| | - John Howard
- Department of Pharmacy, Sharp Coronado Hospital & Villa Long Term Care, Sharp HealthCare, 250 Prospect Place, Coronado, CA 92118, USA
| |
Collapse
|
41
|
Zhang J, Ren L, Zhang L, Gong Y, Xu T, Wang X, Guo C, Zhai L, Yu X, Li Y, Zhu P, Chen R, Jing X, Jing G, Zhou S, Xu M, Wang C, Niu C, Ge Y, Ma B, Shang G, Cui Y, Yao S, Xu J. Single-cell rapid identification, in situ viability and vitality profiling, and genome-based source-tracking for probiotics products. IMETA 2023; 2:e117. [PMID: 38867931 PMCID: PMC10989769 DOI: 10.1002/imt2.117] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 04/22/2023] [Accepted: 05/07/2023] [Indexed: 06/14/2024]
Abstract
Rapid expansion of the probiotics industry demands fast, sensitive, comprehensive, and low-cost strategies for quality assessment. Here, we introduce a culture-free, one-cell-resolution, phenome-genome-combined strategy called Single-Cell Identification, Viability and Vitality tests, and Source-tracking (SCIVVS). For each cell directly extracted from the product, the fingerprint region of D2O-probed single-cell Raman spectrum (SCRS) enables species-level identification with 93% accuracy, based on a reference SCRS database from 21 statutory probiotic species, whereas the C-D band accurately quantifies viability, metabolic vitality plus their intercellular heterogeneity. For source-tracking, single-cell Raman-activated Cell Sorting and Sequencing can proceed, producing indexed, precisely one-cell-based genome assemblies that can reach ~99.40% genome-wide coverage. Finally, we validated an integrated SCIVVS workflow with automated SCRS acquisition where the whole process except sequencing takes just 5 h. As it is >20-fold faster, >10-time cheaper, vitality-revealing, heterogeneity-resolving, and automation-prone, SCIVVS is a new technological and data framework for quality assessment of live-cell products.
Collapse
Affiliation(s)
- Jia Zhang
- Single-Cell Center, CAS Key Laboratory of Biofuels, Shandong Key Laboratory of Energy Genetics, Qingdao Institute of Bioenergy and Bioprocess Technology, Chinese Academy of Sciences Qingdao Shandong China
- Shandong Energy Institute Qingdao Shandong China
- Qingdao New Energy Shandong Laboratory Qingdao Shandong China
- University of Chinese Academy of Sciences Beijing China
| | - Lihui Ren
- Single-Cell Center, CAS Key Laboratory of Biofuels, Shandong Key Laboratory of Energy Genetics, Qingdao Institute of Bioenergy and Bioprocess Technology, Chinese Academy of Sciences Qingdao Shandong China
- Shandong Energy Institute Qingdao Shandong China
- Qingdao New Energy Shandong Laboratory Qingdao Shandong China
- College of Information Science & Engineering Ocean University of China Qingdao Shandong China
| | - Lei Zhang
- Single-Cell Center, CAS Key Laboratory of Biofuels, Shandong Key Laboratory of Energy Genetics, Qingdao Institute of Bioenergy and Bioprocess Technology, Chinese Academy of Sciences Qingdao Shandong China
- Shandong Energy Institute Qingdao Shandong China
- Qingdao New Energy Shandong Laboratory Qingdao Shandong China
- Qingdao Branch of China United Network Communications Co., Ltd. Qingdao Shandong China
| | - Yanhai Gong
- Single-Cell Center, CAS Key Laboratory of Biofuels, Shandong Key Laboratory of Energy Genetics, Qingdao Institute of Bioenergy and Bioprocess Technology, Chinese Academy of Sciences Qingdao Shandong China
- Shandong Energy Institute Qingdao Shandong China
- Qingdao New Energy Shandong Laboratory Qingdao Shandong China
- University of Chinese Academy of Sciences Beijing China
| | - Teng Xu
- Single-Cell Center, CAS Key Laboratory of Biofuels, Shandong Key Laboratory of Energy Genetics, Qingdao Institute of Bioenergy and Bioprocess Technology, Chinese Academy of Sciences Qingdao Shandong China
- Shandong Energy Institute Qingdao Shandong China
- Qingdao New Energy Shandong Laboratory Qingdao Shandong China
- University of Chinese Academy of Sciences Beijing China
| | - Xiaohang Wang
