1
|
Yang S, Qiao J, Zhang M, Kwok LY, Matijašić BB, Zhang H, Zhang W. Prevention and treatment of antibiotics-associated adverse effects through the use of probiotics: A review. J Adv Res 2025; 71:209-226. [PMID: 38844120 DOI: 10.1016/j.jare.2024.06.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 05/18/2024] [Accepted: 06/03/2024] [Indexed: 06/29/2024] Open
Abstract
BACKGROUND The human gut hosts a diverse microbial community, essential for maintaining overall health. However, antibiotics, commonly prescribed for infections, can disrupt this delicate balance, leading to antibiotic-associated diarrhea, inflammatory bowel disease, obesity, and even neurological disorders. Recognizing this, probiotics have emerged as a promising strategy to counteract these adverse effects. AIM OF REVIEW This review aims to offer a comprehensive overview of the latest evidence concerning the utilization of probiotics in managing antibiotic-associated side effects. KEY SCIENTIFIC CONCEPTS OF REVIEW Probiotics play a crucial role in preserving gut homeostasis, regulating intestinal function and metabolism, and modulating the host immune system. These mechanisms serve to effectively alleviate antibiotic-associated adverse effects and enhance overall well-being.
Collapse
Affiliation(s)
- Shuwei Yang
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Huhhot 010018, China
| | - Jiaqi Qiao
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Huhhot 010018, China
| | - Meng Zhang
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Huhhot 010018, China
| | - Lai-Yu Kwok
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Huhhot 010018, China
| | | | - Heping Zhang
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Huhhot 010018, China
| | - Wenyi Zhang
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Huhhot 010018, China.
| |
Collapse
|
2
|
Wang L, Ren B, Wu S, Song H, Xiong L, Wang F, Shen X. Current research progress, opportunities, and challenges of Limosillactobacillus reuteri-based probiotic dietary strategies. Crit Rev Food Sci Nutr 2024:1-21. [PMID: 38920093 DOI: 10.1080/10408398.2024.2369946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/27/2024]
Abstract
Limosillactobacillus reuteri (L. reuteri), a type of Lactobacillus spp., stands out as the most extensively researched probiotic. Its remarkable intestinal adhesion has led to widespread applications in both the food and medical sectors. Notably, recent research highlights the probiotic efficacy of L. reuteri sourced from breast milk, particularly in influencing social behavior and mitigating atopic dermatitis. In this review, our emphasis is on surveying recent literature regarding the promotion of host's health by L. reuteri. We aim to provide a concise summary of the latest regulatory effects and potential mechanisms attributed to L. reuteri in the realms of metabolism, brain- and immune-related functions. The mechanism through which L. reuteri promotes host health by modulating the intestinal microenvironment primarily involves promoting intestinal epithelial renewal, bolstering intestinal barrier function, regulating gut microbiota and its metabolites, and suppressing inflammation and immune responses. Additionally, this review delves into new technologies, identifies shortcomings, and addresses challenges in current L. reuteri research. Finally, the application prospects of L. reuteri are provided. Therefore, a better understanding of the role and mechanisms of L. reuteri will contribute significantly to the development of new probiotic functional foods and enable precise, targeted interventions for various diseases.
Collapse
Affiliation(s)
- Luanfeng Wang
- College of Food Science and Engineering, Nanjing University of Finance and Economics/Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing, China
| | - Bo Ren
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, China
| | - Shufeng Wu
- College of Food Science and Engineering, Nanjing University of Finance and Economics/Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing, China
| | - Haizhao Song
- College of Food Science and Engineering, Nanjing University of Finance and Economics/Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing, China
| | - Ling Xiong
- College of Food Science and Engineering, Nanjing University of Finance and Economics/Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing, China
| | - Fang Wang
- College of Food Science and Engineering, Nanjing University of Finance and Economics/Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing, China
| | - Xinchun Shen
- College of Food Science and Engineering, Nanjing University of Finance and Economics/Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing, China
| |
Collapse
|
3
|
Tang M, Wang C, Xia Y, Tang J, Wang J, Shen L. Clostridioides difficile infection in inflammatory bowel disease: a clinical review. Expert Rev Anti Infect Ther 2024; 22:297-306. [PMID: 38676422 DOI: 10.1080/14787210.2024.2347955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 04/23/2024] [Indexed: 04/28/2024]
Abstract
INTRODUCTION Strong clinical data demonstrate that inflammatory bowel disease (IBD) is an independent risk factor for Clostridiodes difficile infection (CDI) and suggest a globally increased prevalence and severity of C. difficile coinfection in IBD patients (CDI-IBD). In addition to elderly individuals, children are also at higher risk of CDI-IBD. Rapid diagnosis is essential since the clinical manifestations of active IBD and CDI-IBD are indistinguishable. Antibiotics have been well established in the treatment of CDI-IBD, but they do not prevent recurrence. AREAS COVERED Herein, the authors focus on reviewing recent research advances on the new therapies of CDI-IBD. The novel therapies include gut microbiota restoration therapies (such as prebiotics, probiotics and FMT), immunotherapy (such as vaccines and monoclonal antibodies) and diet strategies (such as groningen anti-inflammatory diet and mediterranean diet). Future extensive prospective and placebo-controlled studies are required to evaluate their efficacy and long-term safety. EXPERT OPINION Available studies show that the prevalence of CDI-IBD is not optimistic. Currently, potential treatment options for CDI-IBD include a number of probiotics and novel antibiotics. This review updates the knowledge on the management of CDI in IBD patients, which is timely and important for GI doctors and scientists.
Collapse
Affiliation(s)
- Mengjun Tang
- Central Laboratory, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, China
| | - Chunhua Wang
- Central Laboratory, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, China
| | - Ying Xia
- Central Laboratory, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, China
| | - Jian Tang
- Central Laboratory, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, China
| | - Jiao Wang
- Central Laboratory, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, China
- School of Basic Medicine, Hubei University of Arts and Science, Xiangyang, China
| | - Liang Shen
- Central Laboratory, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, China
- Department of Clinical Laboratory, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, China
| |
Collapse
|
4
|
Dudzicz-Gojowy S, Więcek A, Adamczak M. The Role of Probiotics in the Prevention of Clostridioides difficile Infection in Patients with Chronic Kidney Disease. Nutrients 2024; 16:671. [PMID: 38474799 DOI: 10.3390/nu16050671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 02/21/2024] [Accepted: 02/23/2024] [Indexed: 03/14/2024] Open
Abstract
In patients suffering from chronic kidney disease (CKD), substantial unfavourable alterations in the intestinal microbiota composition, i.e., dysbiosis, have been noted. The main causes of such dysbiosis among others are insufficient dietary fibre content in the diet, fluid restrictions, medications used, and physical activity limitation. One clinically important consequence of dysbiosis in CKD patients is high risk of Clostridioides difficile infection (CDI). In observational studies, it was found that CDI is more frequent in CKD patients than in the general population. This appears to be related to high hospitalization rate and more often antibiotic therapy use, leading up to the occurrence of dysbiosis. Therefore, the use of probiotics in CKD patients may avert changes in the intestinal microbiota, which is the major risk factor of CDI. The aim of this review paper is to summarize the actual knowledge concerning the use of probiotics in CDI prevention in CKD patients in the context of CDI prevention in the general population.
Collapse
Affiliation(s)
- Sylwia Dudzicz-Gojowy
- Department of Nephrology, Transplantation and Internal Medicine, Medical University of Silesia, 40-027 Katowice, Poland
| | - Andrzej Więcek
- Department of Nephrology, Transplantation and Internal Medicine, Medical University of Silesia, 40-027 Katowice, Poland
| | - Marcin Adamczak
- Department of Nephrology, Transplantation and Internal Medicine, Medical University of Silesia, 40-027 Katowice, Poland
| |
Collapse
|
5
|
Huang N, Murphy L, Kandasamy S, Wahi G, Pernica JM. Caregiver knowledge and attitudes relating to paediatric pneumonia and antimicrobial stewardship: a qualitative study. Arch Dis Child 2024; 109:222-226. [PMID: 38041668 DOI: 10.1136/archdischild-2023-326080] [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: 07/09/2023] [Accepted: 11/17/2023] [Indexed: 12/03/2023]
Abstract
OBJECTIVE The objective of this study was to understand caregiver perspectives and experiences relating to the treatment of paediatric community-acquired pneumonia (CAP). DESIGN, SETTING AND PATIENTS This was a phenomenological qualitative study involving interviews with caregivers of young children in Hamilton, Ontario. Caregivers were asked open-ended questions relating to germ theory, pneumonia and the role of antibiotic treatment. The principles of conventional content analysis guided the coding and synthesis of the transcribed interviews. RESULTS Eleven caregivers were interviewed. Many knew that antibiotics were not effective against all types of infections and stated that there was an increased risk of developing resistance with frequent use. However, there were misconceptions that probiotics effectively mitigated antibiotic side effects, and few were familiar with the potential long-term consequences of antibiotic use in children.There was variability in the perceived severity of paediatric CAP. Some participants thought that antibiotic treatment would accelerate recovery and prevent caregivers from feeling helpless. However, others also thought it was inappropriate for physicians to prescribe antibiotics solely to make the caregiver feel better. Many caregivers also felt strongly that clinical follow-up and discussions on treatment risks/benefits would be desirable to counteract feelings of helplessness that result from being sent home without a prescription. CONCLUSION Recognising that parents may have misperceptions about antibiotic use for CAP (and may seek antibiotics without strong rationale) can inform clinicians' efforts to better educate and support caregivers in the emergency department. Care strategies informed by caregiver experiences can improve parent-provider communication and reduce antibiotic misuse.
