1
|
Xia J, Cui Y, Guo Y, Liu Y, Deng B, Han S. The Function of Probiotics and Prebiotics on Canine Intestinal Health and Their Evaluation Criteria. Microorganisms 2024; 12:1248. [PMID: 38930630 PMCID: PMC11205510 DOI: 10.3390/microorganisms12061248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 06/16/2024] [Accepted: 06/18/2024] [Indexed: 06/28/2024] Open
Abstract
Maintaining homeostasis within the intestinal microbiota is imperative for assessing the health status of hosts, and dysbiosis within the intestinal microbiota is closely associated with canine intestinal diseases. In recent decades, the modulation of canine intestinal health through probiotics and prebiotics has emerged as a prominent area of investigation. Evidence indicates that probiotics and prebiotics play pivotal roles in regulating intestinal health by modulating the intestinal microbiota, fortifying the epithelial barrier, and enhancing intestinal immunity. This review consolidates literature on using probiotics and prebiotics for regulating microbiota homeostasis in canines, thereby furnishing references for prospective studies and formulating evaluation criteria.
Collapse
Affiliation(s)
| | | | | | | | - Baichuan Deng
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (J.X.); (Y.C.); (Y.G.); (Y.L.)
| | - Sufang Han
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (J.X.); (Y.C.); (Y.G.); (Y.L.)
| |
Collapse
|
2
|
Vaz SR, Tofoli MH, Avelino MAG, da Costa PSS. Probiotics for infantile colic: Is there evidence beyond doubt? A meta-analysis and systematic review. Acta Paediatr 2024; 113:170-182. [PMID: 37962097 DOI: 10.1111/apa.17036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Revised: 10/27/2023] [Accepted: 10/31/2023] [Indexed: 11/15/2023]
Abstract
AIM This study is a systematic review and meta-analysis of randomised controlled trials that employed probiotics and symbiotics for treating infantile colic. METHODS We performed electronic systematic literature searches in Embase, PubMed and Web of Science, to identify articles published between 1950 and April 2023. Only RCT involving infants with infantile colic under 3 months were included. The treatment plan comprised 15 probiotics, which included Lactobacillus reuteri DSM 17938 and Bifidobacterium animalis lactis BB-12. The probiotics were administered alone or in combination with a prebiotic, vs. no intervention or a placebo. RESULTS Probiotics resulted in an average reduction of 51 min of crying per day (p = 0.001). Further analysis of subgroups showed that the reduction was -39.30 min for vaginal delivery (p = 0.003), -64.66 min for Lactobacillus reuteri DSM 17938 (p = 0.03), -40.45 min for other strains (p < 0.00001), -74.28 min for exclusively breastfed infants (p = 0.0003) and -48.04 min for mixed feeding (p < 0.00001). CONCLUSION All probiotic strains seem effective in treating infantile colic. Exclusively breastfed infants have demonstrated more significant reduction in crying time. However, the available evidence on the effectiveness of probiotics in formula-fed and caesarean-born infants is limited.
Collapse
Affiliation(s)
| | - Marise Helena Tofoli
- Department of Pediatric Gastroenterology, State Hospital for Children and Adolescents, Goiânia, Brazil
| | | | | |
Collapse
|
3
|
Hojsak I, Kolaček S. Role of Probiotics in the Treatment and Prevention of Common Gastrointestinal Conditions in Children. Pediatr Gastroenterol Hepatol Nutr 2024; 27:1-14. [PMID: 38249642 PMCID: PMC10796258 DOI: 10.5223/pghn.2024.27.1.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 07/10/2023] [Accepted: 08/30/2023] [Indexed: 01/23/2024] Open
Abstract
Probiotics are live microorganisms that confer health benefits to the host when administered in adequate amounts. Although recommendations for probiotic use should be strain-specific, many systematic reviews, including recommendations from different societies, recommend probiotic use in general, providing no relevant information for healthcare professionals regarding which probiotic to recommend for which clinical indication, at what dose, and for how long. This narrative review aimed to present the available evidence on the use of probiotics in the prevention and treatment of common gastrointestinal diseases in children, considering the strain and dose used. Furthermore, this study summarizes the evidence on the possible side effects and quality of products containing probiotics.
Collapse
Affiliation(s)
- Iva Hojsak
- Department of Pediatrics, Referral Center for Pediatric Gastroenterology and Nutrition, Children’s Hospital, Zagreb, Croatia
- Department of Pediatrics, University of Zagreb School of Medicine, Zagreb, Croatia
- Department of Pediatrics, University J.J. Strossmayer School of Medicine, Osijek, Croatia
| | - Sanja Kolaček
- Department of Pediatrics, Referral Center for Pediatric Gastroenterology and Nutrition, Children’s Hospital, Zagreb, Croatia
- Department of Pediatrics, University of Zagreb School of Medicine, Zagreb, Croatia
| |
Collapse
|
4
|
Zaib S, Hayat A, Khan I. Probiotics and their Beneficial Health Effects. Mini Rev Med Chem 2024; 24:110-125. [PMID: 37291788 DOI: 10.2174/1389557523666230608163823] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Revised: 05/10/2023] [Accepted: 05/17/2023] [Indexed: 06/10/2023]
Abstract
Probiotics are living microorganisms that are present in cultured milk and fermented food. Fermented foods are a rich source for the isolation of probiotics. They are known as good bacteria. They have various beneficial effects on human health including antihypertensive effects, antihypercholesterolemic effects, prevention of bowel disease, and improving the immune system. Microorganisms including bacteria, yeast, and mold are used as probiotics but the major microorganisms that are used as probiotics are bacteria from the genus Lactobacillus, Lactococcus, Streptococcus, and Bifidobacterium. Probiotics are beneficial in the prevention of harmful effects. Recently, the use of probiotics for the treatment of various oral and skin diseases has also gained significant attention. Clinical studies indicate that the usage of probiotics can alter gut microbiota composition and provoke immune modulation in a host. Due to their various health benefits, probiotics are attaining more interest as a substitute for antibiotics or anti-inflammatory drugs leading to the growth of the probiotic market.
Collapse
Affiliation(s)
- Sumera Zaib
- Department of Basic and Applied Chemistry, Faculty of Sciences and Technology, University of Central Punjab, Lahore, 54590, Pakistan
| | - Aqsa Hayat
- Department of Basic and Applied Chemistry, Faculty of Sciences and Technology, University of Central Punjab, Lahore, 54590, Pakistan
| | - Imtiaz Khan
- Department of Chemistry, Manchester Institute of Biotechnology, The University of Manchester, 131, Princess Street, Manchester M1 7DN, United Kingdom
| |
Collapse
|
5
|
Grenov B. Do probiotics have a role in treatment of diarrhea among children with severe acute malnutrition? Am J Clin Nutr 2023; 117:842-843. [PMID: 37137613 DOI: 10.1016/j.ajcnut.2023.03.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 03/02/2023] [Accepted: 03/03/2023] [Indexed: 05/05/2023] Open
Affiliation(s)
- Benedikte Grenov
- Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
6
|
Horvathova K, Modrackova N, Splichal I, Splichalova A, Amin A, Ingribelli E, Killer J, Doskocil I, Pechar R, Kodesova T, Vlkova E. Defined Pig Microbiota with a Potential Protective Effect against Infection with Salmonella Typhimurium. Microorganisms 2023; 11:microorganisms11041007. [PMID: 37110429 PMCID: PMC10146858 DOI: 10.3390/microorganisms11041007] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 04/04/2023] [Accepted: 04/07/2023] [Indexed: 04/29/2023] Open
Abstract
A balanced microbiota is a main prerequisite for the host's health. The aim of the present work was to develop defined pig microbiota (DPM) with the potential ability to protect piglets against infection with Salmonella Typhimurium, which causes enterocolitis. A total of 284 bacterial strains were isolated from the colon and fecal samples of wild and domestic pigs or piglets using selective and nonselective cultivation media. Isolates belonging to 47 species from 11 different genera were identified by MALDI-TOF mass spectrometry (MALDI-TOF MS). The bacterial strains for the DPM were selected for anti-Salmonella activity, ability to aggregate, adherence to epithelial cells, and to be bile and acid tolerant. The selected combination of 9 strains was identified by sequencing of the 16S rRNA gene as Bacillus sp., Bifidobacterium animalis subsp. lactis, B. porcinum, Clostridium sporogenes, Lactobacillus amylovorus, L. paracasei subsp. tolerans, Limosilactobacillus reuteri subsp. suis, and Limosilactobacillus reuteri (two strains) did not show mutual inhibition, and the mixture was stable under freezing for at least 6 months. Moreover, strains were classified as safe without pathogenic phenotype and resistance to antibiotics. Future experiments with Salmonella-infected piglets are needed to test the protective effect of the developed DPM.
Collapse
Affiliation(s)
- Kristyna Horvathova
- Department of Microbiology, Nutrition and Dietetics, Faculty of Agrobiology, Food and Natural Resources, Czech University of Life Sciences Prague, 165 00 Prague, Czech Republic
| | - Nikol Modrackova
- Department of Microbiology, Nutrition and Dietetics, Faculty of Agrobiology, Food and Natural Resources, Czech University of Life Sciences Prague, 165 00 Prague, Czech Republic
| | - Igor Splichal
- Laboratory of Gnotobiology, Institute of Microbiology, Czech Academy of Sciences, 549 22 Novy Hradek, Czech Republic
| | - Alla Splichalova
- Laboratory of Gnotobiology, Institute of Microbiology, Czech Academy of Sciences, 549 22 Novy Hradek, Czech Republic
| | - Ahmad Amin
- Department of Microbiology, Nutrition and Dietetics, Faculty of Agrobiology, Food and Natural Resources, Czech University of Life Sciences Prague, 165 00 Prague, Czech Republic
| | - Eugenio Ingribelli
- Department of Microbiology, Nutrition and Dietetics, Faculty of Agrobiology, Food and Natural Resources, Czech University of Life Sciences Prague, 165 00 Prague, Czech Republic
| | - Jiri Killer
- Department of Microbiology, Nutrition and Dietetics, Faculty of Agrobiology, Food and Natural Resources, Czech University of Life Sciences Prague, 165 00 Prague, Czech Republic
- Laboratory of Anaerobic Microbiology, Institute of Animal Physiology and Genetics, Czech Academy of Sciences, 142 20 Prague, Czech Republic
| | - Ivo Doskocil
- Department of Microbiology, Nutrition and Dietetics, Faculty of Agrobiology, Food and Natural Resources, Czech University of Life Sciences Prague, 165 00 Prague, Czech Republic
| | - Radko Pechar
- Department of Microbiology, Nutrition and Dietetics, Faculty of Agrobiology, Food and Natural Resources, Czech University of Life Sciences Prague, 165 00 Prague, Czech Republic
| | - Tereza Kodesova
- Department of Microbiology, Nutrition and Dietetics, Faculty of Agrobiology, Food and Natural Resources, Czech University of Life Sciences Prague, 165 00 Prague, Czech Republic
| | - Eva Vlkova
- Department of Microbiology, Nutrition and Dietetics, Faculty of Agrobiology, Food and Natural Resources, Czech University of Life Sciences Prague, 165 00 Prague, Czech Republic
| |
Collapse
|
7
|
Mazur M, Tomczak H, Łodyga M, Plagens-Rotman K, Merks P, Czarnecka-Operacz M. The Intestinal and Skin Microbiome in Patients with Atopic Dermatitis and Their Influence on the Course of the Disease: A Literature Review. Healthcare (Basel) 2023; 11:healthcare11050766. [PMID: 36900771 PMCID: PMC10001192 DOI: 10.3390/healthcare11050766] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 02/22/2023] [Accepted: 02/27/2023] [Indexed: 03/08/2023] Open
Abstract
Bacteria inhabiting the digestive tract are responsible for our health. The microbiome is essential for the development of the immune system and homeostasis of the body. Maintaining homeostasis is very important, but also extremely complicated. The gut microbiome is related to the skin microbiome. It can therefore be assumed that changes in the microbes inhabiting the skin are greatly influenced by the bacteria living in the intestines. Changes in the composition and function of microbes (dysbiosis in the skin and intestines) have recently been linked to changes in the immune response and the development of skin diseases, including atopic dermatitis (AD). This review was compiled by collaborating Dermatologists specializing in atopic dermatitis and psoriasis. A comprehensive review of the current literature was performed using PubMed and limited to relevant case reports and original papers on the skin microbiome in atopic dermatitis. The inclusion criterion was that the paper was published in a peer-reviewed journal in the last 10 years (2012-2022). No limitations on the language of the publication or the type of study were made. It has been shown that any rapid changes in the composition of the microflora may be associated with the appearance of clinical signs and symptoms of the disease. Various studies have proven that the microbiome of many systems (including the intestines) may have a significant impact on the development of the inflammatory process within the skin in the course of AD. It has been shown that an early interaction between the microbiome and immune system may result in a noticeable delay in the onset of atopic diseases. It seems to be of high importance for physicians to understand the role of the microbiome in AD, not only from the pathophysiological standpoint but also in terms of the complex treatment that is required. Perhaps young children diagnosed with AD present specific characteristics of the intestinal microflora. This might be related to the early introduction of antibiotics and dietary manipulations in breastfeeding mothers in the early childhood of AD patients. It is most likely related to the abuse of antibiotics from the first days of life.