- Single-Cell Center, CAS Key Laboratory of Biofuels, Shandong Key Laboratory of Energy Genetics, Qingdao Institute of Bioenergy and Bioprocess Technology, Chinese Academy of Sciences Qingdao Shandong China
- Shandong Energy Institute Qingdao Shandong China
- Qingdao New Energy Shandong Laboratory Qingdao Shandong China
- University of Chinese Academy of Sciences Beijing China
| | - Cheng Guo
- Eastsea Pharma Co., Ltd. Qingdao Shandong China
| | - Lei Zhai
- China National Research Institute of Food and Fermentation Industries Co., Ltd., China Center of Industrial Culture Collection Beijing China
| | - Xuejian Yu
- China National Research Institute of Food and Fermentation Industries Co., Ltd., China Center of Industrial Culture Collection Beijing China
| | - Ying Li
- Qingdao Single-Cell Biotech. Co., Ltd. Qingdao Shandong China
| | - Pengfei Zhu
- Single-Cell Center, CAS Key Laboratory of Biofuels, Shandong Key Laboratory of Energy Genetics, Qingdao Institute of Bioenergy and Bioprocess Technology, Chinese Academy of Sciences Qingdao Shandong China
- Qingdao Single-Cell Biotech. Co., Ltd. Qingdao Shandong China
| | - Rongze Chen
- Single-Cell Center, CAS Key Laboratory of Biofuels, Shandong Key Laboratory of Energy Genetics, Qingdao Institute of Bioenergy and Bioprocess Technology, Chinese Academy of Sciences Qingdao Shandong China
- Shandong Energy Institute Qingdao Shandong China
- Qingdao New Energy Shandong Laboratory Qingdao Shandong China
- University of Chinese Academy of Sciences Beijing China
| | - Xiaoyan Jing
- Single-Cell Center, CAS Key Laboratory of Biofuels, Shandong Key Laboratory of Energy Genetics, Qingdao Institute of Bioenergy and Bioprocess Technology, Chinese Academy of Sciences Qingdao Shandong China
- Shandong Energy Institute Qingdao Shandong China
- Qingdao New Energy Shandong Laboratory Qingdao Shandong China
- University of Chinese Academy of Sciences Beijing China
| | - Gongchao Jing
- Single-Cell Center, CAS Key Laboratory of Biofuels, Shandong Key Laboratory of Energy Genetics, Qingdao Institute of Bioenergy and Bioprocess Technology, Chinese Academy of Sciences Qingdao Shandong China
- Shandong Energy Institute Qingdao Shandong China
- Qingdao New Energy Shandong Laboratory Qingdao Shandong China
- University of Chinese Academy of Sciences Beijing China
| | - Shiqi Zhou
- Single-Cell Center, CAS Key Laboratory of Biofuels, Shandong Key Laboratory of Energy Genetics, Qingdao Institute of Bioenergy and Bioprocess Technology, Chinese Academy of Sciences Qingdao Shandong China
- Shandong Energy Institute Qingdao Shandong China
- Qingdao New Energy Shandong Laboratory Qingdao Shandong China
| | - Mingyue Xu
- Single-Cell Center, CAS Key Laboratory of Biofuels, Shandong Key Laboratory of Energy Genetics, Qingdao Institute of Bioenergy and Bioprocess Technology, Chinese Academy of Sciences Qingdao Shandong China
- Shandong Energy Institute Qingdao Shandong China
- Qingdao New Energy Shandong Laboratory Qingdao Shandong China
| | - Chen Wang
- Single-Cell Center, CAS Key Laboratory of Biofuels, Shandong Key Laboratory of Energy Genetics, Qingdao Institute of Bioenergy and Bioprocess Technology, Chinese Academy of Sciences Qingdao Shandong China
- Shandong Energy Institute Qingdao Shandong China
- Qingdao New Energy Shandong Laboratory Qingdao Shandong China
| | | | - Yuanyuan Ge
- China National Research Institute of Food and Fermentation Industries Co., Ltd., China Center of Industrial Culture Collection Beijing China
| | - Bo Ma
- Single-Cell Center, CAS Key Laboratory of Biofuels, Shandong Key Laboratory of Energy Genetics, Qingdao Institute of Bioenergy and Bioprocess Technology, Chinese Academy of Sciences Qingdao Shandong China
- Shandong Energy Institute Qingdao Shandong China
- Qingdao New Energy Shandong Laboratory Qingdao Shandong China
- University of Chinese Academy of Sciences Beijing China
| | | | - Yunlong Cui
- Eastsea Pharma Co., Ltd. Qingdao Shandong China
| | - Su Yao
- China National Research Institute of Food and Fermentation Industries Co., Ltd., China Center of Industrial Culture Collection Beijing China
| | - Jian Xu