Collapse
Affiliation(s)
- Nelson Huang
- Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Lara Murphy
- Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Sujane Kandasamy
- Department of Health Research Methods, Evidence and Impact, McMaster University, Hamilton, Ontario, Canada
- Department of Child and Youth Studies, Brock University, St Catherines, Ontario, Canada
| | - Gita Wahi
- Department of Health Research Methods, Evidence and Impact, McMaster University, Hamilton, Ontario, Canada
- Department of Pediatrics, McMaster University Faculty of Health Sciences, Hamilton, Ontario, Canada
| | - Jeffrey M Pernica
- Department of Health Research Methods, Evidence and Impact, McMaster University, Hamilton, Ontario, Canada
- Department of Pediatrics, McMaster University Faculty of Health Sciences, Hamilton, Ontario, Canada
| |
Collapse
|
6
|
Zhu Z, Yi B, Tang Z, Chen X, Li M, Xu T, Zhao Z, Tang C. Lactobacillus casei combined with Lactobacillus reuteri alleviate pancreatic cancer by inhibiting TLR4 to promote macrophage M1 polarization and regulate gut microbial homeostasis. BMC Cancer 2023; 23:1044. [PMID: 37904102 PMCID: PMC10614400 DOI: 10.1186/s12885-023-11557-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 10/22/2023] [Indexed: 11/01/2023] Open
Abstract
BACKGROUND Pancreatic cancer is a highly lethal disease with no effective treatments. Lactobacillus casei (L. casei) and Lactobacillus reuteri (L. reuteri) exhibited therapeutic effects on several cancers, but their roles in pancreatic cancer are unknown. This study aims to explore how L. casei & L. reuteri influence pancreatic cancer and the underlying mechanisms. METHODS Pancreatic cancer cells were treated with L. casei & L. reuteri and co-cultured with macrophages in a transwell system in vitro. Pancreatic cancer xenograft model was established and L. casei & L. reuteri was used to treat mice in vivo. MTT, CCK-8 assay or immunohistochemical staining were used to determine the proliferation of pancreatic cancer cells or tumor tissues. Transwell assay was applied to test the migration and invasion of pancreatic cells. RT-qPCR was utilized to assess TLR4 and MyD88 expressions in pancreatic cells or tumor tissues. WB, immunofluorescence staining, or flow cytometry was used to evaluate the M1/M2 polarization of macrophages. Besides, the composition of gut microbiota of tumor-bearing mice was determined by 16 S rRNA sequencing, and ultra-high performance liquid chromatography-mass spectrometry (UPLC-MS) untargeted metabolomics was used to evaluate the metabolic profiles of feces. RESULTS L. casei & L. reuteri inhibited the proliferation, migration, invasion of pancreatic cancer cells and pancreatic cancer cell-induced M2 polarization of macrophages by suppressing TLR4. Meanwhile, L. casei & L. reuteri repressed pancreatic cancer growth and promoted M1 macrophage polarization. Besides, L. casei & L. reuteri reduced fecal Alloprevotella and increased fecal azelate and glutamate in nude mice, while TLR4 inhibitor TAK-242 increased Clostridia UCG-014, azelate, uridine, methionine sulfoxide, oxypurinol, and decreased glyceryl monoester in the feces of pancreatic tumor-bearing mice. Fecal oxypurinol and glyceryl monoester levels were positively or negatively associated with gut Clostridia UCG-014 abundance, respectively. CONCLUSION L. casei & L. reuteri alleviate pancreatic cancer by inhibiting TLR4 to promote macrophage M1 polarization and regulate gut microbial homeostasis.
Collapse
Affiliation(s)
- Zemin Zhu
- Department of Hepatobiliary and Pancreatic Surgery, Zhuzhou Central Hospital, Zhuzhou, China
| | - Bo Yi
- Department of Hepatobiliary and Pancreatic Surgery, Zhuzhou Central Hospital, Zhuzhou, China
| | - Zikai Tang
- Department of Hepatobiliary and Pancreatic Surgery, Zhuzhou Central Hospital, Zhuzhou, China
| | - Xun Chen
- Department of Hepatobiliary and Pancreatic Surgery, Zhuzhou Central Hospital, Zhuzhou, China
- Department of Trauma Center, Zhuzhou Central Hospital, Zhuzhou, China
| | - Ming Li
- Department of Trauma Center, Zhuzhou Central Hospital, Zhuzhou, China
| | - Tao Xu
- Department of Hepatobiliary and Pancreatic Surgery, Zhuzhou Central Hospital, Zhuzhou, China
| | - Zhijian Zhao
- Department of Hepatobiliary and Pancreatic Surgery, Zhuzhou Central Hospital, Zhuzhou, China.
| | - Caixi Tang
- Department of Hepatobiliary and Pancreatic Surgery, Zhuzhou Central Hospital, Zhuzhou, China.
- Department of Trauma Center, Zhuzhou Central Hospital, Zhuzhou, China.
| |
Collapse
|
7
|
Abuqwider J, Di Porzio A, Barrella V, Gatto C, Sequino G, De Filippis F, Crescenzo R, Spagnuolo MS, Cigliano L, Mauriello G, Iossa S, Mazzoli A. Limosilactobacillus reuteri DSM 17938 reverses gut metabolic dysfunction induced by Western diet in adult rats. Front Nutr 2023; 10:1236417. [PMID: 37908302 PMCID: PMC10613642 DOI: 10.3389/fnut.2023.1236417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 09/18/2023] [Indexed: 11/02/2023] Open
Abstract
Introduction Microencapsulation of probiotic bacteria is an efficient and innovative new technique aimed at preserving bacterial survival in the hostile conditions of the gastrointestinal tract. However, understanding whether a microcapsule preserves the effectiveness of the bacterium contained within it is of fundamental importance. Methods Male Wistar rats aged 90 days were fed a control diet or a Western diet for 8 weeks, with rats fed the Western diet divided into three groups: one receiving the diet only (W), the second group receiving the Western diet and free L. reuteri DSM 17938 (WR), and the third group receiving the Western diet and microencapsulated L. reuteri DSM 17938 (WRM). After 8 weeks of treatment, gut microbiota composition was evaluated, together with occludin, one of the tight junction proteins, in the ileum and the colon. Markers of inflammation were also quantified in the portal plasma, ileum, and colon, as well as markers for gut redox homeostasis. Results The Western diet negatively influenced the intestinal microbiota, with no significant effect caused by supplementation with free and microencapsulated L. reuteri. However, L. reuteri, in both forms, effectively preserved the integrity of the intestinal barrier, thus protecting enterocytes from the development of inflammation and oxidative stress. Conclusion From these whole data, it emerges that L. reuteri DSM 17938 can be an effective probiotic in preventing the unhealthy consequences of the Western diet, especially in the gut, and that microencapsulation preserves the probiotic effects, thus opening the formulation of new preparations to be able to improve gut function independent of dietary habits.
Collapse
Affiliation(s)
- Jumana Abuqwider
- Department of Agricultural Sciences, University of Naples Federico II, Naples, Italy
| | - Angela Di Porzio
- Department of Biology, University of Naples Federico II, Naples, Italy
| | - Valentina Barrella
- Department of Biology, University of Naples Federico II, Naples, Italy
- NBFC, National Biodiversity Future Center, Palermo, Italy
| | - Cristina Gatto
- Department of Biology, University of Naples Federico II, Naples, Italy
| | - Giuseppina Sequino
- Department of Agricultural Sciences, University of Naples Federico II, Naples, Italy
| | - Francesca De Filippis
- Department of Agricultural Sciences, University of Naples Federico II, Naples, Italy
| | | | - Maria Stefania Spagnuolo
- Department of Bio-Agrofood Science, Institute for the Animal Production System in the Mediterranean Environment, National Research Council Naples (CNR-ISPAAM), Naples, Italy
| | - Luisa Cigliano
- Department of Biology, University of Naples Federico II, Naples, Italy
| | - Gianluigi Mauriello
- Department of Agricultural Sciences, University of Naples Federico II, Naples, Italy
| | - Susanna Iossa
- Department of Biology, University of Naples Federico II, Naples, Italy
- NBFC, National Biodiversity Future Center, Palermo, Italy
| | - Arianna Mazzoli
- Department of Biology, University of Naples Federico II, Naples, Italy
| |
Collapse
|
8
|
Liu J, Zhu R, Song J, Sohaib M, Wang S, Mao J, Qi J, Xiong X, Zhou W, Guo L. Limosilactobacillus reuteri consumption significantly reduces the total cholesterol concentration without affecting other cardiovascular disease risk factors in adults: A systematic review and meta-analysis. Nutr Res 2023; 117:1-14. [PMID: 37419064 DOI: 10.1016/j.nutres.2023.06.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 06/14/2023] [Accepted: 06/14/2023] [Indexed: 07/09/2023]
Abstract
As one of the most significant probiotics, Limosilactobacillus reuteri (L. reuteri) has been exploited as a nutritional supplement. We hypothesized that L. reuteri consumption might improve the significant risk factors of cardiovascular disease, including blood pressure, blood lipid, and blood glucose. However, previous clinical studies have shown controversial results. This study aims to explore the effect of L. reuteri consumption on these risk factors. PubMed, Embase, Scopus, the Cochrane Library, and Web of Science were searched for eligible randomized controlled trials published before May 2022. A total of 6 studies with 4 different L. reuteri strains and including 512 participants were included. The results showed that L. reuteri consumption significantly reduced total cholesterol (TC) by -0.26 mmol/L compared with the control group. In contrast, it did not affect systolic blood pressure, diastolic blood pressure, fasting blood glucose, high-density lipoprotein cholesterol, low-density lipoprotein cholesterol (LDL-C), or triglycerides. Subgroup analysis showed a significant reduction in TC when participants were <55 years old, had a body mass index between 25 and 30, or had hypercholesterolemia. In addition, TC decreased significantly when L. reuteri supplementation was >5 × 109 colony-forming unit or the length of the intervention was <12 weeks. Strain subgroup analysis showed that L. reuteri NCIMB 30242 significantly reduced TC and LDL-C. In conclusion, L. reuteri consumption has a significant TC-lowering effect, which can effectively reduce the risks of cardiovascular disease associated with hypercholesterolemia. However, the results do not support the effectiveness of L. reuteri consumption on other metabolic outcomes. Further examination of larger sample sizes is needed to confirm these findings.