Collapse
Affiliation(s)
- Małgorzata Mazur
- College of Health, Beauty Care and Education in Poznań, 60-133 Poznań, Poland
| | - Hanna Tomczak
- Central Microbiological Laboratory, H. Święcicki Clinical Hospital at the Poznan University of Medical Sciences, 60-366 Poznan, Poland
| | - Martha Łodyga
- Department of Medicine Berkshire Medical Center, Pittsfield, MA 01201, USA
| | - Katarzyna Plagens-Rotman
- Center for Pediatric, Adolescent Gynecology and Sexology Division of Gynecology, Department of Perinatology and Gynecology, Poznan University of Medical Sciences, 61-758 Poznan, Poland
- Correspondence:
| | - Piotr Merks
- Department of Pharmacology and Clinical Pharmacology, Faculty of Medicine, Collegium Medicum, Cardinal Stefan Wyszyński University, 01-938 Warszawa, Poland
| | - Magdalena Czarnecka-Operacz
- Allergic and Occupational Skin Diseases Unit, Department of Dermatology, Medical University of Poznań, 60-355 Poznan, Poland
| |
Collapse
|
8
|
Probiotics for the Management of Pediatric Gastrointestinal Disorders: Position Paper of the ESPGHAN Special Interest Group on Gut Microbiota and Modifications. J Pediatr Gastroenterol Nutr 2023; 76:232-247. [PMID: 36219218 DOI: 10.1097/mpg.0000000000003633] [Citation(s) in RCA: 48] [Impact Index Per Article: 48.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
BACKGROUND Probiotics, defined as live microorganisms that, when administered in adequate amounts, confer a health benefit on the host, are widely used despite uncertainty regarding their efficacy and discordant recommendations about their use. The European Society for Paediatric Gastroenterology, Hepatology and Nutrition (ESPGHAN) Special Interest Group on Gut Microbiota and Modifications provides updated recommendations for the use of probiotics for the management of selected pediatric gastrointestinal disorders. METHODS All systematic reviews and/or meta-analyses, as well as subsequently published randomized controlled trials (RCTs) (until December 2021), that compared the use of probiotics in all delivery vehicles and formulations, at any dose, with no probiotic (ie, placebo or no treatment), were eligible for inclusion. The recommendations were formulated only if at least 2 RCTs on a similar well-defined probiotic strain were available. The modified Delphi process was used to establish consensus on the recommendations. RESULTS Recommendations for the use of specific probiotic strains were made for the management of acute gastroenteritis, prevention of antibiotic-associated diarrhea, nosocomial diarrhea and necrotizing enterocolitis, management of Helicobacter pylori infection, and management of functional abdominal pain disorders and infant colic. CONCLUSIONS Despite evidence to support the use of specific probiotics in some clinical situations, further studies confirming the effect(s) and defining the type, dose, and timing of probiotics are still often required. The use of probiotics with no documented health benefits should be discouraged.
Collapse
|
9
|
Mazziotta C, Tognon M, Martini F, Torreggiani E, Rotondo JC. Probiotics Mechanism of Action on Immune Cells and Beneficial Effects on Human Health. Cells 2023; 12:cells12010184. [PMID: 36611977 PMCID: PMC9818925 DOI: 10.3390/cells12010184] [Citation(s) in RCA: 119] [Impact Index Per Article: 119.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 12/12/2022] [Accepted: 12/29/2022] [Indexed: 01/03/2023] Open
Abstract
Immune cells and commensal microbes in the human intestine constantly communicate with and react to each other in a stable environment in order to maintain healthy immune activities. Immune system-microbiota cross-talk relies on a complex network of pathways that sustain the balance between immune tolerance and immunogenicity. Probiotic bacteria can interact and stimulate intestinal immune cells and commensal microflora to modulate specific immune functions and immune homeostasis. Growing evidence shows that probiotic bacteria present important health-promoting and immunomodulatory properties. Thus, the use of probiotics might represent a promising approach for improving immune system activities. So far, few studies have been reported on the beneficial immune modulatory effect of probiotics. However, many others, which are mainly focused on their metabolic/nutritional properties, have been published. Therefore, the mechanisms behind the interaction between host immune cells and probiotics have only been partially described. The present review aims to collect and summarize the most recent scientific results and the resulting implications of how probiotic bacteria and immune cells interact to improve immune functions. Hence, a description of the currently known immunomodulatory mechanisms of probiotic bacteria in improving the host immune system is provided.
Collapse
Affiliation(s)
- Chiara Mazziotta
- Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy
- Center for Studies on Gender Medicine, Department of Medical Sciences, University of Ferrara, 64/b, Fossato di Mortara Street, 44121 Ferrara, Italy
| | - Mauro Tognon
- Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy
| | - Fernanda Martini
- Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy
- Center for Studies on Gender Medicine, Department of Medical Sciences, University of Ferrara, 64/b, Fossato di Mortara Street, 44121 Ferrara, Italy
- Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, 44121 Ferrara, Italy
| | - Elena Torreggiani
- Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy
- Correspondence: (E.T.); (J.C.R.); Tel.: +39-053-2455-557 (E.T.); +39-053-245-5536 (J.C.R.)
| | - John Charles Rotondo
- Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy
- Center for Studies on Gender Medicine, Department of Medical Sciences, University of Ferrara, 64/b, Fossato di Mortara Street, 44121 Ferrara, Italy
- Correspondence: (E.T.); (J.C.R.); Tel.: +39-053-2455-557 (E.T.); +39-053-245-5536 (J.C.R.)
| |
Collapse
|
10
|
Hojsak I, Kolaček S, Mihatsch W, Mosca A, Shamir R, Szajewska H, Vandenplas Y. Synbiotics in the Management of Pediatric Gastrointestinal Disorders: Position Paper of the ESPGHAN Special Interest Group on Gut Microbiota and Modifications. J Pediatr Gastroenterol Nutr 2023; 76:102-108. [PMID: 35900172 DOI: 10.1097/mpg.0000000000003568] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
BACKGROUND Synbiotics are a mixture comprising of live microorganisms and substrate(s) selectively utilized by host microorganisms that confers a health benefit on the host. There is an increasing number of studies investigating their role in different diseases and disorders. AIM The purpose of this article is to provide recommendations for the use of synbiotics in the management of pediatric gastrointestinal disorders. The recommendations are developed by the ESPGHAN Special Interest Group on Gut Microbiota and Modifications. METHODS From existing literature databases, we searched and appraised all systematic reviews and/or meta-analyses, and subsequently published randomized controlled trials (RCTs) that compared the use of synbiotics, in all delivery vehicles and formulations, at any dose, compared to no synbiotics. Synbiotics which are part of infant formula were not assessed. The recommendations were formulated only if at least 2 RCTs that used a well-defined synbiotic were available. RESULTS Based on the currently available evidence, no recommendation can be formulated in favor or against the use of evaluated synbiotic combination in the treatment of acute gastroenteritis, prevention of necrotizing enterocolitis, Helicobacter pylori infection, inflammatory bowel disease, functional gastrointestinal disorders, and allergy in infants and children. CONCLUSIONS There is a need for more, well-designed RCTs on the role of synbiotics in gastrointestinal disorders with the same outcome measures to enable the inter-studies comparisons.
Collapse
Affiliation(s)
- Iva Hojsak
- From the Children's Hospital Zagreb, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Sanja Kolaček
- From the Children's Hospital Zagreb, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Walter Mihatsch
- the Department of Pediatrics Ulm University, Ulm, and Neu-Ulm University of Applied Sciences, Neu-Ulm, Germany
| | - Alexis Mosca
- the Hôpital Robert Debré, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Raanan Shamir
- the Institute for Gastroenterology, Nutrition and Liver Diseases, Schneider Children's, Medical Center, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Hania Szajewska
- the Department of Paediatrics, The Medical University of Warsaw, Warsaw, Poland
| | - Yvan Vandenplas
- the Vrije Universiteit Brussel (VUB), UZ Brussel, Kidz Health Castle, Brussels, Belgium
| |
Collapse
|
11
|
Danis R, Mego M, Antonova M, Stepanova R, Svobodnik A, Hejnova R, Wawruch M. Orally Administered Probiotics in the Prevention of Chemotherapy (± Radiotherapy)-Induced Gastrointestinal Toxicity: A Systematic Review With Meta-Analysis of Randomized Trials. Integr Cancer Ther 2022; 21:15347354221144309. [PMID: 36567453 PMCID: PMC9806400 DOI: 10.1177/15347354221144309] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Chemoradiotherapy-induced gastrointestinal toxicity may lead to a significant impairment of the oncological patient's quality of life, as well as to reduced adherence to the treatment, which may have a negative impact on survival and mortality rates. OBJECTIVE The aim of this review was to investigate whether oral probiotic administration prevents chemotherapy (± radiotherapy)-induced gastrointestinal toxicity, particularly diarrhea. METHODS We searched the MEDLINE, Web of Science, and SCOPUS databases for randomized controlled trials in English published between 1990 and 2020. We conducted statistical data analyses expressing the treatment effect size as a risk ratio (RR) together with a 95% confidence interval (CI). Implications are based on trials rated as having a low risk of bias (RoB). RESULTS We included 8 trials (n = 697 participants), from which 3 studies rated as low RoB contained primary endpoint data; the risk of developing grade 3/4 diarrhea in patients receiving probiotics was reduced by 78% compared to the control group (RR = 0.22 [95% CI 0.05-1.08]; P = .06; n = 114 participants). Probiotics showed preventive effects in patients treated with chemotherapy alone (RR = 0.34 [0.12-0.94]; P = .04, n = 121 participants) and in patients with colorectal cancer (RR = 0.56 [0.34-0.92]; P = .02; n = 208 participants). The reduction in the incidence of overall diarrhea was not significant. CONCLUSIONS Probiotics failed to prove a preventive effect of statistical significance against the development of severe and overall diarrhea in cancer patients treated with chemotherapy (± radiotherapy). However, we cannot rule out that the effects of probiotics are clinically relevant, especially in certain subgroups of patients. This needs to be clarified in further well-performed studies.
Collapse
Affiliation(s)
- Radoslav Danis
- Comenius University in Bratislava
Faculty of Medicine, Bratislava, Slovakia,Radoslav Danis, Comenius University in
Bratislava Faculty of Medicine, Spitalska 24, Bratislava 813 72, Slovakia.
| | - Michal Mego
- Comenius University in Bratislava
Faculty of Medicine, Bratislava, Slovakia,National Cancer Institute, Bratislava,
Slovak Republic
| | | | | | | | - Renata Hejnova
- Masaryk University Faculty of Medicine,
Brno, Czech Republic
| | - Martin Wawruch
- Comenius University in Bratislava
Faculty of Medicine, Bratislava, Slovakia
| |
Collapse
|
12
|
The Antimicrobial Effect of Various Single-Strain and Multi-Strain Probiotics, Dietary Supplements or Other Beneficial Microbes against Common Clinical Wound Pathogens. Microorganisms 2022; 10:microorganisms10122518. [PMID: 36557771 PMCID: PMC9781324 DOI: 10.3390/microorganisms10122518] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 12/09/2022] [Accepted: 12/13/2022] [Indexed: 12/24/2022] Open
Abstract
The skin is the largest organ in the human body and is colonized by a diverse microbiota that works in harmony to protect the skin. However, when skin damage occurs, the skin microbiota is also disrupted, and pathogens can invade the wound and cause infection. Probiotics or other beneficial microbes and their metabolites are one possible alternative treatment for combating skin pathogens via their antimicrobial effectiveness. The objective of our study was to evaluate the antimicrobial effect of seven multi-strain dietary supplements and eleven single-strain microbes that contain probiotics against 15 clinical wound pathogens using the agar spot assay, co-culturing assay, and agar well diffusion assay. We also conducted genera-specific and species-specific molecular methods to detect the DNA in the dietary supplements and single-strain beneficial microbes. We found that the multi-strain dietary supplements exhibited a statistically significant higher antagonistic effect against the challenge wound pathogens than the single-strain microbes and that lactobacilli-containing dietary supplements and single-strain microbes were significantly more efficient than the selected propionibacteria and bacilli. Differences in results between methods were also observed, possibly due to different mechanisms of action. Individual pathogens were susceptible to different dietary supplements or single-strain microbes. Perhaps an individual approach such as a 'probiogram' could be a possibility in the future as a method to find the most efficient targeted probiotic strains, cell-free supernatants, or neutralized cell-free supernatants that have the highest antagonistic effect against individual clinical wound pathogens.
Collapse
|
13
|
Conrad LA, LeCroy MN, Rothschild E, Rodgers CRR, Cabana MD. Effect of Early Infant Lactobacillus rhamnosus GG Supplementation in Preventing Viral Respiratory Illness. Clin Pediatr (Phila) 2022; 62:485-491. [PMID: 36401501 DOI: 10.1177/00099228221137427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The role of early probiotic supplementation in infants for the prevention of respiratory viral illnesses is unclear. We examined the association of Lactobacillus rhamnosus GG (LGG) supplementation during the first 6 months of life with the frequency and severity of viral illnesses during the first 24 months of life. We conducted a secondary analysis of data from the randomized controlled Trial of Infant Probiotic Supplementation (n = 184). Parents reported the number of respiratory viral illnesses, and a composite severity score was created based on symptoms. A negative binomial regression model and a mixed-effects linear regression model assessed for differences in the number of episodes and severity of episodes between treatment groups, respectively. There was no significant difference in the incidence rate of viral illness episodes or symptom severity between treatment groups. Daily supplementation with LGG in early infancy does not decrease the number or severity of viral illnesses during the first 2 years of life.
Collapse
Affiliation(s)
- Laura A Conrad
- Department of Pediatrics, The Children's Hospital at Montefiore and Albert Einstein College of Medicine, Bronx, NY, USA
| | - Madison N LeCroy
- Department of Pediatrics, The Children's Hospital at Montefiore and Albert Einstein College of Medicine, Bronx, NY, USA
| | - Evin Rothschild
- Department of Pediatrics, The Children's Hospital at Montefiore and Albert Einstein College of Medicine, Bronx, NY, USA
| | - Caryn R R Rodgers
- Department of Pediatrics, The Children's Hospital at Montefiore and Albert Einstein College of Medicine, Bronx, NY, USA
| | - Michael D Cabana
- Department of Pediatrics, The Children's Hospital at Montefiore and Albert Einstein College of Medicine, Bronx, NY, USA
| |
Collapse
|
14
|
Dekker J, Quilter M, Qian H. Comparison of two probiotics in follow-on formula: Bifidobacterium animalis subsp. lactis HN019 reduced upper respiratory tract infections in Chinese infants. Benef Microbes 2022; 13:341-354. [PMID: 36004715 DOI: 10.3920/bm2022.0041] [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: 11/19/2022]
Abstract
A randomised, double-blind, placebo-controlled trial was performed to investigate the health benefits of probiotic bacteria in infants when delivered in a follow-on infant formula. The study was conducted in Fuyang (Anhui Province, China) during winter and enrolled 192 healthy infants aged six to 12 months. Infants received one of three follow-on formulae daily for 12 weeks: supplemented with 106 cfu/g Bifidobacterium animalis subsp. lactis HN019 (n=64); 106 cfu/g Lacticaseibacillus rhamnosus HN001 (n=64); or without added probiotics (n=64). The primary endpoint was physician-confirmed bacterial or viral infections during the treatment period. Secondary endpoints included parentally reported (confirmed and unconfirmed) infections; antiviral or antibiotic treatments, and hospitalisation; stool frequency and consistency; infant growth; infant temperament; and adverse events. There were 8 cases of confirmed infection, all upper respiratory tract infections (URTIs). Confirmed URTIs were observed in 9.4% of the control group, compared to 3.1% in the HN001 group (P=0.273), and 0.0% in the HN019 group (P=0.028). A similar trend was observed for parentally reported URTIs, with 25.0% in the control group, compared with 14.1% in the HN001 group (P=0.119) and 9.4% in the HN019 group (P=0.019). No infants in the HN019 group were prescribed antibiotics or antivirals, compared with 3 (4.7%) in the HN001 group and 7 (10.9%) in the control group. No infants required hospitalisation. The probiotic-containing formulae were well-tolerated: there were no cases of diarrhoea or differences in stool frequency or characteristics, no differences in infant growth or temperament, and no treatment-related adverse events. This study directly compared the benefits of two different probiotics when added to follow-on infant formula at 106 cfu/g and consumed over a 12-week period. While HN001 showed trends toward reduced infections, HN019 showed better performance in terms of significantly reduced incidence of both physician-confirmed and parentally reported URTIs, and antibiotic/antiviral use compared to a control in Chinese infants. The trial is registered at ClinicalTrials.gov (NCT01724203).