- Single-Cell Center, CAS Key Laboratory of Biofuels, Shandong Key Laboratory of Energy Genetics, Qingdao Institute of Bioenergy and Bioprocess Technology, Chinese Academy of Sciences Qingdao Shandong China
- Shandong Energy Institute Qingdao Shandong China
- Qingdao New Energy Shandong Laboratory Qingdao Shandong China
- University of Chinese Academy of Sciences Beijing China
| |
Collapse
|
42
|
Lee CG, Cha KH, Kim GC, Im SH, Kwon HK. Exploring probiotic effector molecules and their mode of action in gut-immune interactions. FEMS Microbiol Rev 2023; 47:fuad046. [PMID: 37541953 DOI: 10.1093/femsre/fuad046] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 07/17/2023] [Accepted: 08/03/2023] [Indexed: 08/06/2023] Open
Abstract
Probiotics, live microorganisms that confer health benefits when consumed in adequate amounts, have gained significant attention for their potential therapeutic applications. The beneficial effects of probiotics are believed to stem from their ability to enhance intestinal barrier function, inhibit pathogens, increase beneficial gut microbes, and modulate immune responses. However, clinical studies investigating the effectiveness of probiotics have yielded conflicting results, potentially due to the wide variety of probiotic species and strains used, the challenges in controlling the desired number of live microorganisms, and the complex interactions between bioactive substances within probiotics. Bacterial cell wall components, known as effector molecules, play a crucial role in mediating the interaction between probiotics and host receptors, leading to the activation of signaling pathways that contribute to the health-promoting effects. Previous reviews have extensively covered different probiotic effector molecules, highlighting their impact on immune homeostasis. Understanding how each probiotic component modulates immune activity at the molecular level may enable the prediction of immunological outcomes in future clinical studies. In this review, we present a comprehensive overview of the structural and immunological features of probiotic effector molecules, focusing primarily on Lactobacillus and Bifidobacterium. We also discuss current gaps and limitations in the field and propose directions for future research to enhance our understanding of probiotic-mediated immunomodulation.
Collapse
Affiliation(s)
- Choong-Gu Lee
- Natural Product Informatics Research Center, Korea Institute of Science and Technology, 679, Saimdang-ro, Gangneung 25451, Korea
- Division of Bio-Medical Science and Technology, KIST School, University of Science and Technology, 679, Saimdang-ro, Seoul 02792, Korea
- Department of Convergence Medicine, Wonju College of Medicine, Yonsei University, 20, Ilsan-ro, Wonju 26493, Korea
| | - Kwang Hyun Cha
- Natural Product Informatics Research Center, Korea Institute of Science and Technology, 679, Saimdang-ro, Gangneung 25451, Korea
- Division of Bio-Medical Science and Technology, KIST School, University of Science and Technology, 679, Saimdang-ro, Seoul 02792, Korea
- Department of Convergence Medicine, Wonju College of Medicine, Yonsei University, 20, Ilsan-ro, Wonju 26493, Korea
| | - Gi-Cheon Kim
- Department of Microbiology and Immunology, Institute for Immunology and Immunological Diseases, and Brain Korea 21 PLUS Project for Medical Sciences, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seoul 03722, Korea
| | - Sin-Hyeog Im
- Department of Life Sciences, Pohang University of Science and Technology, 77, Cheongam-ro, Pohang 37673, Korea
- Institute for Convergence Research and Education, Yonsei University, 50-1 Yonsei-ro, Seoul 03722, Korea
- ImmunoBiome Inc, Bio Open Innovation Center, 77, Cheongam-ro, Pohang 37673 , Korea
| | - Ho-Keun Kwon
- Department of Microbiology and Immunology, Institute for Immunology and Immunological Diseases, and Brain Korea 21 PLUS Project for Medical Sciences, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seoul 03722, Korea
| |
Collapse
|
43
|