Collapse
Affiliation(s)
- Jinshu Liu
- School of Nursing, Jilin University, Changchun, Jilin, China, 130021; School of Pharmacy, Jilin University, Changchun, Jilin, China, 130021
| | - Ruiting Zhu
- School of Nursing, Jilin University, Changchun, Jilin, China, 130021
| | - Jinping Song
- Rongchang Bio-Pharmaceutical Co. Ltd., Yantai, Shandong, China, 264006
| | | | - Saikun Wang
- School of Nursing, Jilin University, Changchun, Jilin, China, 130021
| | - Jing Mao
- School of Nursing, Jilin University, Changchun, Jilin, China, 130021
| | - Jiahe Qi
- School of Nursing, Jilin University, Changchun, Jilin, China, 130021
| | - Xuance Xiong
- Medical College, Beihua University, Jilin, Jilin, China, 132013
| | - Wei Zhou
- The First Hospital of Jilin University, Changchun, Jilin, China, 130021.
| | - Lirong Guo
- School of Nursing, Jilin University, Changchun, Jilin, China, 130021.
| |
Collapse
|
9
|
Shirley DA, Tornel W, Warren CA, Moonah S. Clostridioides difficile Infection in Children: Recent Updates on Epidemiology, Diagnosis, Therapy. Pediatrics 2023; 152:e2023062307. [PMID: 37560802 PMCID: PMC10471512 DOI: 10.1542/peds.2023-062307] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/31/2023] [Indexed: 08/11/2023] Open
Abstract
Clostridioides (formerly Clostridium) difficile is the most important infectious cause of antibiotic-associated diarrhea worldwide and a leading cause of healthcare-associated infection in the United States. The incidence of C. difficile infection (CDI) in children has increased, with 20 000 cases now reported annually, also posing indirect educational and economic consequences. In contrast to infection in adults, CDI in children is more commonly community-associated, accounting for three-quarters of all cases. A wide spectrum of disease severity ranging from asymptomatic carriage to severe diarrhea can occur, varying by age. Fulminant disease, although rare in children, is associated with high morbidity and even fatality. Diagnosis of CDI can be challenging as currently available tests detect either the presence of organism or disease-causing toxin but cannot distinguish colonization from infection. Since colonization can be high in specific pediatric groups, such as infants and young children, biomarkers to aid in accurate diagnosis are urgently needed. Similar to disease in adults, recurrence of CDI in children is common, affecting 20% to 30% of incident cases. Metronidazole has long been considered the mainstay therapy for CDI in children. However, new evidence supports the safety and efficacy of oral vancomycin and fidaxomicin as additional treatment options, whereas fecal microbiota transplantation is gaining popularity for recurrent infection. Recent advancements in our understanding of emerging epidemiologic trends and management of CDI unique to children are highlighted in this review. Despite encouraging therapeutic advancements, there remains a pressing need to optimize CDI therapy in children, particularly as it pertains to severe and recurrent disease.
Collapse
Affiliation(s)
| | | | - Cirle A. Warren
- Infectious Diseases and International Health, Department of Medicine
- Complicated C. difficile Clinic, UVA Health, University of Virginia, Charlottesville, Virginia
| | - Shannon Moonah
- Infectious Diseases and International Health, Department of Medicine
| |
Collapse
|
10
|
Peng Y, Ma Y, Luo Z, Jiang Y, Xu Z, Yu R. Lactobacillus reuteri in digestive system diseases: focus on clinical trials and mechanisms. Front Cell Infect Microbiol 2023; 13:1254198. [PMID: 37662007 PMCID: PMC10471993 DOI: 10.3389/fcimb.2023.1254198] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 08/07/2023] [Indexed: 09/05/2023] Open
Abstract
Objectives Digestive system diseases have evolved into a growing global burden without sufficient therapeutic measures. Lactobacillus reuteri (L. reuteri) is considered as a new potential economical therapy for its probiotic effects in the gastrointestinal system. We have provided an overview of the researches supporting various L. reuteri strains' application in treating common digestive system diseases, including infantile colic, diarrhea, constipation, functional abdominal pain, Helicobacter pylori infection, inflammatory bowel disease, diverticulitis, colorectal cancer and liver diseases. Methods The summarized literature in this review was derived from databases including PubMed, Web of Science, and Google Scholar. Results The therapeutic effects of L. reuteri in digestive system diseases may depend on various direct and indirect mechanisms, including metabolite production as well as modulation of the intestinal microbiome, preservation of the gut barrier function, and regulation of the host immune system. These actions are largely strain-specific and depend on the activation or inhibition of various certain signal pathways. It is well evidenced that L. reuteri can be effective both as a prophylactic measure and as a preferred therapy for infantile colic, and it can also be recommended as an adjuvant strategy to diarrhea, constipation, Helicobacter pylori infection in therapeutic settings. While preclinical studies have shown the probiotic potential of L. reuteri in the management of functional abdominal pain, inflammatory bowel disease, diverticulitis, colorectal cancer and liver diseases, its application in these disease settings still needs further study. Conclusion This review focuses on the probiotic effects of L. reuteri on gut homeostasis via certain signaling pathways, and emphasizes the importance of these probiotics as a prospective treatment against several digestive system diseases.
Collapse
Affiliation(s)
- Yijing Peng
- Department of Neonatology, Women’s Hospital of Jiangnan University, Wuxi Maternity and Child Health Care Hospital, Wuxi, China
- Wuxi Children’s Hospital, Children’s Hospital of Jiangnan University, Wuxi, China
| | - Yizhe Ma
- Department of Neonatology, Women’s Hospital of Jiangnan University, Wuxi Maternity and Child Health Care Hospital, Wuxi, China
- Department of Pediatric, Jiangyin People’s Hospital of Nantong University, Wuxi, China
| | - Zichen Luo
- Department of Neonatology, Women’s Hospital of Jiangnan University, Wuxi Maternity and Child Health Care Hospital, Wuxi, China
| | - Yifan Jiang
- School of Medicine, Nantong University, Nantong, China
| | - Zhimin Xu
- College of Resources and Environment, Innovative Institute for Plant Health, Zhongkai University of Agriculture and Engineering, Guangzhou, China
| | - Renqiang Yu
- Department of Neonatology, Women’s Hospital of Jiangnan University, Wuxi Maternity and Child Health Care Hospital, Wuxi, China
- Research Institute for Reproductive Health and Genetic Diseases, Women’s Hospital of Jiangnan University, Wuxi Maternity and Child Health Care Hospital, Wuxi, China
| |
Collapse
|
11
|
Darbandi A, Banar M, Koupaei M, Afifirad R, Asadollahi P, Bafandeh E, Rasooli I, Emamie A, Navidifar T, Owlia P. Clinical efficacy of probiotics in prevention of infectious diseases among hospitalized patients in ICU and non-ICU wards in clinical randomized trials: A systematic review. Health Sci Rep 2023; 6:e1469. [PMID: 37547361 PMCID: PMC10400784 DOI: 10.1002/hsr2.1469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 07/13/2023] [Accepted: 07/18/2023] [Indexed: 08/08/2023] Open
Abstract
Background and Aims The present study aimed to review probiotics' clinical efficacy in preventing infectious diseases among hospitalized patients in ICU and non-ICU wards. Methods A search of Medline, EMBASE, The Cochrane Library, Science Direct, Open Grey, and Google Scholar was conducted for eligible publications from 2002 to 2020 following the requirements outlined in the PRISMA guideline. The search strategy was based on the combination of the following terms: "probiotics," "prebiotics," "synbiotics," and "cross-infection." The logical operators "AND" (or the equivalent operator for the databases) and "OR" (e.g., probiotics OR prebiotics OR synbiotics) were used. Results The results indicated that the probiotic consumption caused a significant reduction in antibiotic-associated diarrhea (AAD) and Clostridioides difficile infection (CDI) in 2/8 randomized clinical trials (RCTs) investigating AAD/CDI. Also, 5/12 clinical trials highlighted the considerable effects of probiotics on the reduction or prevention of ventilator associated pneumoniae (VAP), so the mean prevalence of VAP was lower in the probiotic group than in the placebo group. The total rate of nosocomial infections among preterm infants was nonsignificantly higher in the probiotic group compared to the control group. Conclusion This systematic review shows that the administration of probiotics has moderate preventive or mitigating effects on the occurrence of VAP in ICU patients, CDI, AAD, and nosocomial infections among children. Consequently, applying antibiotics along with the proper probiotic species can be advantageous.