Collapse
Affiliation(s)
- J Dekker
- Fonterra Research and Development Centre, Palmerston North, Private Bag 11029, 4442 Palmerston North, New Zealand
| | - M Quilter
- Fonterra Research and Development Centre, Palmerston North, Private Bag 11029, 4442 Palmerston North, New Zealand
| | - H Qian
- School of Public Health, Anhui Medical University, Hefei, China P.R
| |
Collapse
|
15
|
National Consensus for the Management of Acute Gastroenteritis in Jordanian Children: Consensus Recommendations Endorsed by the Jordanian Paediatric Society. Int J Pediatr 2022; 2022:4456232. [PMID: 36082204 PMCID: PMC9448628 DOI: 10.1155/2022/4456232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Accepted: 06/15/2022] [Indexed: 11/22/2022] Open
Abstract
Diarrhoeal diseases are one of the leading worldwide preventable causes of death among children under 5 years of age. Almost half of children do not receive optimal acute gastroenteritis (AGE) treatment in Jordan. With neither regional nor local guidelines available for AGE, consensus recommendations on the management of paediatric AGE in Jordan were developed by a panel of senior paediatricians and paediatric gastroenterologists and are endorsed by the Jordanian Paediatric Society. Recommendations are based on international guidelines and available relevant literature in relation to the AGE landscape and the healthcare system in Jordan. The prevention of diarrhoeal diseases should focus on the improvement of nutrition, hygiene, and sanitation, the introduction of routine vaccination against rotavirus, and the adoption of a standardised approach for AGE management (oral rehydration solution (ORS) use±adjunct therapies, continued feeding, and avoiding routine antibiotic use). Ondansetron, diosmectite, racecadotril, probiotics, and zinc can be considered adjunct to ORS, if needed. Local data gaps should be addressed. The clinical algorithm for the management of paediatric AGE could promote adherence to practice recommendations and by extension improve health outcomes in children.
Collapse
|
16
|
Indian Academy of Pediatrics Consensus Guidelines for Probiotic Use in Childhood Diarrhea. Indian Pediatr 2022. [DOI: 10.1007/s13312-022-2557-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
17
|
Nayebi A, Navashenaq JG, Soleimani D, Nachvak SM. Probiotic supplementation: A prospective approach in the treatment of COVID-19. Nutr Health 2022; 28:163-175. [PMID: 34747257 PMCID: PMC9160438 DOI: 10.1177/02601060211049631] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Background: Despite strategies based on social distancing, the coronavirus disease 2019 (COVID-19) expands globally, and so far, many attempts have been made to achieve effective treatment for patients with COVID-19. This disease infects the lower respiratory tract and may lead to severe acute respiratory syndrome coronavirus (SARS-CoV). COVID-19 also can cause gastrointestinal infections. Therefore, COVID-19 patients with gastrointestinal symptoms are more likely to be complicated by SARS-CoV. In this disease, acquired immune responses are impaired, and uncontrolled inflammatory responses result in cytokine storms, leading to acute lung injury and thrombus formation. Probiotics are living microorganisms that contribute to the health of the host if administered in appropriate doses. Aim: This study aimed to provide evidence to show the importance of gut dysbiosis in viral disease, especially COVID-19. Therefore, we have focused on the impact of probiotics consumption on preventing severe symptoms of the disease. Methods: We have entirely searched SCOPUS, PubMed, and Google Scholar databases to collect evidence regarding the relationship between probiotics and viral infections to expand this relationship to the COVID-19. Results: It has been shown that probiotics directly counteract SARS-CoV in the gastrointestinal and respiratory tracts. Moreover, probiotics suppress severe immune responses and prevent cytokine storms to inhibit pathologic inflammatory conditions in the body via modulation of immune responses. Conclusion: According to available evidence based on their antiviral and respiratory activities, using probiotics might be an adjuvant therapy to reduce the burden and severity of this disease.
Collapse
Affiliation(s)
- Atiyeh Nayebi
- Student Research Committee, Nutritional Sciences Department, School of Nutrition Sciences and Food Technology, Kermanshah University of Medical Sciences, Kermanshah, Iran
- Nutritional Sciences Department, School of Nutrition Sciences and Food Technology, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | | | - Davood Soleimani
- Student Research Committee, Nutritional Sciences Department, School of Nutrition Sciences and Food Technology, Kermanshah University of Medical Sciences, Kermanshah, Iran
- Research Center of Oils and Fats, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Seyyed Mostafa Nachvak
- Student Research Committee, Nutritional Sciences Department, School of Nutrition Sciences and Food Technology, Kermanshah University of Medical Sciences, Kermanshah, Iran
- Nutritional Sciences Department, School of Nutrition Sciences and Food Technology, Kermanshah University of Medical Sciences, Kermanshah, Iran
| |
Collapse
|
18
|
Danis R, Wawruch M. Travellers' diarrhoea - prevention, trends and role of microbiome. Cent Eur J Public Health 2022; 30:20-25. [PMID: 35421294 DOI: 10.21101/cejph.a6740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Accepted: 02/15/2022] [Indexed: 11/15/2022]
Abstract
OBJECTIVES In this review, we present a contemporary look at the management of travellers' diarrhoea (TD), and we discuss the potential role of a microbiome as well as the administration of live microorganisms in order to prevent TD. METHODS We performed a comprehensive search using the PubMed and Web of Science databases for the period 2014-2021, looking for original and review articles on travellers' diarrhoea. RESULTS TD belongs among the most frequent illnesses experienced by travellers. For the most part, it is manifested as an acute yet self limiting condition, and only in a few cases proceeds to a prolonged form. Epidemiological analyses have shown that the majority of TD cases are caused by bacterial infections. In practice, pharmacological therapy is often used in the prevention and treatment of TD, since patients naturally seek preventive measures against the development of its severe course and its impact on planned activities. Bismuth salicylate is a strongly recommended TD prophylaxis but is not available on all European Union markets. Although the antibiotic prophylaxis is not generally recommended in guidelines, some antibiotic or chemotherapeutic agents are accessible over-the-counter in certain countries, and travellers are routinely encouraged to use them preventively. This routine can alter the microbiome of the traveller and promote the spread of drug resistant bacteria in their place of residence. Probiotic administration is considered safe, although the quality of evidence in favour of its prophylactic use in TD is currently low. CONCLUSIONS The challenge for public health authorities is to educate personnel that can directly influence the behaviour of travellers through safe and effective pharmacological alternatives to antibiotics. Manipulation of the gut microbiome using specific probiotic strains can represent a safe and promising intervention.
Collapse
Affiliation(s)
- Radoslav Danis
- Institute of Pharmacology and Clinical Pharmacology, Faculty of Medicine, Comenius University, Bratislava, Slovak Republic
| | - Martin Wawruch
- Institute of Pharmacology and Clinical Pharmacology, Faculty of Medicine, Comenius University, Bratislava, Slovak Republic
| |
Collapse
|
19
|
Huang R, Wu F, Zhou Q, Wei W, Yue J, Xiao B, Luo Z. Lactobacillus and intestinal diseases: mechanisms of action and clinical applications. Microbiol Res 2022; 260:127019. [DOI: 10.1016/j.micres.2022.127019] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 03/27/2022] [Accepted: 03/29/2022] [Indexed: 12/12/2022]
|
20
|
Jeon S, Kim H, Kim J, Seol D, Jo J, Choi Y, Cho S, Kim H. Positive Effect of Lactobacillus acidophilus EG004 on Cognitive Ability of Healthy Mice by Fecal Microbiome Analysis Using Full-Length 16S-23S rRNA Metagenome Sequencing. Microbiol Spectr 2022; 10:e0181521. [PMID: 35019699 PMCID: PMC8754107 DOI: 10.1128/spectrum.01815-21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 12/07/2021] [Indexed: 11/20/2022] Open
Abstract
Evidence for the concept of the "gut-brain axis" (GBA) has risen. Many types of research demonstrated the mechanism of the GBA and the effect of probiotic intake. Although many studies have been reported, most were focused on neurodegenerative disease and, it is still not clear what type of bacterial strains have positive effects. We designed an experiment to discover a strain that positively affects brain function, which can be recognized through changes in cognitive processes using healthy mice. The experimental group consisted of a control group and three probiotic consumption groups, namely, Lactobacillus acidophilus, Lacticaseibacillus paracasei, and Lacticaseibacillus rhamnosus. Three experimental groups fed probiotics showed an improved cognitive ability by cognitive-behavioral tests, and the group fed on L. acidophilus showed the highest score. To provide an understanding of the altered microbial composition effect on the brain, we performed full 16S-23S rRNA sequencing using Nanopore, and operational taxonomic units (OTUs) were identified at species level. In the group fed on L. acidophilus, the intestinal bacterial ratio of Firmicutes and Proteobacteria phyla increased, and the bacterial proportions of 16 species were significantly different from those of the control group. We estimated that the positive results on the cognitive behavioral tests were due to the increased proportion of the L. acidophilus EG004 strain in the subjects' intestines since the strain can produce butyrate and therefore modulate neurotransmitters and neurotrophic factors. We expect that this strain expands the industrial field of L. acidophilus and helps understand the mechanism of the gut-brain axis. IMPORTANCE Recently, the concept of the "gut-brain axis" has risen and suggested that microbes in the GI tract affect the brain by modulating signal molecules. Although many pieces of research were reported in a short period, a signaling mechanism and the effects of a specific bacterial strain are still unclear. Besides, since most of the research was focused on neurodegenerative disease, the study with a healthy animal model is still insufficient. In this study, we show using a healthy animal model that a bacterial strain (Lactobacillus acidophilus EG004) has a positive effect on mouse cognitive ability. We experimentally verified an improved cognitive ability by cognitive behavioral tests. We performed full 16S-23S rRNA sequencing using a Nanopore MinION instrument and provided the gut microbiome composition at the species level. This microbiome composition consisted of candidate microbial groups as a biomarker that shows positive effects on cognitive ability. Therefore, our study suggests a new perspective for probiotic strain use applicable for various industrialization processes.
Collapse
Affiliation(s)
- Soomin Jeon
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, Republic of Korea
| | - Hyaekang Kim
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, Republic of Korea
| | - Jina Kim
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, Republic of Korea
| | - Donghyeok Seol
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, Republic of Korea
- eGnome, Inc., Seoul, Republic of Korea
| | - Jinchul Jo
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, Republic of Korea
| | - Youngseok Choi
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, Republic of Korea
| | - Seoae Cho
- eGnome, Inc., Seoul, Republic of Korea
| | - Heebal Kim
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, Republic of Korea
- eGnome, Inc., Seoul, Republic of Korea
- Interdisciplinary Program in Bioinformatics, Seoul National University, Seoul, Republic of Korea
| |
Collapse
|
21
|
Myojin S, Pak K, Sako M, Kobayashi T, Takahashi T, Sunagawa T, Tsuboi N, Ishikura K, Kubota M, Kubota M, Igarashi T, Morioka I, Miyairi I. Interventions for Shiga toxin-producing Escherichia coli gastroenteritis and risk of hemolytic uremic syndrome: A population-based matched case control study. PLoS One 2022; 17:e0263349. [PMID: 35120154 PMCID: PMC8815883 DOI: 10.1371/journal.pone.0263349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 01/18/2022] [Indexed: 11/25/2022] Open
Abstract
Background The role of antibiotics in the treatment of Shiga toxin-producing Escherichia coli (STEC) infection is controversial. Objectives To evaluate the association between treatment (antibiotics, antidiarrheal agents, and probiotics) for STEC infection and hemolytic uremic syndrome (HUS) development. Patients and methods We performed a population-based matched case-control study using the data from the National Epidemiological Surveillance of Infectious Diseases (NESID) between January 1, 2017 and December 31, 2018. We identified all patients with STEC infection and HUS as cases and matched patients with STEC infection without HUS as controls, with a case-control a ratio of 1:5. Further medical information was obtained by a standardized questionnaire. Multivariable conditional logistic regression model was used. Results 7760 patients with STEC infection were registered in the NESID. 182 patients with HUS and 910 matched controls without HUS were selected. 90 patients with HUS (68 children and 22 adults) and 371 patients without HUS (266 children and 105 adults) were included in the main analysis. The matched ORs of any antibiotics and fosfomycin for HUS in children were 0.56 (95% CI 0.32–0.98), 0.58 (0.34–1.01). The matched ORs for HUS were 2.07 (1.07–4.03), 0.86 (0.46−1.61) in all ages treated with antidiarrheal agent and probiotics. Conclusions Antibiotics, especially fosfomycin, may prevent the development of HUS in children, while use of antidiarrheal agents should be avoided.