Salazar-Parra MA, Cruz-Neri RU, Trujillo-Trujillo XA, Dominguez-Mora JJ, Cruz-Neri HI, Guzmán-Díaz JM, Guzmán-Ruvalcaba MJ, Vega-Gastelum JO, Ascencio-Díaz KV, Zarate-Casas MF, González-Ponce FY, Barbosa-Camacho FJ, Fuentes-Orozco C, Cervantes-Guevara G, Cervantes-Pérez E, Cervantes-Cardona GA, Cortés-Flores AO, González-Ojeda A. Effectiveness of Saccharomyces Boulardii CNCM I-745 probiotic in acute inflammatory viral diarrhoea in adults: results from a single-centre randomized trial. BMC Gastroenterol 2023; 23:229. [PMID: 37400812 DOI: 10.1186/s12876-023-02863-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Accepted: 06/27/2023] [Indexed: 07/05/2023] Open
Abstract
BACKGROUND Probiotics are effective for treating acute infectious diarrhoea caused by bacteria, but there are inconsistent results for the effectiveness of probiotics for diarrhoea caused by viruses. In this article we want to determine whether Sb supplementation has an effect on acute inflammatory viral diarrhoea diagnosed with the multiplex panel PCR test. The aim of this study was to evaluate the efficacy of Saccharomyces boulardii (Sb) as a treatment in patients diagnosed with viral acute diarrhoea. METHODS From February 2021 to December 2021, 46 patients with a confirmed diagnosis of viral acute diarrhoea diagnosed with the polymerase chain reaction multiplex assay were enrolled in a double-blind, randomized placebo-controlled trial. Patients received paracetamol 500 mg as a standard analgesic and 200 mg of Trimebutine as an antispasmodic treatment plus 600 mg of Sb (n = 23, 1 × 109/100 mL Colony forming unit) or a placebo (n = 23) orally once daily for eight days. The improvement in and severity of symptoms were measured using a symptom diary, the Patient Global Impression and the Patient Global Impression of Change scales (days 4 and 8), both answered and recorded by the patient. RESULTS Of the 46 patients who completed treatment, 24 (52%) were men and 22 (48%) were women. The average age was 35.6 ± 12.28 years (range 18 to 61 years). The average duration of the evolution of illness at the time of diagnosis was 0.85 ± 0.73 days (maximum 2 days). On day 4 after the diagnosis, 20% reported pain and 2% reported fever, but on day 8, no patient reported pain or fever. On day 4, 70% of patients in the Sb group and 26% in the placebo group reported improvement (P = 0.03), based on the Patients' Global Impression of Change scale, which assesses patient's rating of overall improvement. These findings suggest that 3 to 4 days of treatment with Sb helped to improve symptoms of diarrhoea caused by a virus. CONCLUSION Treatment with Sb on acute inflammatory diarrhoea of viral aetiology shows no changes regarding the severity of the symptoms; nevertheless, it seems to impact improvement positively. TRIAL REGISTRATION 22CEI00320171130 dated on 16/12/2020, NCT05226052 dated on 07/02/2022.
Collapse
Affiliation(s)
- Marcela Ag Salazar-Parra
- Biomedical Research Unit 02, Western National Medical Center, Mexican Institute of Social Security, Avenida Belisario Domínguez #1000, Colonia Independencia, Guadalajara, Jalisco, 44340, Mexico
- Department of Medical Science, University of Colima, Avenida Universidad #333, Colonia las Víboras, Colima, Colima, 28040, México
| | - Roberto U Cruz-Neri
- Surgeon and Coloproctologist, Puerta de Hierro Sur Medical Center, Tlajomulco de Zúñiga, Avenida Adolfo López Mateos Sur #1401, Colonia La Tijera, Jalisco, 45640, Mexico
| | - Xóchitl Ar Trujillo-Trujillo
- University of Colima, Universitary Center of Biomedical Research, Colonia Villas de San Sebastián, Avenida 25 de Julio #965, Colima, Colima, 28045, Mexico
| | - Juan J Dominguez-Mora
- Puerta de Hierro Medical Center, Tlajomulco de Zúñiga, Avenida Adolfo López Mateos Sur #1401, Colonia La Tijera, Jalisco, 45640, Mexico
| | - Héctor I Cruz-Neri
- High Specialty Geriatric Care Unit, Hospital Civil Fray Antonio Alcalde, Calle Hospital #278, Colonia El Retiro, Guadalajara, Jalisco, 44280, México
| | - Jazmín M Guzmán-Díaz
- Biomedical Research Unit 02, Western National Medical Center, Mexican Institute of Social Security, Avenida Belisario Domínguez #1000, Colonia Independencia, Guadalajara, Jalisco, 44340, Mexico