Collapse
Affiliation(s)
- Atieh Darbandi
- Molecular Microbiology Research CenterShahed UniversityTehranIran
| | - Maryam Banar
- Department of PathobiologySchool of Public Health, Tehran University of Medical SciencesTehranIran
| | - Maryam Koupaei
- Department of Microbiology and ImmunologySchool of Medicine, Kashan University of Medical SciencesKashanIran
| | - Roghayeh Afifirad
- Department of MicrobiologySchool of Medicine, Tehran University of Medical SciencesTehranIran
| | - Parisa Asadollahi
- Department of MicrobiologyFaculty of Medicine, Ilam University of Medical SciencesIlamIran
| | - Elnaz Bafandeh
- Department of Medical BiotechnologyFaculty of Medicine, Lorestan University of Medical SciencesKhorramabadIran
| | - Iraj Rasooli
- Molecular Microbiology Research Center, Faculty of SciencesShahed UniversityTehranIran
| | - Amir Emamie
- Department of PathobiologySchool of Public Health, Tehran University of Medical SciencesTehranIran
| | | | - Parviz Owlia
- Molecular Microbiology Research CenterShahed UniversityTehranIran
- Molecular Microbiology Research Center, Faculty of SciencesShahed UniversityTehranIran
| |
Collapse
|
12
|
Sun X, Kong J, Zhu S, Liu C. A systematic review and meta-analysis: the therapeutic and preventive effect of Lactobacillus reuteri DSM 17,938 addition in children with diarrhea. BMC Gastroenterol 2023; 23:141. [PMID: 37147591 PMCID: PMC10161659 DOI: 10.1186/s12876-023-02778-4] [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] [Received: 10/24/2022] [Accepted: 04/21/2023] [Indexed: 05/07/2023] Open
Abstract
OBJECTIVE To summarize the effect of adding Lactobacillus reuteri in the treatment plan for diarrheal disease in children, and analyze the potential of probiotics in preventing the occurrence of diarrheal disease. METHODS Search for randomized controlled trials of Lactobacillus reuteri for the treatment and prevention of diarrhea in the Pubmed, Web of science, Medline, and Cochrane databases. Data such as the number of diarrhea patients, time, length of stay, clinical symptoms and effect of diarrhea prevention were extracted for meta-analysis. Relative risk and confidence interval (RR and 95% CI) were used as outcome indicators. RESULTS 963 participants in the 9 RCTs came from multiple countries/regions. Compared with placebo/no intervention, the number of diarrhea patients in the Lactobacillus reuteri group was significantly reduced on the day 1 (RR = 0.87, 95%CI: 0.78-0.97) and day 2 (RR = 0.61, 95%CI: 0.44-0.83). Cumulative statistics analysis showed that the effect was stable and significant starting on the 4th day after treatment. A few studies have shown that Lactobacillus reuteri can reduce the time of diarrhea, the number of days with watery stools, and days of hospital stay. However, it has no effect on the occurrence of nosocomial diarrhea (RR = 1.11, 95%CI: 0.68-1.83), rotavirus diarrhea (RR = 1.46, 95%CI: 0.78-2.72), antibiotic-related diarrhea (RR = 1.76, 95%CI: 0.77-4.05), and diarrhea (RR = 1.35, 95%CI: 0.95-1.92). CONCLUSION Addition of Lactobacillus reuteri in the treatment plan has a significant effect on reducing the number of diarrhea and reducing the symptoms of diarrhea, but has no obvious effect on the prevention of diarrhea. Combining probiotics and improving the ability of probiotics to respond is the focus of attention.
Collapse
Affiliation(s)
- Xiaoqi Sun
- Department of Clinical Nutrition, Shengjing Hospital of China Medical University, Shenyang, China.
| | - Juan Kong
- Department of Clinical Nutrition, Shengjing Hospital of China Medical University, Shenyang, China
| | - Shuotong Zhu
- Department of Gerneral Surgery, Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Chengjiang Liu
- Department of Gastroenterology, Anhui Medical University, He Fei, China
| |
Collapse
|
13
|
Liu Y, Hoang TK, Park ES, Freeborn J, Okeugo B, Tran DQ, Rhoads JM. Probiotic-educated Tregs are more potent than naïve Tregs for immune tolerance in stressed new-born mice. Benef Microbes 2023; 14:73-84. [PMID: 36815493 PMCID: PMC10124588 DOI: 10.3920/bm2022.0095] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/24/2023]
Abstract
When new-born mice are subjected to acute maternal separation stress, cow-milk based formula feeding, and brief recurrent hypoxia with cold stress, they develop gut inflammation similar to the phenotype of neonatal necrotizing enterocolitis, characterised by an increase in gut mucosal effector T (Teffs) and reduced Foxp3+ regulatory T (Tregs) cells. The imbalance can be prevented by probiotic Limosilactobacillus reuteri DSM 17938 (LR 17938). We hypothesised that LR 17938 could potentiate a tolerogenic function of Tregs. To analyse whether LR 17938 can educate Tregs to improve their tolerogenic potency during neonatal stress, we isolated T cells (Tregs and Teffs) from 'donor' mice fed with either LR 17938 (107 cfu) or control media. The cells were adoptively transferred (AT) by intraperitoneal injection (5 × 105 cells/mouse) to new-born (d5) recipient mice. Mice were then separated from their dams, fed formula by gavage, and exposed to hypoxia and cold stress (NeoStress) for 4 days. We analysed the percentage of Tregs in CD4+T helper cells in the intestine (INT) and mesenteric lymph nodes (MLN) of recipient mice. We found that: (1) the percentage of Tregs in the INT and MLN following NeoStress were significantly reduced compared to dam-fed unstressed mice; (2) AT of either naïve Tregs or LR-educated Tregs to mice with Neostress increased the percentage of Tregs in the INT and MLN compared to the percentage in NeoStress mice without Treg treatment; however, LR-educated Tregs increased the Tregs significantly more than naïve Tregs; and (3) AT of LR-educated Tregs reduced pro-inflammatory CD44+Foxp3-NonTregs and inflammatory CX3CR1+ dendritic cells in the intestinal mucosa of NeoStress mice. In conclusion, adoptive transfer of Tregs promotes the generation of and/or migration of endogenous Tregs in the intestinal mucosa of recipient mice. Importantly, probiotic-educated Tregs are more potent than naïve Tregs to enhance immune tolerance following neonatal stress.
Collapse
Affiliation(s)
- Y Liu
- Department of Pediatrics, Division of Gastroenterology, McGovern Medical School, University of Texas Health Science Center at Houston, 6431 Fannin Street, MSB 3.137, Houston, TX 77030, USA
| | - T K Hoang
- Department of Pediatrics, Division of Gastroenterology, McGovern Medical School, University of Texas Health Science Center at Houston, 6431 Fannin Street, MSB 3.137, Houston, TX 77030, USA
| | - E S Park
- Department of Pediatrics, Division of Gastroenterology, McGovern Medical School, University of Texas Health Science Center at Houston, 6431 Fannin Street, MSB 3.137, Houston, TX 77030, USA
| | - J Freeborn
- Department of Pediatrics, Division of Gastroenterology, McGovern Medical School, University of Texas Health Science Center at Houston, 6431 Fannin Street, MSB 3.137, Houston, TX 77030, USA
| | - B Okeugo
- Department of Pediatrics, Division of Gastroenterology, McGovern Medical School, University of Texas Health Science Center at Houston, 6431 Fannin Street, MSB 3.137, Houston, TX 77030, USA
| | - D Q Tran
- Department of Pediatrics, Division of Gastroenterology, McGovern Medical School, University of Texas Health Science Center at Houston, 6431 Fannin Street, MSB 3.137, Houston, TX 77030, USA
| | - J M Rhoads
- Department of Pediatrics, Division of Gastroenterology, McGovern Medical School, University of Texas Health Science Center at Houston, 6431 Fannin Street, MSB 3.137, Houston, TX 77030, USA
| |
Collapse
|
14
|
Calvigioni M, Bertolini A, Codini S, Mazzantini D, Panattoni A, Massimino M, Celandroni F, Zucchi R, Saba A, Ghelardi E. HPLC-MS-MS quantification of short-chain fatty acids actively secreted by probiotic strains. Front Microbiol 2023; 14:1124144. [PMID: 36937254 PMCID: PMC10020375 DOI: 10.3389/fmicb.2023.1124144] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 02/20/2023] [Indexed: 03/06/2023] Open
Abstract
Introduction Short-chain fatty acids (SCFAs) are the main by-products of microbial fermentations occurring in the human intestine and are directly involved in the host's physiological balance. As impaired gut concentrations of acetic, propionic, and butyric acids are often associated with systemic disorders, the administration of SCFA-producing microorganisms has been suggested as attractive approach to solve symptoms related to SCFA deficiency. Methods In this research, nine probiotic strains (Bacillus clausii NR, OC, SIN, and T, Bacillus coagulans ATCC 7050, Bifidobacterium breve DSM 16604, Limosilactobacillus reuteri DSM 17938, Lacticaseibacillus rhamnosus ATCC 53103, and Saccharomyces boulardii CNCM I-745) commonly included in commercial formulations were tested for their ability to secrete SCFAs by using an improved protocol in high-performance liquid chromatography coupled to tandem mass spectrometry (HPLC-MS-MS). Results The developed method was highly sensitive and specific, showing excellent limits of detection and quantification of secreted SCFAs. All tested microorganisms were shown to secrete acetic acid, with only B. clausii and S. boulardii additionally able to produce propionic and butyric acids. Quantitative differences in the secretion of SCFAs were also evidenced. Discussion The experimental approach described in this study may contribute to the characterization of probiotics as SCFA-producing organisms, a crucial stage toward their application to improve SCFA deficiency.