Collapse
Affiliation(s)
- Shota Myojin
- Division of Infectious Diseases, Department of Medical Subspecialties, National Center for Child Health and Development, Tokyo, Japan
- Department of Pediatrics and Child Health, Nihon University School of Medicine, Tokyo, Japan
| | - Kyongsun Pak
- Biostatistics Unit, Department of Data Science, Clinical Research Center, National Center for Child Health and Development, Tokyo, Japan
| | - Mayumi Sako
- Department of Clinical Research Promotion, Clinical Research Center, National Center for Child Health and Development, Tokyo, Japan
| | - Tohru Kobayashi
- Department of Data Science, Clinical Research Center, National Center for Child Health and Development, Tokyo, Japan
| | - Takuri Takahashi
- Infectious Disease Surveillance Center, National Institute of Infectious Diseases, Tokyo, Japan
| | - Tomimasa Sunagawa
- Infectious Disease Surveillance Center, National Institute of Infectious Diseases, Tokyo, Japan
| | - Norihiko Tsuboi
- Division of Critical Care Medicine, Department of Critical Care and Anesthesia, National Center for Child Health and Development, Tokyo, Japan
| | - Kenji Ishikura
- Department of Pediatrics, Kitasato University School of Medicine, Kanagawa, Japan
| | - Masaya Kubota
- Division of Neurology, Department of Medical Subspecialties, National Center for Child Health and Development, Tokyo, Japan
| | - Mitsuru Kubota
- Department of General Pediatrics & Interdisciplinary Medicine, National Center for Child Health and Development, Tokyo, Japan
| | | | - Ichiro Morioka
- Department of Pediatrics and Child Health, Nihon University School of Medicine, Tokyo, Japan
| | - Isao Miyairi
- Division of Infectious Diseases, Department of Medical Subspecialties, National Center for Child Health and Development, Tokyo, Japan
- Department of Pediatrics, Hamamatsu University School of Medicine, Shizuoka, Japan
- * E-mail:
| |
Collapse
|
22
|
Gueniche A, Liboutet M, Cheilian S, Fagot D, Juchaux F, Breton L. Vitreoscilla filiformis Extract for Topical Skin Care: A Review. Front Cell Infect Microbiol 2022; 11:747663. [PMID: 34976852 PMCID: PMC8717924 DOI: 10.3389/fcimb.2021.747663] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 11/23/2021] [Indexed: 12/13/2022] Open
Abstract
The term probiotic has been defined by experts as live microorganisms, which when administered in adequate amounts, confer a health benefit on the host. Probiotics are, thus, by definition, live microorganisms, and the viability of probiotics is a prerequisite for certain benefits, such as the release of metabolites at the site or adhesion properties, for example. However, some semi-active or non-replicative bacterial preparations may retain a similar activity to the live forms. On cosmetic, lysates or fractions are generally used. Topically applied Vitreoscilla filiformis extract has shown to have some similar biological activity of probiotics in the gut, for example, regulating immunity by optimisation of regulatory cell function, protecting against infection, and helping skin barrier function for better recovery and resistance. Due to their mode of action and efficacy, V. filiformis extract (lysate including membrane and cytosol) may be considered as non-replicative probiotic fractions, and this review article presents all its properties.
Collapse
Affiliation(s)
- Audrey Gueniche
- L'Oreal Research and Innovation, Luxury Division Dept, Chevilly-La-Rue, France
| | - Muriel Liboutet
- L'Oreal Research and Innovation, Luxury Division Dept, Chevilly-La-Rue, France
| | - Stephanie Cheilian
- L'Oreal Research and Innovation, Luxury Division Dept, Chevilly-La-Rue, France
| | - Dominique Fagot
- L'Oreal Research and Innovation, Advanced Research Dept, Aulnay-sous-Bois, France
| | - Franck Juchaux
- L'Oreal Research and Innovation, Advanced Research Dept, Aulnay-sous-Bois, France
| | - Lionel Breton
- L'Oreal Research and Innovation, Advanced Research Dept, Aulnay-sous-Bois, France
| |
Collapse
|
23
|
Jaber RM, Alnshash BM, Qasem NW. Infectious Diseases in Children. Fam Med 2022. [DOI: 10.1007/978-3-030-54441-6_150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
24
|
Michels M, Jesus GFA, Voytena APL, Rossetto M, Ramlov F, Córneo E, Feuser P, Gelain D, Dal-Pizzol F. Immunomodulatory Effect of Bifidobacterium, Lactobacillus, and Streptococcus Strains of Paraprobiotics in Lipopolysaccharide-Stimulated Inflammatory Responses in RAW-264.7 Macrophages. Curr Microbiol 2021; 79:9. [PMID: 34905100 DOI: 10.1007/s00284-021-02708-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Accepted: 10/11/2021] [Indexed: 12/17/2022]
Abstract
The discovery of the potential of paraprobiotics to exert different immunological benefits suggests that further studies should be carried out to determine their potential and mechanisms of action in modulating the immune system. The objective of this study was to investigate the immune response of several microbial-associated molecular patterns (MAMPS) used at different doses in macrophage cell lines RAW-264.7 stimulated with lipopolysaccharide (LPS). Two experiments were conducted. The first was performed to determine a dose response curve for each paraprobiotic (Bifidobacterium lactis, Lactobacillus casei, Lactobacillus gasseri, Lactobacillus paracasei, and Streptococcus thermophilus). Further experiments were carried using only two doses (0.01 g/ml and 0.1 g/ml). RAW-264.7 cells were cultivated in Dubelcco's Modified Eagle's medium supplemented with fetal bovine serum and penicillin/streptomycin. Cells were incubated with LPS (1 μg/ml) and six concentrations of MAMPs were added. RAW-264.7 viability, myeloperoxidase activity, nitrite/nitrate concentration, reactive oxygen species production, oxidative damage, and inflammatory parameters were measured. In the LPS group, there was a significant reduction in cell viability. Myeloperoxidase and nitrite/nitrate concentrations demonstrated a better effect at 0.01 and 0.1 g/ml doses. There was a significant reduction in interleukin-6 (IL-6) levels at 0.1 g/ml dose in all paraprobiotics. IL-10 levels decreased in the LPS group and increased at 0.1 g/ml dose in all paraprobiotics. The dichlorofluorescin diacetate results were reinforced by the observed in oxidative damage. Paraprobiotics are likely to contribute to the improvement of intestinal homeostasis, immunomodulation, and host metabolism.
Collapse
Affiliation(s)
- Monique Michels
- Gabbia Biotechnology, Barra Velha, SC, Brazil. .,Laboratory of Experimental Pathophysiology, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, Brazil. .,Programa de Pós-Graduação em Ciências da Saúde, Universidade do Extremo Sul Catarinense, Avenida Universitária, 1105 - Bairro Universitário, Criciúma, SC, CEP: 888006-000, Brazil.
| | | | | | | | | | - Emily Córneo
- Laboratory of Experimental Pathophysiology, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, Brazil
| | - Paulo Feuser
- Laboratory of Experimental Pathophysiology, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, Brazil
| | - Daniel Gelain
- Departament of Biochemistry, Institute of Basic Health Sciences, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Felipe Dal-Pizzol
- Laboratory of Experimental Pathophysiology, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, Brazil
| |
Collapse
|
25
|
Effectiveness of probiotics and synbiotics in reducing duration of acute infectious diarrhea in pediatric patients in developed countries: a systematic review and meta-analysis. Eur J Pediatr 2021; 180:2907-2920. [PMID: 33825068 DOI: 10.1007/s00431-021-04046-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 03/11/2021] [Accepted: 03/21/2021] [Indexed: 02/07/2023]
Abstract
Acute diarrhea is one of the most frequent causes of doctor visits and hospital admissions for children. Our objective was to evaluate the association between probiotics administration and reduction of acute infectious diarrhea duration in children dwelling in developed countries. Bibliographic databases, gray literature, and reference lists were searched up to September 29, 2019. Double-blind, randomized controlled trials that examined probiotics efficacy in children with acute infectious diarrhea residing in developed countries were included. Data were synthesized by generic inverse variance method using fixed- and random-effects model. Twenty trials met the eligibility criteria (n = 3469 patients) and were included in the qualitative synthesis, and 19 studies in meta-analysis. Twelve trials (n = 840) were assessed as high/unclear risk of bias and eight (n = 2629) as low risk of bias. Comparisons revealed a moderate effectiveness of probiotics in low risk of bias studies (MD = - 13.45 h; 95% CI - 24.26, - 2.62; p = 0.02, Bayesian meta-analysis pooled effect MD = - 0.38, 95% CrI - 2.3, 1.58) and a notable effect in studies with high/unclear risk for bias (MD = - 19.70 h; 95% CI - 28.09, - 11.31; p = 0.0004). In trials of optimal methodological quality (n = 1989), probiotics effect was absent (MD = - 3.32 h; 95% CI - 8.78, 2.13, p = 0.23).Conclusion: Outcomes suggest that probiotics do not demonstrate sufficient clinical impact in reducing diarrhea duration in children in the developed countries.Systematic Review Registration: This review is registered at PROSPERO (ID: CRD42020152966). What is Known: • Probiotics, due to the conflicting study results, are administered without adequate evidence as an adjuvant therapeutic agent for eliminating duration of acute infectious diarrhea in pediatric patients. What is New: • In developed countries, probiotics are demonstrated as ineffective in reducing the duration of acute infectious diarrhea in children.
Collapse
|
26
|
Ghosh A, Sundaram B, Bhattacharya P, Mohanty N, Dheivamani N, Mane S, Acharyya B, Kamale V, Poddar S, Khobragade A, Thomas W, Prabhudesai S, Choudhary A, Mitra M. Effect of Saccharomyces boulardii CNCM-I 3799 and Bacillus subtilis CU-1 on Acute Watery Diarrhea: A Randomized Double-Blind Placebo-Controlled Study in Indian Children. Pediatr Gastroenterol Hepatol Nutr 2021; 24:423-431. [PMID: 34557395 PMCID: PMC8443851 DOI: 10.5223/pghn.2021.24.5.423] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 04/22/2021] [Accepted: 05/20/2021] [Indexed: 11/14/2022] Open
Abstract
PURPOSE To assess the effect of combination probiotic Saccharomyces boulardii CNCM-I 3799 and Bacillus subtilis CU-1 in outpatient management of acute watery diarrhea in children. METHODS A randomized double-blind placebo-controlled study was conducted in 180 participants aged six months to five years with acute mild to moderate diarrhea. All were enrolled from six centers across India and centrally randomized to receive S. boulardii CNCM-I 3799 and B. subtilis CU-1 or a placebo along with oral rehydration salts and zinc supplementation. Each participant was followed up for three months to assess recurrence of diarrhea. RESULTS The mean duration of diarrhea in the probiotic and placebo groups were 54.16 hours and 59.48 hours, respectively. The difference in the duration of diarrhea in those administered with probiotic or placebo within 24 hours of diarrhea onset was 25.21 hours. Furthermore, the difference in duration of diarrhea was 13.84 hours (p<0.05) for participants who were administered with probiotics within 48 hours. There were no significant differences in the stool frequencies between the two arms. After three months, 15% in the probiotic group and 18.5% in the placebo group reported episodes of diarrhea. The mean duration of diarrhea was considerably lower in the probiotic group, 31.02 hours versus 48 hours in placebo (p=0.017). CONCLUSION S. boulardii CNCM-I 3799 and B. subtilis CU-1 combination was effective in reducing the duration of diarrhea when administered within 48 hours of diarrhea onset. Similarly, it reduced recurrence of diarrhea and its intensity in the subsequent three months.
Collapse
Affiliation(s)
- Apurba Ghosh
- Department of Pediatrics, Institute of Child Health, Kolkata, India
| | | | - Piyali Bhattacharya
- Department of Pediatrics, Sanjay Gandhi Post Graduate Institute of Medical Science, Lucknow, India
| | - Nimain Mohanty
- Department of Pediatrics, MGM Medical College, Navi Mumbai, India
| | - Nirmala Dheivamani
- Department of Pediatric Gastroenterology, Institute of Child Health, Chennai, India
| | - Sushant Mane
- Department of Pediatrics, Grant Govt. Medical College & Sir J.J. Group of Hospitals, Mumbai, India
| | - Bhaswati Acharyya
- Department of Pediatric Gastroenterology, Institute of Child Health, Kolkata, India
| | - Vijay Kamale
- Department of Pediatrics, MGM Medical College, Navi Mumbai, India
| | - Sumon Poddar
- Department of Microbiology, Institute of Child Health, Kolkata, India
| | - Akash Khobragade
- Department of Medical & Clinical Pharmacology, Grant Govt. Medical College & Sir J.J. Group of Hospitals, Mumbai, India
| | - Winston Thomas
- Department of Pediatrics, Institute of Child Health, Chennai, India
| | - Sumant Prabhudesai
- Department of Pediatric Intensive Care Unit, Kanchi Kamakoti Childs Trust Hospital, Chennai, India
| | - Ankita Choudhary
- Department of Pediatrics, MGM Medical College, Navi Mumbai, India
| | - Monjori Mitra
- Department of Pediatrics, Institute of Child Health, Kolkata, India
| |
Collapse
|
27
|
Xiong L, Li Y, Li J, Yang J, Shang L, He X, Liu L, Luo Y, Xie X. Intestinal microbiota profiles in infants with acute gastroenteritis caused by rotavirus and norovirus infection: a prospective cohort study. Int J Infect Dis 2021; 111:76-84. [PMID: 34411719 DOI: 10.1016/j.ijid.2021.08.024] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 07/18/2021] [Accepted: 08/12/2021] [Indexed: 11/19/2022] Open
Abstract
OBJECTIVE To compare the intestinal microbiota profiles in infants following rotavirus (RV) and human norovirus (HNoV) infection. METHODS Faecal specimens from 18 infants {mean age 11.8 months [standard deviation (SD) 3.0] months} with acute gastroenteritis caused by RV (G9P8) and 24 infants [mean age 8.8 (SD 6.4) months] with acute gastroenteritis caused by HNoV (GII) infection were collected prospectively. The faecal microbiome was assessed by 16S rRNA amplicon pyrosequencing. Alpha diversity, beta diversity, deferentially abundant taxa and microbial functions were assessed by bioinformatic analysis. RESULTS The Chao1 index for the HNoV group was significantly higher compared with the control group (P=0.0003), and was lower for the RV group compared with the HNoV group (P=0.0078). No significant difference in beta diversity was observed between the RV and HNoV groups. The RV group showed greater abundance of Actinobacteria at phylum level and Bifidobacterium spp., Streptococcus spp., Enterococcus spp. and Lactobacillus spp. at genus level. The HNoV group showed richness in Fusobacteria and Cyanobacteria at phylum level, and Enterococcus spp. and Streptococcus spp. at genus level. Bacillus was the characteristic genus in infected infants. In comparison with the control group, the viral group (P≤0.01), the RV group (P=0.002) and the HNoV group (P≤0.01) showed significant differences in potentially pathogenic bacteria. CONCLUSIONS Changes in microbiotic structure were observed in infants following RV and HNoV infection. The Chao 1 index of alpha diversity increased significantly in the HNoV group. Bacillus was the characteristic genus in infected infants. An increase in pathogenic bacteria, particularly Streptococcus spp. and Enterococcus spp., was detected in infected infants.