| | - Mario J Guzmán-Ruvalcaba
- Biomedical Research Unit 02, Western National Medical Center, Mexican Institute of Social Security, Avenida Belisario Domínguez #1000, Colonia Independencia, Guadalajara, Jalisco, 44340, Mexico
| | - Jesús O Vega-Gastelum
- Biomedical Research Unit 02, Western National Medical Center, Mexican Institute of Social Security, Avenida Belisario Domínguez #1000, Colonia Independencia, Guadalajara, Jalisco, 44340, Mexico
| | - Kriscia V Ascencio-Díaz
- Biomedical Research Unit 02, Western National Medical Center, Mexican Institute of Social Security, Avenida Belisario Domínguez #1000, Colonia Independencia, Guadalajara, Jalisco, 44340, Mexico
| | - Maria F Zarate-Casas
- Biomedical Research Unit 02, Western National Medical Center, Mexican Institute of Social Security, Avenida Belisario Domínguez #1000, Colonia Independencia, Guadalajara, Jalisco, 44340, Mexico
| | - Fanny Y González-Ponce
- Biomedical Research Unit 02, Western National Medical Center, Mexican Institute of Social Security, Avenida Belisario Domínguez #1000, Colonia Independencia, Guadalajara, Jalisco, 44340, Mexico
| | - Francisco J Barbosa-Camacho
- Biomedical Research Unit 02, Western National Medical Center, Mexican Institute of Social Security, Avenida Belisario Domínguez #1000, Colonia Independencia, Guadalajara, Jalisco, 44340, Mexico
| | - Clotilde Fuentes-Orozco
- Biomedical Research Unit 02, Western National Medical Center, Mexican Institute of Social Security, Avenida Belisario Domínguez #1000, Colonia Independencia, Guadalajara, Jalisco, 44340, Mexico
| | - Gabino Cervantes-Guevara
- Departamento de Bienestar y Desarrollo Sustentable, Centro Universitario del Norte, Universidad de Guadalajara, Carretera Federal No. 23, Km. 191, Colotlán, Jalisco, C.P. 46200, México
| | - Enrique Cervantes-Pérez
- Departamento de Medicina Interna, Centro Universitario de Ciencias de la Salud, Hospital Civil de Guadalajara Fray Antonio Alcalde, Universidad de Guadalajara, Calle Hospital 278, Col. El Retiro, Guadalajara, Jalisco, 95100, México
| | - Guillermo Alonso Cervantes-Cardona
- Departamento de Disciplinas Filosófico, Metodológicas e Instrumentales, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Sierra Mojada 950 Edificio "N" planta alta, Col. Independencia, Guadalajara, Jalisco, 44340, México
| | - Ana Olivia Cortés-Flores
- Surgical Oncology, Anker Global Oncology, Av. Juan Palomar y Arias 530, Guadalajara, Monraz, Guadalajara, Jal. Mexico, 44670, Mexico
| | - Alejandro González-Ojeda
- Biomedical Research Unit 02, Western National Medical Center, Mexican Institute of Social Security, Avenida Belisario Domínguez #1000, Colonia Independencia, Guadalajara, Jalisco, 44340, Mexico.
| |
Collapse
|
44
|
Kaufmann B, Seyfried N, Hartmann D, Hartmann P. Probiotics, prebiotics, and synbiotics in nonalcoholic fatty liver disease and alcohol-associated liver disease. Am J Physiol Gastrointest Liver Physiol 2023; 325:G42-G61. [PMID: 37129252 PMCID: PMC10312326 DOI: 10.1152/ajpgi.00017.2023] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 04/26/2023] [Accepted: 04/26/2023] [Indexed: 05/03/2023]
Abstract
The use of probiotics, prebiotics, and synbiotics has become an important therapy in numerous gastrointestinal diseases in recent years. Modifying the gut microbiota, this therapeutic approach helps to restore a healthy microbiome. Nonalcoholic fatty liver disease and alcohol-associated liver disease are among the leading causes of chronic liver disease worldwide. A disrupted intestinal barrier, microbial translocation, and an altered gut microbiome metabolism, or metabolome, are crucial in the pathogenesis of these chronic liver diseases. As pro-, pre-, and synbiotics modulate these targets, they were identified as possible new treatment options for liver disease. In this review, we highlight the current findings on clinical and mechanistic effects of this therapeutic approach in nonalcoholic fatty liver disease and alcohol-associated liver disease.