Collapse
Affiliation(s)
- Marco Calvigioni
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Andrea Bertolini
- Department of Surgical, Medical and Molecular Pathology and Critical Care Medicine, University of Pisa, Pisa, Italy
| | - Simone Codini
- Department of Surgical, Medical and Molecular Pathology and Critical Care Medicine, University of Pisa, Pisa, Italy
| | - Diletta Mazzantini
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Adelaide Panattoni
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Mariacristina Massimino
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Francesco Celandroni
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Riccardo Zucchi
- Department of Surgical, Medical and Molecular Pathology and Critical Care Medicine, University of Pisa, Pisa, Italy
| | - Alessandro Saba
- Department of Surgical, Medical and Molecular Pathology and Critical Care Medicine, University of Pisa, Pisa, Italy
| | - Emilia Ghelardi
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
- Research Center Nutraceuticals and Food for Health–Nutrafood, University of Pisa, Pisa, Italy
| |
Collapse
|
15
|
Schneider R, Sant'Anna A. Using probiotics in paediatric populations. Paediatr Child Health 2022; 27:482-502. [PMID: 36583073 PMCID: PMC9792287 DOI: 10.1093/pch/pxac087] [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: 03/01/2021] [Accepted: 04/25/2022] [Indexed: 12/28/2022] Open
Abstract
This statement defines probiotics and reviews the most recent literature on their use in paediatrics. Many studies have examined the potential benefit of probiotics, but significant variation in the strains and doses of probiotics used, the patient populations studied, and in study design, have led to heterogeneous results. Present evidence suggests that probiotics can decrease mortality and lower incidence of necrotizing enterocolitis in preterm and low birth weight neonates. Probiotics may also be beneficial in reducing feeding intolerance. In infants, probiotics may be considered to reduce symptoms of colic. In older children, probiotics can be considered to prevent antibiotic-associated diarrhea and Clostridium difficile -associated diarrhea. Probiotic supplements used in conjunction with standard therapy can help with Helicobacter pylori eradication and decrease the side effects of treatment. Lactobacillus species can be considered to treat irritable bowel syndrome. Probiotics can also be considered to help prevent atopic dermatitis and eczema. To optimize paediatric policy and practice, large, quality studies are needed to determine what types and combinations of probiotics are most efficacious.
Collapse
Affiliation(s)
- Rilla Schneider
- Canadian Paediatric Society, Nutrition and Gastroenterology Committee, Ottawa, Ontario, Canada
| | - Ana Sant'Anna
- Canadian Paediatric Society, Nutrition and Gastroenterology Committee, Ottawa, Ontario, Canada
| |
Collapse
|
16
|
Schneider R, Sant'Anna A. L’utilisation des probiotiques dans la population pédiatrique. Paediatr Child Health 2022; 27:482-502. [PMID: 36583070 PMCID: PMC9792288 DOI: 10.1093/pch/pxac086] [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: 03/01/2021] [Accepted: 04/25/2022] [Indexed: 12/28/2022] Open
Abstract
Le présent document de principes définit les probiotiques et fournit une analyse des publications scientifiques les plus récentes sur leur utilisation en pédiatrie. De nombreuses études ont évalué les avantages potentiels des probiotiques, mais en raison des variations importantes dans les souches et les doses utilisées, des populations de patients étudiées et des méthodologies privilégiées, les résultats sont hétérogènes. Selon les données probantes à jour, les probiotiques peuvent réduire le taux de mortalité et l’incidence d’entérocolite nécrosante chez les nouveau-nés prématurés et de petit poids à la naissance. Ils peuvent également être bénéfiques pour réduire l’intolérance alimentaire. Chez les nourrissons, on peut envisager de les utiliser pour limiter les symptômes de coliques, et chez les enfants plus âgés, pour prévenir la diarrhée associée aux antibiotiques ou au Clostridium difficile . Les suppléments de probiotiques utilisés conjointement avec un traitement standard peuvent contribuer à éradiquer l’Helicobacter pylori et à atténuer les effets secondaires du traitement. On peut envisager d’utiliser des espèces de Lactobacillus pour traiter le syndrome du côlon irritable ou de recourir à des probiotiques pour contribuer à prévenir la dermatite atopique et l’eczéma. Afin d’optimiser les politiques et les pratiques en pédiatrie, de vastes études de qualité devront être réalisées pour déterminer les types et les combinaisons de probiotiques les plus efficaces.
Collapse
Affiliation(s)
- Rilla Schneider
- Société canadienne de pédiatrie, comité de nutrition et de gastroentérologie, Ottawa (Ontario)Canada
| | - Ana Sant'Anna
- Société canadienne de pédiatrie, comité de nutrition et de gastroentérologie, Ottawa (Ontario)Canada
| |
Collapse
|
17
|
Dargenio VN, Cristofori F, Dargenio C, Giordano P, Indrio F, Celano G, Francavilla R. Use of Limosilactobacillus reuteri DSM 17938 in paediatric gastrointestinal disorders: an updated review. Benef Microbes 2022; 13:221-242. [PMID: 35212258 DOI: 10.3920/bm2021.0151] [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
Strains of lactobacilli are the most widely used probiotics and can be found in a large variety of food products and food supplements throughout the world. In this study, the evidence on Limosilactobacillus reuteri DSM 17938 (LR DSM 17938) has been reviewed. This species secretes reuterin and other substances singularly or in microvesicles, inhibiting pathogen growth and interacting with the intestinal microbiota and mucosa, restoring homeostasis. The use of LR DSM 17938 has been exploited in several pathological conditions. Preclinical research has shown that this probiotic can ameliorate dysbiosis and, by interacting with intestinal mucosal cells, can raise the pain threshold and promote gastrointestinal motility. These aspects are amongst the significant components in functional gastrointestinal disorders, such as colic and regurgitation in infants, functional abdominal pain and functional constipation in children and adolescents. This strain can decrease the duration of acute diarrhoea and hospitalization for acute gastroenteritis but does not seem to prevent nosocomial diarrhoea and antibiotic-associated diarrhoea. Because of its ability to survive in the gastric environment, it has been tested in Helicobacter pylori infection, showing a significant decrease of antibiotic-associated side effects and a tendency to increase the eradication rate. Finally, all these studies have shown the excellent safety of LR DSM 17938 even at higher dosages. In conclusion data from various clinical trials here reviewed can guide the clinician to find the correct dose, frequency of administration, and therapy duration.