Collapse
Affiliation(s)
- Lijing Xiong
- Department of Paediatric Gastroenterology, Hepatology and Nutrition, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, People's Republic of China
| | - Yang Li
- Department of Paediatric Gastroenterology, Hepatology and Nutrition, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, People's Republic of China
| | - Jing Li
- Department of Paediatric Gastroenterology, Hepatology and Nutrition, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, People's Republic of China
| | - Jing Yang
- Department of Paediatric Gastroenterology, Hepatology and Nutrition, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, People's Republic of China
| | - Lihong Shang
- Department of Paediatric Gastroenterology, Hepatology and Nutrition, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, People's Republic of China
| | - Xiaoqing He
- Department of Paediatric Gastroenterology, Hepatology and Nutrition, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, People's Republic of China
| | - Lirong Liu
- Department of Paediatric Gastroenterology, Hepatology and Nutrition, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, People's Republic of China
| | - Yurong Luo
- Department of Paediatric Gastroenterology, Hepatology and Nutrition, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, People's Republic of China
| | - Xiaoli Xie
- Department of Paediatric Gastroenterology, Hepatology and Nutrition, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, People's Republic of China.
| |
Collapse
|
28
|
Merenstein D, Fraser CM, Roberts RF, Liu T, Grant-Beurmann S, Tan TP, Smith KH, Cronin T, Martin OA, Sanders ME, Lucan SC, Kane MA. Bifidobacterium animalis subsp. lactis BB-12 Protects against Antibiotic-Induced Functional and Compositional Changes in Human Fecal Microbiome. Nutrients 2021; 13:nu13082814. [PMID: 34444974 PMCID: PMC8398419 DOI: 10.3390/nu13082814] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 08/11/2021] [Indexed: 01/04/2023] Open
Abstract
The administration of broad-spectrum antibiotics is often associated with antibiotic-associated diarrhea (AAD), and impacts gastrointestinal tract homeostasis, as evidenced by the following: (a) an overall reduction in both the numbers and diversity of the gut microbiota, and (b) decreased short-chain fatty acid (SCFA) production. Evidence in humans that probiotics may enhance the recovery of microbiota populations after antibiotic treatment is equivocal, and few studies have addressed if probiotics improve the recovery of microbial metabolic function. Our aim was to determine if Bifidobacterium animalis subsp. lactis BB-12 (BB-12)-containing yogurt could protect against antibiotic-induced fecal SCFA and microbiota composition disruptions. We conducted a randomized, allocation-concealed, controlled trial of amoxicillin/clavulanate administration (days 1-7), in conjunction with either BB-12-containing or control yogurt (days 1-14). We measured the fecal levels of SCFAs and bacterial composition at baseline and days 7, 14, 21, and 30. Forty-two participants were randomly assigned to the BB-12 group, and 20 participants to the control group. Antibiotic treatment suppressed the fecal acetate levels in both the control and probiotic groups. Following the cessation of antibiotics, the fecal acetate levels in the probiotic group increased over the remainder of the study and returned to the baseline levels on day 30 (-1.6% baseline), whereas, in the control group, the acetate levels remained suppressed. Further, antibiotic treatment reduced the Shannon diversity of the gut microbiota, for all the study participants at day 7. The magnitude of this change was larger and more sustained in the control group compared to the probiotic group, which is consistent with the hypothesis that BB-12 enhanced microbiota recovery. There were no significant baseline clinical differences between the two groups. Concurrent administration of amoxicillin/clavulanate and BB-12 yogurt, to healthy subjects, was associated with a significantly smaller decrease in the fecal SCFA levels and a more stable taxonomic profile of the microbiota over time than the control group.
Collapse
Affiliation(s)
- Daniel Merenstein
- Department of Family Medicine, Georgetown University Medical Center, Washington, DC 20057, USA; (T.P.T.); (K.H.S.); (T.C.)
- Department of Human Science, School of Nursing and Health Studies, Georgetown University Medical Center, Washington, DC 20057, USA
- Correspondence: (D.M.); (C.M.F.); (M.A.K.); Tel.: +1-202-687-2745 (D.M.); +1-410-706-3879 (C.M.F.); +1-410-706-5097 (M.A.K.)
| | - Claire M. Fraser
- Institute for Genomic Sciences, Departments of Medicine and Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (S.G.-B.); (O.A.M.)
- Correspondence: (D.M.); (C.M.F.); (M.A.K.); Tel.: +1-202-687-2745 (D.M.); +1-410-706-3879 (C.M.F.); +1-410-706-5097 (M.A.K.)
| | - Robert F. Roberts
- Department of Food Science, The Pennsylvania State University, University Park, PA 16802, USA;
| | - Tian Liu
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, MD 21201, USA;
| | - Silvia Grant-Beurmann
- Institute for Genomic Sciences, Departments of Medicine and Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (S.G.-B.); (O.A.M.)
| | - Tina P. Tan
- Department of Family Medicine, Georgetown University Medical Center, Washington, DC 20057, USA; (T.P.T.); (K.H.S.); (T.C.)
| | - Keisha Herbin Smith
- Department of Family Medicine, Georgetown University Medical Center, Washington, DC 20057, USA; (T.P.T.); (K.H.S.); (T.C.)
| | - Tom Cronin
- Department of Family Medicine, Georgetown University Medical Center, Washington, DC 20057, USA; (T.P.T.); (K.H.S.); (T.C.)
| | - Olivia A. Martin
- Institute for Genomic Sciences, Departments of Medicine and Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (S.G.-B.); (O.A.M.)
- Department of Surgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | | | - Sean C. Lucan
- Department of Family and Social Medicine, Albert Einstein College of Medicine, Montefiore Health System, Bronx, NY 10461, USA;
| | - Maureen A. Kane
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, MD 21201, USA;
- Correspondence: (D.M.); (C.M.F.); (M.A.K.); Tel.: +1-202-687-2745 (D.M.); +1-410-706-3879 (C.M.F.); +1-410-706-5097 (M.A.K.)
| |
Collapse
|
29
|
Depoorter L, Vandenplas Y. Probiotics in Pediatrics. A Review and Practical Guide. Nutrients 2021; 13:2176. [PMID: 34202742 PMCID: PMC8308463 DOI: 10.3390/nu13072176] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 06/17/2021] [Accepted: 06/21/2021] [Indexed: 02/07/2023] Open
Abstract
The potential benefit of the administration of probiotics in children has been studied in many settings globally. Probiotics products contain viable micro-organisms that confer a health benefit on the host. Beneficial effects of selected probiotic strains for the management or prevention of selected pediatric conditions have been demonstrated. The purpose of this paper is to provide an overview of current available evidence on the efficacy of specific probiotics in selected conditions to guide pediatricians in decision-making on the therapeutic or prophylactic use of probiotic strains in children. Evidence to support the use of certain probiotics in selected pediatric conditions is often available. In addition, the administration of probiotics is associated with a low risk of adverse events and is generally well tolerated. The best documented efficacy of certain probiotics is for treatment of infectious gastroenteritis, and prevention of antibiotic-associated, Clostridioides difficile-associated and nosocomial diarrhea. Unfortunately, due to study heterogeneity and in some cases high risk of bias in published studies, a broad consensus is lacking for specific probiotic strains, doses and treatment regimens for some pediatric indications. The current available evidence thus limits the systematic administration of probiotics. The most recent meta-analyses and reviews highlight the need for more well-designed, properly powered, strain-specific and dedicated-dose response studies.
Collapse
Affiliation(s)
| | - Yvan Vandenplas
- Vrije Universiteit Brussel (VUB), UZ Brussel, KidZ Health Castle, 1090 Brussels, Belgium;
| |
Collapse
|
30
|
Ivashkin VT, Maev IV, Abdulganieva DI, Alekseenko SA, Gorelov AV, Zakharova IN, Zolnikova OY, Ivashkina NY, Korochanskaya NV, Mammayev SN, Poluektova EA, Trukhmanov AS, Usenko DV, Uspensky YP, Tsukanov VV, Shifrin OS, Berezhnaya IV, Ivashkin KV, Lapina TL, Maslennikov RV, Nikolaeva SV, Sugyan NG, Ulyanin AI. Practical Recommendations of Scientific Society for the Study of Human Microbiome and the Russian Gastroenterological Association on Use of Probiotics, Prebiotics, Synbiotics and Functional Foods in Treatment and Prevention of Gastroenterological Diseases in Children and Adults. RUSSIAN JOURNAL OF GASTROENTEROLOGY, HEPATOLOGY, COLOPROCTOLOGY 2021. [DOI: 10.22416/1382-4376-2021-31-2-65-91] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Aim. The practical guidelines are intended for primary care physicians, general practitioners, paediatricians, gastroenterologists and general internists to advance the treatment and prevention of gastroenterological diseases in adults and children in therapies with probiotics, prebiotics, synbiotics and their enriched functional foods.Key points. Probiotics are live microorganisms that sustain health of the host when supplied in adequate amounts. Prebiotics include human-indigestible but accessible to gut microbiota substances expediting specific changes in the composition and/or activity of gastrointestinal microbiota that favour the host health. The mechanism of probiotic action comprises the quorum resistance maintenance, nutrient substrate metabolism and end metabolite recycling, macroorganism-sustaining substrate production, as well as the mediation of local and adaptive immune responses.The Russian Federation regulates market differently for biologically active food additives (BAFA), medicinal products (drugs) and functional food products (FFP). We overview the probiotic strains regulated in Russia as BAFAs, drugs and FFPs and provide recommendations on the use of these strains in treatment and prevention of gastroenterological diseases in children and adults.Conclusion. The clinical efficacy of probiotics, prebiotics, synbiotics and fortified functional foods depends on the prebiotic and strain properties and is verified in appropriate comparative clinical trials. Not all probiotics registered in Russia as BAFAs, drugs and FFPs have a strain identity, which provides no warranty of the clinical effect expected. The FFP legislation demands improved regulation mechanisms and control for therapeutic efficacy.
Collapse
Affiliation(s)
- V. T. Ivashkin
- Sechenov First Moscow State University (Sechenov University)
| | - I. V. Maev
- Yevdokimov Moscow State University of Medicine and Dentistry
| | | | | | - A. V. Gorelov
- Sechenov First Moscow State University (Sechenov University); Central Research Institute of Epidemiology
| | - I. N. Zakharova
- Russian Medical Academy of Continuous Professional Education
| | | | | | | | | | | | | | - D. V. Usenko
- Russian Medical Academy of Continuous Professional Education
| | | | - V. V. Tsukanov
- Research Institute for Medical Problems in the North — Division of Krasnoyarsk Scientific Centre of the Siberian Branch of the RAS
| | - O. S. Shifrin
- Sechenov First Moscow State University (Sechenov University)
| | | | - K. V. Ivashkin
- Sechenov First Moscow State University (Sechenov University)
| | - T. L. Lapina
- Sechenov First Moscow State University (Sechenov University)
| | | | | | - N. G. Sugyan
- Russian Medical Academy of Continuous Professional Education
| | - A. I. Ulyanin
- Sechenov First Moscow State University (Sechenov University)
| |
Collapse
|
31
|
Agudelo-Chaparro J, Ciro-Velásquez HJ, Sepúlveda-Valencia JU, Pérez-Monterroza EJ. Microencapsulation of Lactobacillus rhamnosus ATCC 7469 by spray drying using maltodextrin, whey protein concentrate and trehalose. FOOD SCI TECHNOL INT 2021; 28:476-488. [PMID: 34058894 DOI: 10.1177/10820132211020621] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
This study aimed to microencapsulate Lactobacillus rhamnosus (L. rhamnosus) ATCC 7469 with whey protein concentrate (WPC), maltodextrin and trehalose by spray drying and to assess the impact of microencapsulation on cell viability and the properties of the dried powders. Spray-drying conditions, including inlet air temperature, air flow rate and feed pump, were fixed as independent variables, while probiotic survival, moisture content, water activity and effective yield were established as dependent variables. The survival of encapsulated L. rhamnosus by spray drying was optimized with response surface methodology, and the stability of the powder was assessed. The optimum spray-drying conditions were an inlet air temperature, air flow rate and feed pump rate of 169 °C, 33 m3·h-1 and 16 mL·min-1, respectively, survival of 70%, air aspiration of 84% and outlet air temperature of 52 °C, achieving an overall desirability of 0.96. The physicochemical and structural characteristics of the produced powder were acceptable for application with regard to residual water content, hygroscopicity, water activity, and particle size. The results indicated that a protein-trehalose-maltodextrin mixture is a good wall material to encapsulate L. rhamnosus, showing important thermal protection during the drying process and increasing survival. However, a decrease in this capacity is observed at an air outlet temperature of approximately 101 °C. The possible effects of the wall materials and the drying conditions on survival are also discussed.
Collapse
Affiliation(s)
- Jacqueline Agudelo-Chaparro
- Department of Agricultural and Food Engineering, Universidad Nacional de Colombia, Sede Medellín, Medellín, Colombia
| | - Héctor J Ciro-Velásquez
- Department of Agricultural and Food Engineering, Universidad Nacional de Colombia, Sede Medellín, Medellín, Colombia
| | - José U Sepúlveda-Valencia
- Department of Agricultural and Food Engineering, Universidad Nacional de Colombia, Sede Medellín, Medellín, Colombia
| | | |
Collapse
|
32
|
Wzorek-Łyczko K, Piwowarczyk A, Kuchar E. Protocol of the study: the effectiveness of pleuran in the treatment of acute gastroenteritis in children-a randomised, placebo-controlled, double-blind trial (EPTAGE). BMJ Open 2021; 11:e042370. [PMID: 33707267 PMCID: PMC7957130 DOI: 10.1136/bmjopen-2020-042370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Abstract
INTRODUCTION Acute gastroenteritis is one of the most common causes of children's morbidity and mortality globally. Oral or intravenous rehydration was proven effective in reducing the mortality rates in acute gastroenteritis, although it does not affect the course of the disease. Attempts to identify new therapeutic methods effective in reducing the symptoms of diarrhoea are of interest. Pleuran's potential immunomodulatory effect in acute gastrointestinal infection relies on the stimulation of innate immunity. The effectiveness of pleuran (β-(1,3/1,6)-d-glucan) administration to treat acute infectious diarrhoea remains unknown. This study evaluates the efficacy of pleuran in reducing diarrhoea duration and the severity of acute gastroenteritis symptoms in children. METHODS AND ANALYSIS Our study is a randomised, double-blind, placebo-controlled superiority trial with two parallel groups and a 1:1 allocation ratio. A total of 120 children aged 2-10 years hospitalised or requiring a visit to the emergency department because of acute gastroenteritis will be randomly assigned to receive either pleuran oral suspension in the experimental group or matching placebo in the control group. The primary outcome measure will be the duration of diarrhoea. We will analyse the results in both intention-to-treat and per-protocol approaches. ETHICS AND DISSEMINATION The Bioethics Committee of The Medical University of Warsaw approved the study protocol (approval number: KB/45/2018). Written informed consent of the patients' caregivers participating in the study will be obligatory. The results of this study will be published in a medical journal, regardless of whether they confirm or deny the research hypothesis. TRIAL REGISTRATION NUMBER NCT03988257; Pre-results.