Collapse
Affiliation(s)
- Benedikt Kaufmann
- Department of Surgery, Klinikum rechts der Isar, Technical University Munich, Munich, Germany
| | - Nick Seyfried
- Department of Surgery, Klinikum rechts der Isar, Technical University Munich, Munich, Germany
| | - Daniel Hartmann
- Department of Surgery, Klinikum rechts der Isar, Technical University Munich, Munich, Germany
| | - Phillipp Hartmann
- Department of Surgery, Klinikum rechts der Isar, Technical University Munich, Munich, Germany
- Department of Pediatrics, University of California San Diego, La Jolla, California, United States
| |
Collapse
|
45
|
Doar NW, Samuthiram SD. Qualitative Analysis of the Efficacy of Probiotic Strains in the Prevention of Antibiotic-Associated Diarrhea. Cureus 2023; 15:e40261. [PMID: 37440799 PMCID: PMC10335840 DOI: 10.7759/cureus.40261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/17/2023] [Indexed: 07/15/2023] Open
Abstract
Antibiotic-associated diarrhea is often managed by the withdrawal of the culprit antibiotics or the administration of alternative antibiotics when a Clostridium difficile infection (CDI) is suspected, an infection that tends to be the most common causative agent of the disease. Probiotics are also gaining popularity as alternative therapies, and it was hypothesized in this article that a Lactobacillus strain is the most efficacious probiotic for the prevention of antibiotic-associated diarrhea. This article conducted a literature review investigating the relative efficacy of the Lactobacillus, Bifidobacterium, and Saccharomyces probiotic strains as effective alternative therapies for antibiotic-associated diarrhea. The literature searched was from the PubMed database. The inclusion filters were: random control trials (RCTs), clinical trials, meta-analysis, last 10 years, full-text articles available in English, and all articles published in peer-reviewed journals. All three probiotic genera had strains that demonstrated significant efficacy in the prevention of antibiotic-associated diarrhea. However, Saccharomyces boulardii I-745 tends to outperform all the strains as the most effective and the one with the fewest, if any, adverse effects. Whenever probiotics are considered for the prevention of antibiotic-associated diarrhea (AAD) in both pediatric and adult patients, S. boulardii I-745 should probably be prioritized.
Collapse
Affiliation(s)
- Nyier W Doar
- Medicine, Interfaith Medical Center, New York, USA
| | | |
Collapse
|
46
|
Kullar R, Goldstein EJC, Johnson S, McFarland LV. Lactobacillus Bacteremia and Probiotics: A Review. Microorganisms 2023; 11:microorganisms11040896. [PMID: 37110319 PMCID: PMC10145752 DOI: 10.3390/microorganisms11040896] [Citation(s) in RCA: 47] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 03/26/2023] [Accepted: 03/29/2023] [Indexed: 04/29/2023] Open
Abstract
Lactobacilli are widely found in nature, are commensal microbes in humans, and are commonly used as probiotics. Concerns about probiotic safety have arisen due to reports of bacteremia and other Lactobacillus-associated infections. We reviewed the literature for articles on the pathogenicity of Lactobacillus spp. bacteremia and reports of probiotics in these patients. Our aim is to review these articles and update the present knowledge on the epidemiology of Lactobacillus spp. bacteremia and determine the role of probiotics in Lactobacillus bacteremia. Lactobacillus bacteremia is infrequent but has a higher risk of mortality and risk factors, including severe underlying diseases, immune system suppression, admission to intensive care units, and use of central venous catheters. A variety of Lactobacillus species may cause bacteremia and may or may not be associated with probiotic exposure. To determine if oral probiotics are the source of these infections, the blood isolates and the oral probiotic strain(s) must be compared by sensitive identification methods. The prevalence of Lactobacillus bacteremia is infrequent but is more common in patients taking probiotics compared to those not taking probiotics. Three probiotics (Lacticaseibacillus rhamnosus GG, Lactiplantibacillus plantarum, and Lacticaseibacillus paracasei) were directly linked with blood isolates from bacteremia patients using molecular identification assays.
Collapse
Affiliation(s)
- Ravina Kullar
- Expert Stewardship Inc., Newport Beach, CA 92663, USA
| | | | | | | |
Collapse
|
47
|
Ng QX, Lim YL, Yaow CYL, Ng WK, Thumboo J, Liew TM. Effect of Probiotic Supplementation on Gut Microbiota in Patients with Major Depressive Disorders: A Systematic Review. Nutrients 2023; 15:1351. [PMID: 36986088 PMCID: PMC10052013 DOI: 10.3390/nu15061351] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 03/06/2023] [Accepted: 03/09/2023] [Indexed: 03/14/2023] Open
Abstract
There is accumulating evidence on the beneficial effects of probiotic supplementation for patients with depressive disorders. However, prior reviews on the topic have largely focused on clinical effectiveness with limited emphasis on the underlying mechanisms of action and effects of probiotics on gut microbiota. In accordance with PRISMA guidelines, a systematic literature search of Medline, EMBASE and the Cochrane Library using combinations of the key words, ("depress*" OR "MDD" OR "suicide"), ("probiotic" OR "Lactobacillus" OR "Bifidobacterium") AND ("gut" OR "gut micr*" OR "microbiota"), as well as grey literature was performed. We found seven clinical trials involving patients with major depressive disorder (MDD). The small number of studies and heterogeneous sources of data precluded meta-analysis. Most trials (other than one open-label trial) had a low-to-moderate risk of bias, which was largely due to a lack of control for the effects of diet on gut microbiota. Probiotic supplementation yielded only modest effects on depressive symptoms and there were no consistent effects on gut microbiota diversity, and in most instances, no significant alterations in gut microbiota composition were observed after four to eight weeks of probiotic intervention. There is also a lack of systematic reporting on adverse events and no good longer-term data. Patients with MDD may require a longer time to show clinical improvement and the microbial host environment may also need longer than eight weeks to produce significant microbiota alterations. To advance this field, further larger-scale and longer-term studies are required.