Collapse
Affiliation(s)
- V N Dargenio
- Interdisciplinary Department of Medicine, Paediatric Section. University of Bari Aldo Moro, Children's Hospital 'Giovanni XXIII', Via Amendola 207, 70126 Bari, Italy
| | - F Cristofori
- Interdisciplinary Department of Medicine, Paediatric Section. University of Bari Aldo Moro, Children's Hospital 'Giovanni XXIII', Via Amendola 207, 70126 Bari, Italy
| | - C Dargenio
- Interdisciplinary Department of Medicine, Paediatric Section. University of Bari Aldo Moro, Children's Hospital 'Giovanni XXIII', Via Amendola 207, 70126 Bari, Italy
| | - P Giordano
- Interdisciplinary Department of Medicine, Paediatric Section. University of Bari Aldo Moro, Children's Hospital 'Giovanni XXIII', Via Amendola 207, 70126 Bari, Italy
| | - F Indrio
- Department of Paediatrics, University of Foggia, Via Pinto 1, 71100 Foggia, Italy
| | - G Celano
- Department of Soil, Plant and Food Science, University of Bari Aldo Moro, Via Amendola 265/a, 70126 Bari, Italy
| | - R Francavilla
- Interdisciplinary Department of Medicine, Paediatric Section. University of Bari Aldo Moro, Children's Hospital 'Giovanni XXIII', Via Amendola 207, 70126 Bari, Italy
| |
Collapse
|
18
|
Transglutaminase-catalyzed modification of fish skin gelatin enhanced the protection of microcapsules to Limosilactobacillus reuteri. FOOD BIOSCI 2022. [DOI: 10.1016/j.fbio.2022.101961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
19
|
Bao W, He Y, Yu J, Liu M, Yang X, Ta N, Zhang E, Liang C. Regulatory Effect of Lactiplantibacillus plantarum 2-33 on Intestinal Microbiota of Mice With Antibiotic-Associated Diarrhea. Front Nutr 2022; 9:921875. [PMID: 35757257 PMCID: PMC9218693 DOI: 10.3389/fnut.2022.921875] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2022] [Accepted: 05/13/2022] [Indexed: 12/20/2022] Open
Abstract
Diarrhea is one of the common adverse reactions in antibiotic treatment, which is usually caused by the imbalance of intestinal flora, and probiotics play an important role in the structure of intestinal flora. Therefore, this experiment studied the regulatory effect of Lactiplantibacillus plantarum 2-33 on antibiotic-associated diarrhea (AAD) mice. First, the AAD mice model was established by the mixed antibiotic solution of gentamicin sulfate and cefradine. Then, the physiological indexes and diarrhea of mice were observed and recorded by gastric perfusion of low dose (1.0 × 107 CFU/ml), medium dose (1.0 × 108CFU/ml), and high dose (1.0 × 109 CFU/ml) strain 2-33. 16S rRNA gene V3-V4 regions were sequenced in colon contents of mice in control group, model group, self-healing group, and experimental group, respectively, and the diversity of intestinal flora and gene function prediction were analyzed. The results showed that the intestinal flora of AAD mice was not significantly regulated by gastric perfusion of strain 2-33 to 7 days, but the relative abundance and diversity of intestinal flora of AAD mice were significantly improved by gastric perfusion to 14 days (p < 0.05). In addition, at the genus level, the relative abundance of Lactobacillus increased significantly, and the relative abundance of Enterococcus and Bacillus decreased significantly (p < 0.05). In addition, the regulation of strain 2-33 on intestinal flora of AAD mice was time- and dose-dependent, short-term gastric perfusion, and low dose had no significant effect (p > 0.05). Strain 2-33 can significantly increase the levels of anti-inflammatory cytokines IL-4 and IL-10, significantly decrease the levels of proinflammatory cytokines TNF-α and IFN-γ (p < 0.05), and can also adjust carbohydrate metabolism, amino acid metabolism, and energy metabolism to normal levels, thus accelerating the recovery of intestinal flora structure of AAD mice. In summary, strain 2-33 can improve the structure and diversity of intestinal flora of AAD mice, balance the level of substance and energy metabolism, and play a positive role in relieving diarrhea, maintaining and improving the intestinal microecological balance.
Collapse
Affiliation(s)
- Wuyundalai Bao
- College of Food Science and Engineering, Inner Mongolia Agricultural University, Hohhot, China
| | - Yuxing He
- College of Food Science and Engineering, Inner Mongolia Agricultural University, Hohhot, China
| | - Jinghe Yu
- College of Food Science and Engineering, Inner Mongolia Agricultural University, Hohhot, China
| | - Mingchao Liu
- College of Food Science and Engineering, Inner Mongolia Agricultural University, Hohhot, China
| | - Xiaofeng Yang
- College of Food Science and Engineering, Inner Mongolia Agricultural University, Hohhot, China
| | - Na Ta
- College of Food Science and Engineering, Inner Mongolia Agricultural University, Hohhot, China
| | - Enxin Zhang
- College of Food Science and Engineering, Inner Mongolia Agricultural University, Hohhot, China
| | - Chengyuan Liang
- College of Food Science and Engineering, Inner Mongolia Agricultural University, Hohhot, China
| |
Collapse
|
20
|
Ragan MV, Wala SJ, Goodman SD, Bailey MT, Besner GE. Next-Generation Probiotic Therapy to Protect the Intestines From Injury. Front Cell Infect Microbiol 2022; 12:863949. [PMID: 35837474 PMCID: PMC9273849 DOI: 10.3389/fcimb.2022.863949] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Accepted: 05/25/2022] [Indexed: 12/20/2022] Open
Abstract
Probiotics are live microorganisms that, when administered in adequate amounts, provide health benefits to the host. Some strains of the probiotic Lactobacillus reuteri (L. reuteri) have both antimicrobial and anti-inflammatory properties that may be exploited for the treatment and prevention of different gastrointestinal diseases, including necrotizing enterocolitis (NEC) and Clostridioides difficile (C. difficile) infection. Our laboratory has developed a new delivery system for L. reuteri in which the probiotic is incubated with biocompatible, semipermeable, porous dextranomer microspheres (DM) that can be loaded with beneficial and diffusible cargo. L. reuteri can be induced to form a biofilm by incubating the bacteria on the surface of these microspheres, which enhances the efficacy of the probiotic. Loading the DM with sucrose or maltose induces L. reuteri to produce more biofilm, further increasing the efficacy of the probiotic. Using a rat model of NEC, L. reuteri administered in its biofilm state significantly increases animal survival, reduces the incidence of NEC, preserves gut barrier function, and decreases intestinal inflammation. In a murine model of Clostridiodes difficile infection, L. reuteri administered in its biofilm state decreases colitis when administered either before or after C. difficile induction, demonstrating both prophylactic and therapeutic efficacy. There are currently no FDA-approved probiotic preparations for human use. An FDA-approved phase I clinical trial of L. reuteri in its biofilm state in healthy adults is currently underway. The results of this trial will be used to support a phase 1 clinical trial in neonates, with the goal of utilizing L. reuteri in its biofilm state to prevent NEC in premature neonates in the future.
Collapse
Affiliation(s)
- Mecklin V. Ragan
- Center for Perinatal Research, Department of Pediatric Surgery, Columbus, OH, United States
| | - Samantha J. Wala
- Center for Perinatal Research, Department of Pediatric Surgery, Columbus, OH, United States
| | - Steven D. Goodman
- Nationwide Children’s Hospital, The Ohio State University, Columbus, OH, United States
| | - Michael T. Bailey
- Nationwide Children’s Hospital, The Ohio State University, Columbus, OH, United States
| | - Gail E. Besner
- Center for Perinatal Research, Department of Pediatric Surgery, Columbus, OH, United States
- *Correspondence: Gail E. Besner,
| |
Collapse
|
21
|
Effects of Lacidophilin Tablets, Yogurt, and Bifid Triple Viable Capsules on the Gut Microbiota of Mice with Antibiotic-Associated Diarrhea. THE CANADIAN JOURNAL OF INFECTIOUS DISEASES & MEDICAL MICROBIOLOGY = JOURNAL CANADIEN DES MALADIES INFECTIEUSES ET DE LA MICROBIOLOGIE MEDICALE 2022; 2022:6521793. [PMID: 35360462 PMCID: PMC8964159 DOI: 10.1155/2022/6521793] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 02/08/2022] [Accepted: 02/23/2022] [Indexed: 12/14/2022]
Abstract
Antibiotic-associated diarrhea (AAD) is a common morbidity caused by antibiotic use and is characterized by the dysbiosis of the gut microbiota. Several clinical trials have shown that probiotics can prevent AAD. This study aimed at investigating the effects of Lacidophilin tablets (LB), yogurt (YG), and bifid triple viable capsules (BT) on the gut microbiota of mice with AAD. Mice with diarrhea were randomly allocated to treatment groups or the control group and were treated with either LB, YG, BT, or vehicle control. The body weight, diarrhea scores, cecum index, and cecal length were determined. Fecal samples of all mice were analyzed using 16S rRNA high-throughput sequencing. The results showed that LB, YG, and BT significantly decreased the diarrhea scores and inhibited increases in the cecum index and cecal length induced by AAD. In addition, they significantly changed the composition and richness of the gut microbiota. Specifically, they increased the abundance of the phylum Firmicutes and decreased the abundance of the phyla Bacteroidetes and the family Bacteroidaceae. Treatment with LB and YG also decreased the abundance of the phylum Proteobacteria and only LB could mediate the reduced levels of Lactobacillaceae in AAD mice. At the genus level, YG and BT treatment decreased the abundance of Bacteroides or Parasutterella. To our surprise, only LB treatment dramatically increased the abundance of Lactobacillus and decreased that of potential pathogens, such as Bacteroides, Parabacteroides, and Parasutterella, to almost normal values. Our findings indicate that LB, YG, and BT ameliorated diarrhea by regulating the composition and structure of the gut microbiota and that LB plays an important role in regulating the gut microbiota.