Collapse
Affiliation(s)
- Katarzyna Wzorek-Łyczko
- The Department of Pediatrics with Clinical Assessment Unit, Medical University of Warsaw, Warsaw, Poland
| | - Anna Piwowarczyk
- The Department of Pediatrics with Clinical Assessment Unit, Medical University of Warsaw, Warsaw, Poland
| | - Ernest Kuchar
- The Department of Pediatrics with Clinical Assessment Unit, Medical University of Warsaw, Warsaw, Poland
| |
Collapse
|
33
|
Tomczak H, Wrońska M, Pecyna P, Hampelska K. The issue of the correct use of probiotics
in the absence of recommendations. POSTEP HIG MED DOSW 2021. [DOI: 10.5604/01.3001.0014.7701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Antibiotics are important for saving both human health and life. Antibiotics destroy all bacteria
within their spectrum, because they do not distinguish between good and bad bacteria.
Even if an antibiotic therapy lasts only a few days, it may cause diarrhoea and mycosis.
Antibiotics destroy most bacterial species in the intestines. These changes may affect one’s
whole life. Today it is a challenge for medicine to be able to manipulate the microbiome so
as to restore normal relations between microorganisms. At present, when antibiotics are
abused, probiotics are very often applied. However, as there are no recommendations,
a lot of mistakes can be made when using them. Both drugs and dietary supplements can be
classified as probiotics. Medicinal probiotics are subject to very strict registration requirements
and their use is associated with a specific disease or ailment. Probiotic microorganisms
must be classified according to their genus, species and strain. These preparations
may contain one or more probiotic strains depending on its application. At present there
are no established schemes or rules concerning the dosage of probiotic preparations. This
issue arouses numerous controversies. It is assumed that the probiotic should be applied
at a dose which proved to have a beneficial effect in tests conducted on humans. Patients
usually make decisions on the choice and dosage of preparations themselves. Individualised
probiotic therapy is the key to success. There is no universal preparation – a specific probiotic
should be used in a particular clinical case.
Collapse
Affiliation(s)
- Hanna Tomczak
- Central Microbiology Laboratory, H. Święcicki Clinical Hospital at the Medical University in Poznań, Poznań, Poland
| | - Marta Wrońska
- Central Microbiology Laboratory, H. Święcicki Clinical Hospital at the Medical University in Poznań, Poznań, Poland
| | - Paulina Pecyna
- Department of Genetics and Pharmaceutical Microbiology, Poznań University of Medical Sciences, Poznań, Poland
| | - Katarzyna Hampelska
- Central Microbiology Laboratory, H. Święcicki Clinical Hospital at the Medical University in Poznań, Poznań, Poland
| |
Collapse
|
34
|
Barbuti RC, Schiavon LL, Oliveira CP, Alvares-DA-Silva MR, Sassaki LY, Passos MDCF, Farias AQ, Barros LL, Barreto BP, Albuquerque GBDMLD, Alves AM, Navarro-Rodriguez T, Bittencourt PL. GUT MICROBIOTA, PREBIOTICS, PROBIOTICS, AND SYNBIOTICS IN GASTROINTESTINAL AND LIVER DISEASES: PROCEEDINGS OF A JOINT MEETING OF THE BRAZILIAN SOCIETY OF HEPATOLOGY (SBH), BRAZILIAN NUCLEUS FOR THE STUDY OF HELICOBACTER PYLORI AND MICROBIOTA (NBEHPM), AND BRAZILIAN FEDERATION OF GASTROENTEROLOGY (FBG). ARQUIVOS DE GASTROENTEROLOGIA 2021; 57:381-398. [PMID: 33331485 DOI: 10.1590/s0004-2803.202000000-72] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Accepted: 04/06/2020] [Indexed: 02/07/2023]
Abstract
Over the last years, there is growing evidence that microorganisms are involved in the maintenance of our health and are related to various diseases, both intestinal and extraintestinal. Changes in the gut microbiota appears to be a key element in the pathogenesis of hepatic and gastrointestinal disorders, including non-alcoholic fatty liver disease, alcoholic liver disease, liver cirrhosis, inflammatory bowel disease, irritable bowel syndrome, and Clostridium difficile - associated diarrhea. In 2019, the Brazilian Society of Hepatology (SBH) in cooperation with the Brazilian Nucleus for the Study of Helicobacter Pylori and Microbiota (NBEHPM), and Brazilian Federation of Gastroenterology (FBG) sponsored a joint meeting on gut microbiota and the use of prebiotics, probiotics, and synbiotics in gastrointestinal and liver diseases. This paper summarizes the proceedings of the aforementioned meeting. It is intended to provide practical information about this topic, addressing the latest discoveries and indicating areas for future studies.
Collapse
Affiliation(s)
- Ricardo Correa Barbuti
- Universidade de São Paulo, Faculdade de Medicina, Departamento de Gastroenterologia, São Paulo, SP, Brasil
| | - Leonardo Lucca Schiavon
- Universidade Federal de Santa Catarina, Faculdade de Medicina, Departamento de Clínica Médica, Florianópolis, SC, Brasil
| | - Cláudia P Oliveira
- Universidade de São Paulo, Faculdade de Medicina, Departamento de Gastroenterologia, São Paulo, SP, Brasil
| | - Mário Reis Alvares-DA-Silva
- Universidade Federal do Rio Grande do Sul, Faculdade de Medicina, Departamento de Medicina Interna, Porto Alegre, RS, Brasil
| | | | | | - Alberto Queiroz Farias
- Universidade de São Paulo, Faculdade de Medicina, Departamento de Gastroenterologia, São Paulo, SP, Brasil
| | - Luisa Leite Barros
- Universidade de São Paulo, Faculdade de Medicina, Departamento de Gastroenterologia, São Paulo, SP, Brasil
| | - Bruno Paes Barreto
- Universidade do Estado do Pará, Centro de Ciências Biológicas e da Saúde, Belém, PA, Brasil.,Centro Universitário do Estado do Pará (CESUPA), Belém, PA, Brasil
| | | | - Amanda Mandarino Alves
- Universidade de São Paulo, Faculdade de Medicina, Departamento de Gastroenterologia, São Paulo, SP, Brasil
| | - Tomás Navarro-Rodriguez
- Universidade de São Paulo, Faculdade de Medicina, Departamento de Gastroenterologia, São Paulo, SP, Brasil
| | | |
Collapse
|
35
|
Lee YY, Leow AHR, Chai PF, Raja Ali RA, Lee WS, Goh KL. Use of probiotics in clinical practice with special reference to diarrheal diseases: A position statement of the Malaysian Society of Gastroenterology and Hepatology. JGH OPEN 2021; 5:11-19. [PMID: 33490608 PMCID: PMC7812487 DOI: 10.1002/jgh3.12469] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 11/18/2020] [Accepted: 11/24/2020] [Indexed: 12/17/2022]
Abstract
Probiotics comprise a large group of microorganisms, which have different properties and thus confer different benefits. The use of probiotics has shown promising results in the management of diarrheal diseases. While the availability of probiotic products has flourished in the marketplace, there is limited guidance on the selection of probiotics for clinical use. This position paper is aimed at informing clinicians about the proper selection criteria of probiotics based on current evidence on strain-specific efficacy and safety for the management of diarrheal diseases. Members of the working group discussed issues on probiotic use in clinical practice, which were then drafted into statements. Literature to support or refute the statements were gathered through a search of medical literature from 2011 to 2020. Recommendations were formulated based on the drafted statements and evidence gathered, revised as necessary, and finalized upon agreement of all members. Twelve statements and recommendations were developed covering the areas of quality control in the manufacturing of probiotics, criteria for selection of probiotics, and established evidence for use of probiotics in diarrheal diseases in adults and children. Recommendations for the use of specific probiotic strains in clinical practice were categorized as proven and probable efficacy based on strength of evidence. Robust evidence is available to support the use of probiotics for diarrheal diseases in clinical practice. Based on the results obtained, we strongly advocate the careful evaluation of products, including manufacturing practices, strain-specific evidence, and contraindications for at-risk populations when choosing probiotics for use in clinical practice.
Collapse
Affiliation(s)
- Yeong-Yeh Lee
- Department of Medicine, School of Medical Sciences Universiti Sains Malaysia Kota Bharu Malaysia.,Gut Research Group, Faculty of Medicine Universiti Kebangsaan Malaysia Kuala Lumpur Malaysia
| | - Alex H-R Leow
- Gastroenterology Unit, Department of Medicine, Faculty of Medicine Universiti Kebangsaan Malaysia Jalan Universiti Kuala Lumpur Malaysia.,University of Malaya Medical Centre Jalan Universiti Kuala Lumpur Malaysia
| | - Pei-Fan Chai
- University of Malaya Medical Centre Jalan Universiti Kuala Lumpur Malaysia
| | - Raja Affendi Raja Ali
- Gut Research Group, Faculty of Medicine Universiti Kebangsaan Malaysia Kuala Lumpur Malaysia.,Endoscopy Centre Pantai Hospital Kuala Lumpur Malaysia
| | - Way-Seah Lee
- Department of Paediatrics, Faculty of Medicine University of Malaya Kuala Lumpur Malaysia
| | - Khean-Lee Goh
- Department of Medicine, Faculty of Medicine University of Malaya Kuala Lumpur Malaysia
| |
Collapse
|
36
|
Bioactive Compounds Produced by the Accompanying Microflora in Bulgarian Yoghurt. Processes (Basel) 2021. [DOI: 10.3390/pr9010114] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Bulgarian yoghurt is associated with health benefits and longevity of consumers. The specific microflora producing bioactive metabolites is responsible for this effect. The present study examines the biodiversity in four homemade yoghurts from regions containing endemic microflora. Metagenome sequencing indicated Lactobacillus delbrueckii subsp. bulgaricus and Streptococcus thermophilus were predominant in all samples. In addition, yoghurts contained accompanying lactic acid bacteria (LAB) including Lacticaseibacillus paracasei, Lb. helveticus, Limosilactobacillus fermentum, Lb. rhamnosus, Lactococcus lactis, Pediococcus acidilactici, Leuconostoc mesenteroides, and Leuc. pseudomesenteroides. A negligible amount of pollutant strains was found. Twenty-four LAB strains were isolated from the yoghurts and identified. Lb. delbrueckii subsp. bulgaricus strains were genotyped by randomly amplified polymorphic DNA–PCR (RAPD), multi-locus sequence typing (MLST), and pulse field gel electrophoresis (PFGE), which demonstrated their uniqueness and non-commercial origin. To estimate the bioactive metabolites produced by the accompanying microflora, yoghurts fermented by single LAB strains were analyzed using liquid chromatography and mass spectrometry (LC-MS). The fermented samples contained large amounts of free essential amino acids (arginine, citrulline, tryptophan, lysine, and histidine), the neuroprotector indole-3-propionic acid (IPA), and significant quantities of the cyclic antimicrobial peptides cyclo(phenylalanyl-prolyl) and cyclo(leucyloprolyl). The disclosure of these special qualities draws attention to the accompanying microflora as a source of potential probiotic strains that can fortify the yoghurts’ content with bioactive compounds.
Collapse
|
37
|
Schnadower D, Sapien RE, Casper TC, Vance C, Tarr PI, O'Connell KJ, Levine AC, Roskind CG, Rogers AJ, Bhatt SR, Mahajan P, Powell EC, Olsen CS, Gorelick MH, Dean JM, Freedman SB. Association between Age, Weight, and Dose and Clinical Response to Probiotics in Children with Acute Gastroenteritis. J Nutr 2021; 151:65-72. [PMID: 33274370 PMCID: PMC7779240 DOI: 10.1093/jn/nxaa313] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 06/25/2020] [Accepted: 09/17/2020] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Gastroenteritis is a common and impactful disease in childhood. Probiotics are often used to treat acute gastroenteritis (AGE); however, in a large multicenter randomized controlled trial (RCT) in 971 children, Lactobacillus rhamnosus GG (LGG) was no better than placebo in improving patient outcomes. OBJECTIVES We sought to determine whether the effect of LGG is associated with age, weight z score and weight percentile adjusted for age and sex, or dose per kilogram administered. METHODS This was a preplanned secondary analysis of a multicenter double-blind RCT of LGG 1 × 1010 CFU twice daily for 5 d or placebo in children 3-48 mo of age with AGE. Our primary outcome was moderate to severe gastroenteritis. Secondary outcomes included diarrhea and vomiting frequency and duration, chronic diarrhea, and side effects. We used multivariable linear and nonlinear models testing for interaction effects to assess outcomes by age, weight z score and weight percentile adjusted for age and sex, and dose per kilogram of LGG received. RESULTS A total of 813 children (84%) were included in the analysis; 413 received placebo and 400 LGG. Baseline characteristics were similar between treatment groups. There were no differential interaction effects across ranges of age (P-interaction = 0.32), adjusted weight z score (P-interaction = 0.43), adjusted weight percentile (P-interaction = 0.45), or dose per kilogram of LGG received (P-interaction = 0.28) for the primary outcome. Whereas we found a statistical association favoring placebo at the extremes of adjusted weight z scores for the number of vomiting episodes (P-interaction = 0.02) and vomiting duration (P-interaction = 0.0475), there were no statistically significant differences in other secondary outcome measures (all P-interactions > 0.05). CONCLUSIONS LGG does not improve outcomes in children with AGE regardless of the age, adjusted weight z score, and adjusted weight percentile of participants, or the probiotic dose per kilogram received. These results further strengthen the conclusions of low risk of bias clinical trials which demonstrate that LGG provides no clinical benefit in children with AGE.This trial was registered at clinicaltrials.gov as NCT01773967.