Collapse
Affiliation(s)
- Qin Xiang Ng
- Health Services Research Unit, Singapore General Hospital, Singapore 169608, Singapore
- MOH Holdings Pte Ltd., 1 Maritime Square, Singapore 099253, Singapore
| | - Yu Liang Lim
- MOH Holdings Pte Ltd., 1 Maritime Square, Singapore 099253, Singapore
- Department of Gastroenterology and Hepatology, Tan Tock Seng Hospital, Singapore 308433, Singapore
| | | | - Wee Khoon Ng
- Department of Gastroenterology and Hepatology, Tan Tock Seng Hospital, Singapore 308433, Singapore
| | - Julian Thumboo
- Health Services Research Unit, Singapore General Hospital, Singapore 169608, Singapore
- Department of Rheumatology and Immunology, Singapore General Hospital, Singapore 169608, Singapore
- SingHealth Duke-NUS Medicine Academic Clinical Programme, Duke-NUS Medical School, Singapore 169857, Singapore
| | - Tau Ming Liew
- SingHealth Duke-NUS Medicine Academic Clinical Programme, Duke-NUS Medical School, Singapore 169857, Singapore
- Department of Psychiatry, Singapore General Hospital, Singapore 169608, Singapore
- Saw Swee Hock School of Public Health, National University of Singapore, Singapore 117549, Singapore
| |
Collapse
|
48
|
Wombwell E. Saccharomyces boulardii prophylaxis for targeted antibiotics and infectious indications to reduce healthcare facility-onset Clostridioides difficile infection. Microbes Infect 2023; 25:105041. [PMID: 36058513 DOI: 10.1016/j.micinf.2022.105041] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 08/22/2022] [Accepted: 08/25/2022] [Indexed: 02/03/2023]
Abstract
BACKGROUND Probiotic prophylaxis for Clostridioides difficile infection (CDI) is controversial stemming from deficits in strain and disease specificity considerations and concern for adverse effects. Here risk for healthcare facility-onset CDI (HO-CDI) dependent on concomitant antibiotic and infectious indication is assessed to identify opportunities for targeted prophylaxis. METHODS Retrospective matched-cohort study from January 2016 through March 2019. Patient-admissions administered high risk antibiotics were categorized by Saccharomyces boulardii administration and matched 1:1 to non-recipients. Unadjusted and adjusted HO-CDI risk estimated using Cox proportional hazards regression. RESULTS S. boulardii administration was associated with 48% risk reduction for HO-CDI compared to non-recipients (aHR 0.52, 95% CI: 0.31-0.87). Patient-admissions administered antibiotics and S. boulardii for a pneumonia indication exhibited a 57% reduction in risk for HO-CDI (aHR 0.43, 95% CI: 0.19-0.95). Administration of S. boulardii with ceftriaxone was associated with a 76% reduced risk of HO-CDI (aHR 0.24, 95% CI: 0.11-0.53) compared to ceftriaxone without S. boulardii, number needed to treat of 100. CONCLUSIONS S. boulardii administration is associated with a significant HO-CDI risk reduction for inpatients receiving antibiotics associated with CDI. Institutions interested in targeted use of S. boulardii to limit potential adverse effects may consider prophylaxis for inpatients with pneumonia or receiving ceftriaxone.
Collapse
Affiliation(s)
- Eric Wombwell
- Division of Pharmacy Practice and Administration, University of Missouri-Kansas City School of Pharmacy, Kansas City, Missouri, USA; Department of Pharmacy, Centerpoint Medical Center, Independence, Missouri, USA.
| |
Collapse
|
49
|
Ribaldone DG, Pellicano R, Fagoonee S, Actis GC. Modulation of the gut microbiota: opportunities and regulatory aspects. Minerva Gastroenterol (Torino) 2023; 69:128-140. [PMID: 35179341 DOI: 10.23736/s2724-5985.22.03152-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The human gut is an intensively colonized organ containing microorganisms that can be health-promoting or pathogenic. This feature led to the development of functional foods aiming to fortify the former category at the expense of the latter. Since long, cultured products, including probiotics fortification, have been used for humans as live microbial feed additions. This review presents some of the microbes used as probiotics and discusses how supplementation with probiotics may help initiate and/or restore eubiotic composition of gut microbiota. Additionally, it considers safety and regulatory aspects of probiotics.