Collapse
|
22
|
Gu X, Sim JX, Lee WL, Cui L, Chan YF, Chang ED, Teh YE, Zhang AN, Armas F, Chandra F, Chen H, Zhao S, Lee Z, Thompson JR, Ooi EE, Low JG, Alm EJ, Kalimuddin S. Gut Ruminococcaceae levels at baseline correlate with risk of antibiotic-associated diarrhea. iScience 2022; 25:103644. [PMID: 35005566 PMCID: PMC8718891 DOI: 10.1016/j.isci.2021.103644] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 11/19/2021] [Accepted: 12/13/2021] [Indexed: 12/11/2022] Open
Abstract
Antibiotic-associated diarrhea (AAD) affects a significant proportion of patients receiving antibiotics. We sought to understand if differences in the gut microbiome would influence the development of AAD. We administered a 3-day course of amoxicillin-clavulanate to 30 healthy adult volunteers, and analyzed their stool microbiome, using 16S rRNA gene sequencing, at baseline and up to 4 weeks post antibiotic administration. Lower levels of gut Ruminococcaceae were significantly and consistently observed from baseline until day 7 in participants who developed AAD. Overall, participants who developed AAD experienced a greater decrease in microbial diversity. The probability of AAD could be predicted based on qPCR-derived levels of Faecalibacterium prausnitzii at baseline. Our findings suggest that a lack of gut Ruminococcaceae influences development of AAD. Quantification of F. prausnitzii in stool prior to antibiotic administration may help identify patients at risk of AAD, and aid clinicians in devising individualized treatment regimens to minimize such adverse effects.
Collapse
Affiliation(s)
- Xiaoqiong Gu
- Antimicrobial Resistance Interdisciplinary Research Group, Singapore-MIT Alliance for Research and Technology, 1 Create Way, Singapore 138602, Singapore
- Campus for Research Excellence and Technological Enterprise (CREATE), Singapore 138602, Singapore
| | - Jean X.Y. Sim
- Department of Infectious Diseases, Singapore General Hospital, Academia Level 3, 20 College Road, Singapore 169856, Singapore
| | - Wei Lin Lee
- Antimicrobial Resistance Interdisciplinary Research Group, Singapore-MIT Alliance for Research and Technology, 1 Create Way, Singapore 138602, Singapore
- Campus for Research Excellence and Technological Enterprise (CREATE), Singapore 138602, Singapore
| | - Liang Cui
- Antimicrobial Resistance Interdisciplinary Research Group, Singapore-MIT Alliance for Research and Technology, 1 Create Way, Singapore 138602, Singapore
- Campus for Research Excellence and Technological Enterprise (CREATE), Singapore 138602, Singapore
| | - Yvonne F.Z. Chan
- Department of Infectious Diseases, Singapore General Hospital, Academia Level 3, 20 College Road, Singapore 169856, Singapore
| | - Ega Danu Chang
- Antimicrobial Resistance Interdisciplinary Research Group, Singapore-MIT Alliance for Research and Technology, 1 Create Way, Singapore 138602, Singapore
- Campus for Research Excellence and Technological Enterprise (CREATE), Singapore 138602, Singapore
| | - Yii Ean Teh
- Department of Infectious Diseases, Singapore General Hospital, Academia Level 3, 20 College Road, Singapore 169856, Singapore
| | - An-Ni Zhang
- Department of Biological Engineering, Massachusetts Institute of Technology, 21 Ames Street, Cambridge, MA 02142, USA
| | - Federica Armas
- Antimicrobial Resistance Interdisciplinary Research Group, Singapore-MIT Alliance for Research and Technology, 1 Create Way, Singapore 138602, Singapore
- Campus for Research Excellence and Technological Enterprise (CREATE), Singapore 138602, Singapore
| | - Franciscus Chandra
- Antimicrobial Resistance Interdisciplinary Research Group, Singapore-MIT Alliance for Research and Technology, 1 Create Way, Singapore 138602, Singapore
- Campus for Research Excellence and Technological Enterprise (CREATE), Singapore 138602, Singapore
| | - Hongjie Chen
- Antimicrobial Resistance Interdisciplinary Research Group, Singapore-MIT Alliance for Research and Technology, 1 Create Way, Singapore 138602, Singapore
- Campus for Research Excellence and Technological Enterprise (CREATE), Singapore 138602, Singapore
| | - Shijie Zhao
- Department of Biological Engineering, Massachusetts Institute of Technology, 21 Ames Street, Cambridge, MA 02142, USA
| | - Zhanyi Lee
- Antimicrobial Resistance Interdisciplinary Research Group, Singapore-MIT Alliance for Research and Technology, 1 Create Way, Singapore 138602, Singapore
- Campus for Research Excellence and Technological Enterprise (CREATE), Singapore 138602, Singapore
| | - Janelle R. Thompson
- Singapore Centre for Environmental Life Sciences Engineering, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore
- Asian School of the Environment, Nanyang Technological University, 62 Nanyang Drive, Singapore 637459, Singapore
| | - Eng Eong Ooi
- Program in Emerging Infectious Diseases, Duke-NUS Medical School, 8 College Road, Singapore 169857, Singapore
- Viral Research and Experimental Medicine Center, SingHealth Duke-NUS Academic Medical Centre (ViREMiCS), 20 College Road, Singapore 169856, Singapore
- Saw Swee Hock School of Public Health, National University of Singapore, 12 Science Drive 2, Singapore 117549, Singapore
| | - Jenny G. Low
- Department of Infectious Diseases, Singapore General Hospital, Academia Level 3, 20 College Road, Singapore 169856, Singapore
- Program in Emerging Infectious Diseases, Duke-NUS Medical School, 8 College Road, Singapore 169857, Singapore
- Viral Research and Experimental Medicine Center, SingHealth Duke-NUS Academic Medical Centre (ViREMiCS), 20 College Road, Singapore 169856, Singapore
| | - Eric J. Alm
- Antimicrobial Resistance Interdisciplinary Research Group, Singapore-MIT Alliance for Research and Technology, 1 Create Way, Singapore 138602, Singapore
- Campus for Research Excellence and Technological Enterprise (CREATE), Singapore 138602, Singapore
- Department of Biological Engineering, Massachusetts Institute of Technology, 21 Ames Street, Cambridge, MA 02142, USA
- Center for Microbiome Informatics and Therapeutics, Massachusetts Institute of Technology, Building E25-321, Cambridge, MA 02139, USA
- Broad Institute of MIT and Harvard, 415 Main Street, Cambridge, MA 02142, USA
| | - Shirin Kalimuddin
- Department of Infectious Diseases, Singapore General Hospital, Academia Level 3, 20 College Road, Singapore 169856, Singapore
- Program in Emerging Infectious Diseases, Duke-NUS Medical School, 8 College Road, Singapore 169857, Singapore
| |
Collapse
|
23
|
Pyle S. Human Gut Microbiota and the Influence of Probiotics, Prebiotics, and Micronutrients. COMPREHENSIVE GUT MICROBIOTA 2022:271-288. [DOI: 10.1016/b978-0-12-819265-8.00076-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
24
|
Matsuura T, Kyokane K, Yamada S, Kuno Y. The development of the cure of the functional intestinal disorder based on the differences of gut microbiota in aged patients: A randomized clinical trial. Medicine (Baltimore) 2021; 100:e27696. [PMID: 34871255 PMCID: PMC8568417 DOI: 10.1097/md.0000000000027696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 08/02/2021] [Accepted: 10/20/2021] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Constipation, which is not an organic disease in the lower gastrointestinal tract, is a gastrointestinal symptom characteristic of elderly patients. Complaints of dyschezia increase with age, and it is difficult to treat in many cases. This study aimed to determine the appropriate treatment and its effects on intestinal immunity in elderly patients experiencing chronic constipation. METHODS Patients experiencing difficulty defecating were randomly divided into 2 groups. Group A was given only laxatives, whereas Group B was given laxatives combined with probiotics as an intervention. Both groups were compared based on the degree of improvement in constipation and its effects on the intestinal environment. RESULTS There was a significant improvement in constipation of elderly patients when probiotics were administered in combination with a laxative, suggesting that it may be a more effective treatment. Furthermore, the changes in the intestinal flora, examined before and after the intervention, tended to be associated with improvement of constipation. CONCLUSION The results indicated that the improvement of intestinal flora was somewhat achieved by relieving constipation. Because intestinal bacteria significantly influence intestinal immunity and, thus, systemic immunity of the entire body, the development of better treatments for constipation would help to improve both the intestinal environment and immune function in the elderly.
Collapse
|
25
|
Yang Y, Zhang W, Huan H, Xia W, Chen Y, Wang P, Liu Y. Construction of an Integrated mCherry Red Fluorescent Protein Expression System for Labeling and Tracing in Lactiplantibacillus plantarum WCFS1. Front Microbiol 2021; 12:690270. [PMID: 34239511 PMCID: PMC8258168 DOI: 10.3389/fmicb.2021.690270] [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: 04/02/2021] [Accepted: 05/20/2021] [Indexed: 11/17/2022] Open
Abstract
Thorough intestinal adhesion and colonization greatly promote the probiotic properties of lactic acid bacteria (LAB). Labeling and tracing with fluorescent proteins are effective and reliable for studying the in vivo physiological activities of LAB including localization, adhesion, and colonization. Lactiplantibacillus plantarum WCFS1 was successfully traced with a red fluorescent protein (RFP), which was expressed by the bacteria-carrying recombinant plasmids. In this study, we aimed to construct a stable RFP mCherry expression system, whose encoding gene was integrated into the bacterial chromosome via double-crossed homologous recombination, and use it for labeling WCFS1 with the goal of avoiding the potential loss of non-chromosomal plasmids along with intestinal growth. First, the constitutive expression of the mCherry protein was improved after adjusting the length of the spacer between the promoter and the gene start codon. Then, the optimized mCherry gene expression cassette was integrated into the chromosome of WCFS1. The resulting strain had normal unimpaired growth and strong fluorescent signals, even after 100 generations, indicating its stability. Furthermore, quantitative polymerase chain reaction (PCR) results revealed a strong positive correlation between the fluorescence intensity of the strain and the number of viable cells, demonstrating its potential usage for the quantification of in vivo WCFS1 cells. Finally, the increased adhesion ability of WCFS1 due to the recombinant expression of the bsh gene was visualized and evaluated using fluorescence intensity, the results of which were consistent with those obtained using the previously established quantification methods. These results suggest that the chromosomal-integrated mCherry labeling system can be extensively used to examine the distribution, colonization, and survival of LAB in vivo in order to determine the mechanism of its probiotic function.