Collapse
Affiliation(s)
- David Schnadower
- Division of Emergency Medicine, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Robert E Sapien
- Department of Emergency Medicine, University of New Mexico, Albuquerque, NM, USA
| | - T Charles Casper
- Department of Pediatrics, University of Utah, Salt Lake City, UT, USA
| | - Cheryl Vance
- Departments of Emergency Medicine and Pediatrics, University of California, Davis, School of Medicine, Sacramento, CA, USA
| | - Phillip I Tarr
- Division of Gastroenterology, Hepatology, & Nutrition, Department of Pediatrics, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Karen J O'Connell
- Division of Emergency Medicine, Children's National Health System, Department of Pediatrics and Emergency Medicine, The George Washington School of Medicine and Health Sciences, Washington, DC, USA
| | - Adam C Levine
- Department of Emergency Medicine, Rhode Island Hospital/Hasbro Children's Hospital and Brown University, Providence, RI, USA
| | - Cindy G Roskind
- Division of Emergency Medicine, Department of Pediatrics, Columbia University College of Physicians & Surgeons, New York, NY, USA
| | - Alexander J Rogers
- Departments of Emergency Medicine and Pediatrics, University of Michigan, Ann Arbor, MI, USA
| | - Seema R Bhatt
- Division of Emergency Medicine, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Prashant Mahajan
- Departments of Emergency Medicine and Pediatrics, University of Michigan, Ann Arbor, MI, USA
| | - Elizabeth C Powell
- Division of Emergency Medicine, Department of Pediatrics, Ann & Robert H Lurie Children's Hospital, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Cody S Olsen
- Department of Pediatrics, University of Utah, Salt Lake City, UT, USA
| | - Marc H Gorelick
- Central Administration, Children's Minnesota, Minneapolis, MN, USA
| | - J Michael Dean
- Department of Pediatrics, University of Utah, Salt Lake City, UT, USA
| | - Stephen B Freedman
- Sections of Pediatric Emergency Medicine and Gastroenterology, Department of Pediatrics, Alberta Children's Hospital, Alberta Children's Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | | |
Collapse
|
38
|
María Remes Troche J, Coss Adame E, Ángel Valdovinos Díaz M, Gómez Escudero O, Eugenia Icaza Chávez M, Antonio Chávez-Barrera J, Zárate Mondragón F, Antonio Ruíz Velarde Velasco J, Rafael Aceves Tavares G, Antonio Lira Pedrín M, Cerda Contreras E, Carmona Sánchez RI, Guerra López H, Solana Ortiz R. Lactobacillus acidophilus LB: a useful pharmabiotic for the treatment of digestive disorders. Therap Adv Gastroenterol 2020; 13:1756284820971201. [PMID: 33281937 PMCID: PMC7692339 DOI: 10.1177/1756284820971201] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 10/07/2020] [Indexed: 02/04/2023] Open
Abstract
Dysbiosis, a loss of balance between resident bacterial communities and their host, is associated with multiple diseases, including inflammatory bowel diseases (nonspecific chronic ulcerative colitis and Crohn's disease), and digestive functional disorders. Probiotics, prebiotics, synbiotic organisms and, more recently, pharmabiotics, have been shown to modulate the human microbiota. In this review, we provide an overview of the key concepts relating to probiotics, prebiotics, synbiotic organisms, and pharmabiotics, with a focus on available clinical evidence regarding the specific use of a unique pharmabiotic, the strain Lactobacillus acidophilus LB (Lactobacillus boucardii), for the management of gastrointestinal disorders. Since it does not contain living organisms, the administration of L. acidophilus LB is effective and safe as an adjuvant in the treatment of acute diarrhea, chronic diarrhea, and antibiotic-associated diarrhea, even in the presence of immunosuppression.
Collapse
Affiliation(s)
- José María Remes Troche
- Instituto e Investigaciones Médico Biológicas de la Universidad Veracruzana, Veracruz, Mexico
| | - Enrique Coss Adame
- Gastroenterology Department, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Miguel Ángel Valdovinos Díaz
- National Institute of Medical Sciences and Nutrition Salvador Zubirán, Vasco de Quiroga 15, Belisario Domínguez Secc 16, Mexico City 14080, Mexico
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Cielecka-Piontek J, Dziedziński M, Szczepaniak O, Kobus-Cisowska J, Telichowska A, Szymanowska D. Survival of commercial probiotic strains and their effect on dark chocolate synbiotic snack with raspberry content during the storage and after simulated digestion. ELECTRON J BIOTECHN 2020. [DOI: 10.1016/j.ejbt.2020.09.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
|
40
|
De Castro JA, Kesavelu D, Lahiri KR, Chaijitraruch N, Chongsrisawat V, Jog PP, Liaw YH, Nguyen GK, Nguyen TVH, Pai UA, Phan HND, Quak SH, Tanpowpong P, Guno MJ. Recommendations for the adjuvant use of the poly-antibiotic-resistant probiotic Bacillus clausii (O/C, SIN, N/R, T) in acute, chronic, and antibiotic-associated diarrhea in children: consensus from Asian experts. Trop Dis Travel Med Vaccines 2020; 6:21. [PMID: 33110611 PMCID: PMC7583175 DOI: 10.1186/s40794-020-00120-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 09/28/2020] [Indexed: 11/17/2022] Open
Abstract
This paper proposes recommendations for probiotics in pediatric gastrointestinal diseases in the Asia-Pacific region. Evidence-based recommendations and randomized controlled trials in the region are included. Cultural aspects, health management issues and economic factors were also considered. Final recommendations were approved by utilizing a modified Delphi process and applying the Likert scale in an electronic voting process. Bacillus clausii was recommended as an adjunct treatment with oral rehydration solution for acute viral diarrhea. B. clausii may also be considered for prevention of antibiotic-associated diarrhea, Clostridium difficile-induced diarrhea, and as adjunct treatment of Helicobacter pylori. There is insufficient evidence for recommendations in other conditions. Despite a diversity of epidemiological, socioeconomical and health system conditions, similar recommendations currently apply to most Asia-Pacific countries. Ideally, these need to be validated with local randomized-controlled trials.
Collapse
Affiliation(s)
- Jo-Anne De Castro
- De La Salle University Medical Center, Dasmariñas, Cavite Philippines
| | | | | | - Nataruks Chaijitraruch
- Faculty of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, Bangkok, Thailand
| | - Voranush Chongsrisawat
- Faculty of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, Bangkok, Thailand
| | | | - Yun Haw Liaw
- KPJ Sabah Specialist Hospital, Kota Kinabalu, Sabah Malaysia
| | | | | | | | - Huu Nguyet Diem Phan
- University of Medicine and Pharmacy of Ho Chi Minh City, Ho Chi Minh City, Vietnam
| | | | - Pornthep Tanpowpong
- Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Mary Jean Guno
- Institute of Pediatrics, Section of Pediatric Gastroenterology, Hepatology and Nutrition, The Medical City, Pasig City, Philippines
| |
Collapse
|
41
|
Stavropoulou E, Bezirtzoglou E. Probiotics in Medicine: A Long Debate. Front Immunol 2020; 11:2192. [PMID: 33072084 PMCID: PMC7544950 DOI: 10.3389/fimmu.2020.02192] [Citation(s) in RCA: 118] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 08/11/2020] [Indexed: 12/12/2022] Open
Abstract
During the last years probiotics gained the attention of clinicians for their use in the prevention and treatment of multiple diseases. Probiotics main mechanisms of action include enhanced mucosal barrier function, direct antagonism with pathogens, inhibition of bacterial adherence and invasion capacity in the intestinal epithelium, boosting of the immune system and regulation of the central nervous system. It is accepted that there is a mutual communication between the gut microbiota and the liver, the so-called “microbiota-gut-liver axis” as well as a reciprocal communication between the intestinal microbiota and the central nervous system through the “microbiota-gut-brain axis.” Moreover, recently the “gut-lung axis” in bacterial and viral infections is considerably discussed for bacterial and viral infections, as the intestinal microbiota amplifies the alveolar macrophage activity having a protective role in the host defense against pneumonia. The importance of the normal human intestinal microbiota is recognized in the preservation of health. Disease states such as, infections, autoimmune conditions, allergy and other may occur when the intestinal balance is disturbed. Probiotics seem to be a promising approach to prevent and even reduce the symptoms of such clinical states as an adjuvant therapy by preserving the balance of the normal intestinal microbiota and improving the immune system. The present review states globally all different disorders in which probiotics can be given. To date, Stronger data in favor of their clinical use are provided in the prevention of gastrointestinal disorders, antibiotic-associated diarrhea, allergy and respiratory infections. We hereby discuss the role of probiotics in the reduction of the respiratory infection symptoms and we focus on the possibility to use them as an adjuvant to the therapeutic approach of the pandemic COVID-19. Nevertheless, it is accepted by the scientific community that more clinical studies should be undertaken in large samples of diseased populations so that the assessment of their therapeutic potential provide us with strong evidence for their efficacy and safety in clinical use.
Collapse
Affiliation(s)
- Elisavet Stavropoulou
- CHUV (Centre Hospitalier Universitaire Vaudois), Lausanne, Switzerland.,Department of Infectious Diseases, Central Institute, Valais Hospital, Sion, Switzerland
| | - Eugenia Bezirtzoglou
- Laboratory of Hygiene and Environmental Protection, Department of Medicine, Democritus University of Thrace, Alexandroupolis, Greece
| |
Collapse
|
42
|
Posovszky C, Buderus S, Classen M, Lawrenz B, Keller KM, Koletzko S. Acute Infectious Gastroenteritis in Infancy and Childhood. DEUTSCHES ARZTEBLATT INTERNATIONAL 2020; 117:615-624. [PMID: 33263539 PMCID: PMC7805585 DOI: 10.3238/arztebl.2020.0615] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/26/2020] [Revised: 01/26/2020] [Accepted: 06/29/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND Despite the introduction of vaccination against rotavirus, and even though it can often be treated on an outpatient basis, acute infectious gastroenteritis is nevertheless the second most common non-traumatic cause of emergency hospitaliza - tion in children aged 1 to 5 years, accounting for approximately 9% of cases (39 410 cases in 2017). The most common path - ogens are viruses (47% rotavirus, 29% norovirus, and 14% adenovirus). METHODS This review is based on publications retrieved by a selective search in PubMed employing the terms "acute gastro - enteritis children" AND "dehydration" OR "rehydration" OR "prevention," and by manual searching (based, for example, on reference lists and expert knowledge), with subsequent evaluation including consideration of the relevant guidelines. RESULTS The degree of dehydration can be judged from weight loss and other clinical findings. In 17 randomized controlled trials conducted on a total of 1811 children with mild or moderate dehydration, oral rehydration with oral rehydration solution was just as effective as intravenous rehydration with respect to weight gain, duration of diarrhea, and fluid administration, and was associated with shorter hospital stays (weighted mean difference, -1.2 days; 95% confidence interval [-2.38; -0.02]). Oral rehydration therapy failed in 4% of patients [1; 7]. In children who are vomiting or who refuse oral rehydration solution, continuous nasogastric application is just as effective as intravenous rehydration and is the treatment of first choice. CONCLUSION In Germany, children with mild or moderate dehydration are often hospitalized for intravenous rehydration therapy, despite the good evidence supporting ambulatory oral rehydration. Obstacles to intersectoral care, the nursing shortage, and inadequate reimbursement must all be overcome in order to reduce unnecessary hospitalizations and thereby lessen the risk of nosocomial infection.
Collapse
Affiliation(s)
- Carsten Posovszky
- Department of Pediatric and Adolescent Medicine, University Medical Center Ulm
| | - Stephan Buderus
- Department of Pediatrics, GFO-Kliniken Bonn, St. Marienhospital Bonn
| | - Martin Classen
- Department of Pediatric and Adolescent Medicine, Klinikum Links der Weser and Klinikum Bremen-Mitte, Bremen
| | | | | | - Sibylle Koletzko
- Department of Pediatric and Adolescent Medicine, Dr. von Hauner Children’s Hospital, LMU Klinikum der Universität München
- Department of Pediatrics, Gastroenterology and Nutrition, School of Medicine Collegium Medicum University of Warmia and Mazury, Olsztyn, Poland
| |
Collapse
|
43
|
Dioso CM, Vital P, Arellano K, Park H, Todorov SD, Ji Y, Holzapfel W. Do Your Kids Get What You Paid for? Evaluation of Commercially Available Probiotic Products Intended for Children in the Republic of the Philippines and the Republic of Korea. Foods 2020; 9:E1229. [PMID: 32899215 PMCID: PMC7555838 DOI: 10.3390/foods9091229] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 08/28/2020] [Accepted: 08/31/2020] [Indexed: 12/18/2022] Open
Abstract
A wide range of probiotic products is available on the market and can be easily purchased over the counter and unlike pharmaceutical drugs, their commercial distribution is not strictly regulated. In this study, ten probiotic preparations commercially available for children's consumption in the Republic of the Philippines (PH) and the Republic of Korea (SK) have been investigated. The analyses included determination of viable counts and taxonomic identification of the bacterial species present in each formulation. The status of each product was assessed by comparing the results with information and claims provided on the label. In addition to their molecular identification, safety assessment of the isolated strains was conducted by testing for hemolysis, biogenic amine production and antibiotic resistance. One out of the ten products contained lower viable numbers of recovered microorganisms than claimed on the label. Enterococcus strains, although not mentioned on the label, were isolated from four products. Some of these isolates produced biogenic amines and were resistant to one or several antibiotics. Metagenomic analyses of two products revealed that one product did not contain most of the microorganisms declared in its specification. The study demonstrated that some commercial probiotic products for children did not match their label claims. Infants and young children belong to the most vulnerable members of society, and food supplements including probiotics destined for this consumer group require careful checking and strict regulation before commercial distribution.
Collapse
Affiliation(s)
- Clarizza May Dioso
- Institute of Biology, College of Science, University of the Philippines Diliman, Quezon City 1101, Philippines;
- Advanced Green Energy and Environment Department, Handong Global University, Pohang, Gyungbuk 37554, Korea; (K.A.); (H.P.); (S.D.T.)
| | - Pierangeli Vital
- Natural Sciences Research Institute, University of the Philippines Diliman, Quezon City 1101, Philippines;
| | - Karina Arellano
- Advanced Green Energy and Environment Department, Handong Global University, Pohang, Gyungbuk 37554, Korea; (K.A.); (H.P.); (S.D.T.)
| | - Haryung Park
- Advanced Green Energy and Environment Department, Handong Global University, Pohang, Gyungbuk 37554, Korea; (K.A.); (H.P.); (S.D.T.)