Collapse
Affiliation(s)
| | | | - Sharmila Fagoonee
- Institute of Biostructures and Bioimaging (CNR) c/o Molecular Biotechnology Center, Turin, Italy
| | | |
Collapse
|
50
|
Fagnant HS, Isidean SD, Wilson L, Bukhari AS, Allen JT, Agans RT, Lee DM, Hatch-McChesney A, Whitney CC, Sullo E, Porter CK, Karl JP. Orally Ingested Probiotic, Prebiotic, and Synbiotic Interventions as Countermeasures for Gastrointestinal Tract Infections in Nonelderly Adults: A Systematic Review and Meta-Analysis. Adv Nutr 2023; 14:539-554. [PMID: 36822240 DOI: 10.1016/j.advnut.2023.02.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 01/26/2023] [Accepted: 02/15/2023] [Indexed: 02/24/2023] Open
Abstract
Meta-analyses have not examined the prophylactic use of orally ingested probiotics, prebiotics, and synbiotics for preventing gastrointestinal tract infections (GTIs) of various etiologies in adult populations, despite evidence that these gut microbiota-targeted interventions can be effective in treating certain GTIs. This systematic review and meta-analysis aimed to estimate the effects of prophylactic use of orally ingested probiotics, prebiotics, and synbiotics on GTI incidence, duration, and severity in nonelderly, nonhospitalized adults. CENTRAL, PubMed, Scopus, and Web of Science were searched through January 2022. English-language, peer-reviewed publications of randomized, placebo-controlled studies testing an orally ingested probiotic, prebiotic, or synbiotic intervention of any dose for ≥1 wk in adults who were not hospitalized, immunosuppressed, or taking antibiotics were included. Results were analyzed using random-effects meta-analyses of intention-to-treat (ITT) and complete case (CC) cohorts. Heterogeneity was explored by subgroup meta-analysis and meta-regression. The risk of bias was assessed using the Cochrane risk-of-bias 2 tool. Seventeen publications reporting 20 studies of probiotics (n = 16), prebiotics (n = 3), and synbiotics (n = 1) were identified (n > 6994 subjects). In CC and ITT analyses, risk of experiencing ≥1 GTI was reduced with probiotics (CC analysis-risk ratio: 0.86; 95% CI: 0.73, 1.01) and prebiotics (risk ratio: 0.80; 95% CI: 0.66, 0.98). No effects on GTI duration or severity were observed. Sources of heterogeneity included the study population and number of probiotic strains administered but were often unexplained, and a high risk of bias was observed for most studies. The specific effects of individual probiotic strains and prebiotic types could not be assessed owing to a lack of confirmatory studies. Findings indicated that both orally ingested probiotics and prebiotics, relative to placebo, demonstrated modest benefit for reducing GTI risk in nonelderly adults. However, results should be interpreted cautiously owing to the low number of studies, high risk of bias, and unexplained heterogeneity that may include probiotic strain-specific or prebiotic-specific effects. This review was registered at PROSPERO as CRD42020200670.
Collapse
Affiliation(s)
- Heather S Fagnant
- US Army Research Institute of Environmental Medicine, Natick, MA, United States
| | - Sandra D Isidean
- Naval Medical Research Center, Silver Spring, MD, United States; Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, United States
| | - Lydia Wilson
- The George Washington University, Washington, DC, United States
| | - Asma S Bukhari
- US Army Research Institute of Environmental Medicine, Natick, MA, United States
| | - Jillian T Allen
- US Army Research Institute of Environmental Medicine, Natick, MA, United States; Oak Ridge Institute of Science and Education, Belcamp, MD, United States
| | - Richard T Agans
- U.S. Air Force School of Aerospace Medicine, Dayton, OH, United States
| | - Dustin M Lee
- Brooke Army Medical Center, Fort Sam Houston, TX, United States
| | | | - Claire C Whitney
- US Army Research Institute of Environmental Medicine, Natick, MA, United States
| | - Elaine Sullo
- The George Washington University, Washington, DC, United States
| | - Chad K Porter
- Naval Medical Research Center, Silver Spring, MD, United States
| | - J Philip Karl
- US Army Research Institute of Environmental Medicine, Natick, MA, United States.
| |
Collapse
|