- HEM Inc., Business Incubator, Handong Global University, Pohang, Gyungbuk 37554, Korea;
| | - Svetoslav Dimitrov Todorov
- Advanced Green Energy and Environment Department, Handong Global University, Pohang, Gyungbuk 37554, Korea; (K.A.); (H.P.); (S.D.T.)
| | - Yosep Ji
- HEM Inc., Business Incubator, Handong Global University, Pohang, Gyungbuk 37554, Korea;
| | - Wilhelm Holzapfel
- Advanced Green Energy and Environment Department, Handong Global University, Pohang, Gyungbuk 37554, Korea; (K.A.); (H.P.); (S.D.T.)
- HEM Inc., Business Incubator, Handong Global University, Pohang, Gyungbuk 37554, Korea;
| |
Collapse
|
44
|
Abstract
Since the publication of the 2014 European Society for Paediatric Gastroenterology, Hepatology, and Nutrition Working Group (WG) on Probiotics and Prebiotics guidelines for the management of acute gastroenteritis (AGE), new evidence concerning the efficacy of probiotics has become available. This document provides updated recommendations on the use of probiotics for the treatment of AGE in previously presumed healthy infants and children. A systematic literature search was performed. All pooled analyses were explicitly performed for the current report. The WG graded the recommendations and assessed the certainty of the supporting evidence using the Grading of Recommendations, Assessment Development, and Evaluations tool. The recommendations were formulated if at least 2 randomized controlled trials that used a given probiotic were available. Despite the large number of identified trials, the WG could not identify 2 randomized controlled trial of high quality for any strain that provided benefit when used for treating AGE. The WG made weak recommendations for (in descending order in terms of the number of trials evaluating any given strain): Saccharomyces boulardii (low to very low certainty of evidence); Lactobacillus rhamnosus GG (very low certainty of evidence); L reuteri DSM 17938 (low to very low certainty of evidence); and L rhamnosus 19070-2 and L reuteri DSM 12246 (very low certainty of evidence). The WG made a strong recommendation against L helveticus R0052 and L rhamnosus R0011 (moderate certainty of evidence) and a weak recommendation against Bacillus clausii strains O/C, SIN, N/R, and T (very low certainty of evidence).
Collapse
|
45
|
Su GL, Ko CW, Bercik P, Falck-Ytter Y, Sultan S, Weizman AV, Morgan RL. AGA Clinical Practice Guidelines on the Role of Probiotics in the Management of Gastrointestinal Disorders. Gastroenterology 2020; 159:697-705. [PMID: 32531291 DOI: 10.1053/j.gastro.2020.05.059] [Citation(s) in RCA: 192] [Impact Index Per Article: 48.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Grace L Su
- Division of Gastroenterology and Hepatology, University of Michigan, Ann Arbor, Michigan; Gastroenterology Section, Veterans Administration Ann Arbor Healthcare System, Ann Arbor, Michigan
| | - Cynthia W Ko
- Division of Gastroenterology, University of Washington Medical School, Seattle, Washington
| | - Premysl Bercik
- Division of Gastroenterology, McMaster University, Hamilton, Ontario, Canada
| | - Yngve Falck-Ytter
- Division of Gastroenterology, Case Western Reserve University, Cleveland, Ohio; Louis Stokes Veterans Affairs Medical Center, Cleveland, Ohio
| | - Shahnaz Sultan
- Division of Gastroenterology, University of Minnesota, Minneapolis, Minnesota
| | - Adam V Weizman
- Division of Gastroenterology, Mount Sinai Hospital, Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Rebecca L Morgan
- Department of Health Research Methods, Evidence and Impact, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
46
|
Preidis GA, Weizman AV, Kashyap PC, Morgan RL. AGA Technical Review on the Role of Probiotics in the Management of Gastrointestinal Disorders. Gastroenterology 2020; 159:708-738.e4. [PMID: 32531292 PMCID: PMC8018518 DOI: 10.1053/j.gastro.2020.05.060] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Affiliation(s)
- Geoffrey A. Preidis
- Section of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, Baylor College of Medicine and Texas Children’s Hospital, Houston, Texas
| | - Adam V. Weizman
- Division of Gastroenterology, Mount Sinai Hospital, Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Purna C. Kashyap
- Enteric Neuroscience Program, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Rebecca L. Morgan
- Health Research Methods, Evidence and Impact, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
47
|
Protective action of Bacillus clausii probiotic strains in an in vitro model of Rotavirus infection. Sci Rep 2020; 10:12636. [PMID: 32724066 PMCID: PMC7387476 DOI: 10.1038/s41598-020-69533-7] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 07/06/2020] [Indexed: 02/06/2023] Open
Abstract
Rotavirus is the most common cause of acute gastroenteritis (AGE) in young children. Bacillus clausii (B. clausii) is a spore-forming probiotic that is able to colonize the gut. A mixture of four B. clausii strains (O/C, T, SIN and N/R) is commonly used for the treatment of AGE, and it has been demonstrated that it can reduce the duration and severity of diarrhea in children with AGE. Few studies have sought to characterize the mechanisms responsible for such beneficial effects. Intestinal effects of probiotics are likely to be strain-specific. We conducted a series of in vitro experiments investigating the activities of this mixture of B. clausii strains on biomarkers of mucosal barrier integrity and immune function in a cellular model of Rotavirus infection. B. clausii protected enterocytes against Rotavirus-induced decrease in trans-epithelial electrical resistance, and up-regulated expression of mucin 5AC and tight junction proteins (occludin and zonula occludens-1), all of which are important for effective mucosal barrier function. B. clausii also inhibited reactive oxygen species production and release of pro-inflammatory cytokines (interleukin-8 and interferon-β) in Rotavirus-infected cells, and down-regulated pro-inflammatory Toll-like receptor 3 pathway gene expression. Such mechanisms likely contributed to the observed protective effects of B. clausii against reduced cell proliferation and increased apoptosis in Rotavirus-infected enterocytes.
Collapse
|
48
|
Martinelli M, Banderali G, Bobbio M, Civardi E, Chiara A, D'Elios S, Lo Vecchio A, Olivero M, Peroni D, Romano C, Stronati M, Turra R, Viola I, Staiano A, Villani A. Probiotics' efficacy in paediatric diseases: which is the evidence? A critical review on behalf of the Italian Society of Pediatrics. Ital J Pediatr 2020; 46:104. [PMID: 32711569 PMCID: PMC7382135 DOI: 10.1186/s13052-020-00862-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 07/06/2020] [Indexed: 12/12/2022] Open
Abstract
During the last decade several paediatric studies have been published with different possible indications for probiotics, leading to a global increase of probiotics' market. Nevertheless, different study designs, multiple single/combined strains and small sample size still leave many uncertainties regarding their efficacy. In addition, different regulatory and quality control issues make still very difficult the interpretation of the clinical data. The objective of this review is to critically summarise the current evidence on probiotics' efficacy and safety on a different number of pathologies, including necrotizing enterocolitis, acute infectious diarrhoea, allergic diseases and functional gastrointestinal disorders in order to guide paediatric healthcare professionals on using evidence-based probiotics' strains. To identify relevant data, literature searches were performed including Medline-PubMed, the Cochrane Library and EMBASE databases. Considering probiotics strain-specific effects, the main focus was on individual probiotic strains and not on probiotics in general.
Collapse
Affiliation(s)
- Massimo Martinelli
- Department of Translational Medical Science, Section of Pediatrics, University of Naples "Federico II", Via S. Pansini, 5, 80131, Naples, Italy
| | - Giuseppe Banderali
- Clinical Department of Pediatrics and Neonatology, San Paolo Hospital, ASST Santi Paolo e Carlo, University of Milan, Milan, Italy
| | | | - Elisa Civardi
- Neonatal Intensive Care Unit, Fondazione IRCCS Policlinico San Matteo Pavia, Pavia, Italy
| | - Alberto Chiara
- Neonatal Intensive Care Unit, Fondazione IRCCS Policlinico San Matteo Pavia, Pavia, Italy
| | - Sofia D'Elios
- Department of Clinical and Experimental Medicine, Section of Pediatrics, University of Pisa, Pisa, Italy
| | - Andrea Lo Vecchio
- Department of Translational Medical Science, Section of Pediatrics, University of Naples "Federico II", Via S. Pansini, 5, 80131, Naples, Italy
| | | | - Diego Peroni
- Department of Clinical and Experimental Medicine, Section of Pediatrics, University of Pisa, Pisa, Italy
| | - Claudio Romano
- Pediatric Gastroenterology and Cystic Fibrosis Unit, University of Messina, Messina, Italy
| | - Mauro Stronati
- Neonatal Intensive Care Unit, Fondazione IRCCS Policlinico San Matteo Pavia, Pavia, Italy
| | | | - Irene Viola
- Pediatric Gastroenterology and Cystic Fibrosis Unit, University of Messina, Messina, Italy
| | - Annamaria Staiano
- Department of Translational Medical Science, Section of Pediatrics, University of Naples "Federico II", Via S. Pansini, 5, 80131, Naples, Italy.
| | - Alberto Villani
- Pediatric and Infectious Disease Unit, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| |
Collapse
|
49
|
Greuter T, Michel MC, Thomann D, Weigmann H, Vavricka SR. Randomized, Placebo-Controlled, Double-Blind and Open-Label Studies in the Treatment and Prevention of Acute Diarrhea With Enterococcus faecium SF68. Front Med (Lausanne) 2020; 7:276. [PMID: 32656217 PMCID: PMC7326027 DOI: 10.3389/fmed.2020.00276] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Accepted: 05/18/2020] [Indexed: 11/13/2022] Open
Abstract
Enterococcus faecium SF68® (SF68) is a licensed pharmaceutical for treatment and prevention of diarrhea in Austria, Italy and Switzerland. However, as for other probiotics, evidence for its efficacy is based on small to medium-sized studies. Four unpublished studies on the treatment of acute diarrhea and the prevention of antibiotic-associated diarrhea were analyzed: one randomized, double blind, placebo-controlled trial (RCT) for treatment (n = 1,143), one open-label study for treatment (n = 5,093), one RCT for prevention (n = 1,397) and one open-label study for prevention (n = 4,340). Patients in the treatment-arm and the open-label studies received SF68 (b.i.d. for the prevention studies, t.i.d. for the treatment trials) for 7 days. Primary end points were time to resolution of diarrhea (treatment) and percentage of development of diarrhea (prevention). The primary endpoint of the treatment study was met with a decreased time to resolution of diarrhea in SF68-treated patients compared to controls (median 3 vs. 4 days, p < 0.001). Time to resolution of secondary symptoms was also significantly reduced. Preventive treatment with SF68 was more effective than placebo with development of diarrhea in 8.6 vs. 16.2% (p < 0.001). Results from the open-label studies were consistent with the RCTs. The incidence of adverse events were low (1.1 and 1.4% in the RCT and 4.7 and 7.4% in the open-label studies). SF68 is effective and safe in the treatment of acute diarrhea and prevention of antibiotic-associated diarrhea.
Collapse
Affiliation(s)
- Thomas Greuter
- Division of Gastroenterology and Hepatology, University Hospital Zurich, Zurich, Switzerland.,Center for Gastroenterology, Zurich, Switzerland
| | - Martin C Michel
- Department of Pharmacology, Johannes Gutenberg University, Mainz, Germany
| | | | | | - Stephan R Vavricka
- Division of Gastroenterology and Hepatology, University Hospital Zurich, Zurich, Switzerland.,Department of Internal Medicine, GZO Zurich Regional Health Center, Wetzikon, Switzerland
| |
Collapse
|
50
|
Dziechciarz P, Krenke K, Szajewska H, Horvath A. Lactobacillus rhamnosus GG Usage in the Prevention of Gastrointestinal and Respiratory Tract Infections in Children with Gastroesophageal Reflux Disease Treated with Proton Pump Inhibitors: A Randomized Double-Blinded Placebo-Controlled Trial. Pediatr Gastroenterol Hepatol Nutr 2020; 23:251-258. [PMID: 32483546 PMCID: PMC7231745 DOI: 10.5223/pghn.2020.23.3.251] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 12/17/2019] [Accepted: 02/11/2020] [Indexed: 01/25/2023] Open
Abstract
PURPOSE Proton-pump inhibitors (PPIs) are frequently used to treat gastroesophageal reflux disease (GERD) in children, but recent evidence suggests a potential association between PPI treatment and some types of infections. The aim of this study was to assess the effectiveness of Lactobacillus rhamnosus GG (LGG) for the prevention of gastrointestinal and respiratory tract infections in children with GERD treated with PPI (omeprazol). METHODS Children younger than 5 years with GERD were assigned by a computer-generated list to receive LGG (109 colony-forming units) or placebo, twice daily, concomitantly with PPI treatment for 4-6 weeks; they were followed up for 12 weeks after therapy. The primary outcome measures were the percentage of children with a minimum of one episode of respiratory tract infection and the percentage of children with a minimum of one episode of gastrointestinal infection during the study. RESULTS Of 61 randomized children, 59 patients (LGG n=30; placebo n=29, mean age 11.3 months) were analyzed. There was no significant difference found between the LGG and placebo groups, either for the proportion of children with at least one respiratory tract infection (22/30 vs. 25/29, respectively; relative risk [RR] 0.85, 95% confidence interval [CI] 0.66-1.10) or for the proportion of children with at least one gastrointestinal infection (9/30 vs. 9/29, respectively; RR 0.97, 95% CI 0.45-2.09). CONCLUSION LGG was not effective in the prevention of infectious complications in children with GERD receiving PPI. Caution is needed in interpreting these results, as the study was terminated early due to slow subject recruitment.
Collapse
Affiliation(s)
- Piotr Dziechciarz
- Department of Pediatrics, Medical University of Warsaw, Warsaw, Poland
| | - Katarzyna Krenke
- Department of Pediatric Pneumonology and Allergy, Medical University of Warsaw, Warsaw, Poland
| | - Hania Szajewska
- Department of Pediatrics, Medical University of Warsaw, Warsaw, Poland
| | - Andrea Horvath
- Department of Pediatrics, Medical University of Warsaw, Warsaw, Poland
| |
Collapse
|