Collapse
Affiliation(s)
- Yao Yang
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, China
| | - Wenjun Zhang
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, China
| | - Hailin Huan
- Jiangsu Academy of Agricultural Sciences, Nanjing, China
| | - Wenxu Xia
- Geneception (Shanghai) Bio-technology Co., Ltd., Shanghai, China
| | - Ying Chen
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, China
| | - Peijuan Wang
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, China
| | - Yanrong Liu
- Nanjing Institute of Product Quality Inspection, Nanjing, China
| |
Collapse
|
26
|
Li B, Qiu H, Zheng N, Wu G, Gu Y, Zhong J, Hong Y, Ma J, Zhou W, Sheng L, Li H. Integrated Metagenomic and Transcriptomic Analyses Reveal the Dietary Dependent Recovery of Host Metabolism From Antibiotic Exposure. Front Cell Dev Biol 2021; 9:680174. [PMID: 34222250 PMCID: PMC8250461 DOI: 10.3389/fcell.2021.680174] [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] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Accepted: 05/20/2021] [Indexed: 01/10/2023] Open
Abstract
The balance of gut microbiome is essential for maintaining host metabolism homeostasis. Despite widespread antibiotic use, the potential long-term detrimental consequences of antibiotics for host health are getting more and more attention. However, it remains unclear whether diet affects the post-antibiotic recovery of gut microbiome and host metabolism. In this study, through metagenomic sequencing and hepatic transcriptome analysis, we investigated the divergent impacts of short-term vancomycin (Vac), or combination of ciprofloxacin and metronidazole (CM) treatment on gut microbiome and host metabolism, as well as their recovery extent from antibiotic exposure on chow diet (CD) and high-fat diet (HFD). Our results showed that short-term Vac intervention affected insulin signaling, while CM induced more functional changes in the microbiome. However, Vac-induced long-term (45 days) changes of species were more apparent when recovered on CD than HFD. The effects of antibiotic intervention on host metabolism were long-lasting, antibiotic-specific, and diet-dependent. The number of differentially expressed gene was doubled by Vac than CM, but was comparable after recovery on CD as revealed by the hepatic transcriptomic analysis. In contrast, HFD intake during recovery could worsen the extent of post-antibiotic recovery by altering infection, immunity, and cancer-related pathways in short-term Vac-exposed rats and by shifting endocrine system-associated pathways in CM-exposed rats. Together, the presented data demonstrated the long-term recovery extent after different antibiotic exposure was diet-related, highlighting the importance of dietary management during post-antibiotic recovery.
Collapse
Affiliation(s)
- Bingbing Li
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Huihui Qiu
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ningning Zheng
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Gaosong Wu
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yu Gu
- Human Phenome Institute, Fudan University, Shanghai, China
| | - Jing Zhong
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Huzhou Key Laboratory of Molecular Medicine, Huzhou Central Hospital, Huzhou, China
| | - Ying Hong
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Junli Ma
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Wen Zhou
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| | - Lili Sheng
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Houkai Li
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
27
|
[Systematic review: clinical evidence of probiotics in the prevention of antibiotic-associated diarrhoea]. MMW Fortschr Med 2021; 163:19-26. [PMID: 33844181 DOI: 10.1007/s15006-021-9762-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
BACKGROUND Antibiotic-associated diarrhoea (AAD) is the most common intestinal side effect of an antibiotic therapy. Various probiotics or probiotic combinations are often used preventively while taking antibiotics for the prevention of AAD. METHOD This review is based on a systematic literature research in MEDLINE and EMBASE. 7 probiotics are presented with regard to their effectiveness and evidence in the prevention of AAD. Only preparations classified by the World Gastroenerology Organization (WGO) with evidence levels 1-3 for the prevention of AAD were taken into account. 37 clinical studies, including 33 RCTs, were evaluated. RESULTS Saccharomyces (S.) boulardii CNCM I-745 is the most extensively studied probiotic regarding the prevention of AAD. It has shown evidence-based efficacy in all patient groups (outpatients and hospitalized children and adults). Lactobacillus rhamnosus GG also has a good evidence regarding the prevention of AAD in children and outpatient adults. The other probiotics and probiotic combinations evaluated in the present study only show efficacy in hospitalized patients or only show very limited evidence regarding their efficacy in the prevention of AAD due to the underlying study design or the small number of patients. CONCLUSION The effect of probiotics is strain-specific, no general statement can be made about the efficacy of probiotics in the prevention of AAD. In principle, it is advisable to select a probiotic with an evidence-based effect such as S. boulardii CNCM I-745 or Lactobacillus rhamnosus GG to prevent AAD.
Collapse
|
28
|
Forster CS, Hsieh MH, Cabana MD. Perspectives from the Society for Pediatric Research: Probiotic use in urinary tract infections, atopic dermatitis, and antibiotic-associated diarrhea: an overview. Pediatr Res 2021; 90:315-327. [PMID: 33288875 PMCID: PMC8180529 DOI: 10.1038/s41390-020-01298-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Revised: 11/03/2020] [Accepted: 11/09/2020] [Indexed: 02/08/2023]
Abstract
Probiotics have received significant attention within both the scientific and lay communities for their potential health-promoting properties, including the treatment or prevention of various conditions in children. In this article, we review the published data on use of specific probiotic strains for three common pediatric conditions: the prevention of urinary tract infections and antibiotic-associated diarrhea and the treatment of atopic dermatitis. Research into the utility of specific probiotic strains is of varying quality, and data are often derived from small studies and case series. We discuss the scientific merit of these studies, their overall findings regarding the utility of probiotics for these indications, issues in reporting of methods, and results from these clinical trials, as well as future areas of investigation.
Collapse
Affiliation(s)
- Catherine S. Forster
- grid.239560.b0000 0004 0482 1586Department of Pediatrics, Children’s National Health System, Washington, DC USA
| | - Michael H. Hsieh
- grid.239560.b0000 0004 0482 1586Department of Pediatrics, Children’s National Health System, Washington, DC USA
| | - Michael D. Cabana
- grid.251993.50000000121791997Department of Pediatrics, Children’s Hospital at Montefiore and the Albert Einstein School of Medicine, Bronx, NY USA
| |
Collapse
|
29
|
Łukasik J, Guo Q, Boulos L, Szajewska H, Johnston BC. Probiotics for the prevention of antibiotic-associated adverse events in children-A scoping review to inform development of a core outcome set. PLoS One 2020; 15:e0228824. [PMID: 32469907 PMCID: PMC7259577 DOI: 10.1371/journal.pone.0228824] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2020] [Accepted: 05/08/2020] [Indexed: 11/19/2022] Open
Abstract
INTRODUCTION Routine use of probiotics during antibiotic therapy in children remains a subject of discussion. To facilitate synthesis of individual study results and guideline formulation, it is important to assess predefined, similar, and clinically important outcomes. Core outcome sets are a proposed solution for this issue. The aim of this review was to document choice, design, and heterogeneity of outcomes in studies that assessed the effects of probiotics used for the prevention of antibiotic-associated adverse events in children. METHODS A scoping literature search covering three major databases was performed. Studies that evaluated oral probiotics' use concomitant with antibiotic therapy in children were included. Data on outcome definitions, measurement instruments, and follow-up were extracted. The outcomes were assigned to predefined core areas and domains. Data were analyzed descriptively. RESULTS Thirty-seven studies were included in this review. Diarrhea, the most commonly reported outcome, had diagnostic criteria clearly defined only in 21 studies. In total, 16 different definitions of diarrhea were identified. Diarrhea duration, severity, and etiology were reported in 9, 4, and 7 studies, respectively. Twenty studies assessed gastrointestinal symptoms other than diarrhea. Seven studies reported outcomes related to resource use or the economic impact of the intervention. Only 2 studies assessed outcomes related to life impact. None of the studies predefined adverse events of probiotic use. CONCLUSIONS Identified outcomes were characterized by substantial heterogeneity. The majority of outcomes were not designed to evaluate endpoints of real-life relevance. Results from this review suggest the need for a new core outcome set consisting of outcomes important for decision-making.
Collapse
Affiliation(s)
- Jan Łukasik
- Department of Pediatrics, Medical University of Warsaw, Warsaw, Poland
| | - Qin Guo
- Department of Pediatrics, West China Second University Hospital, Chengdu, China
| | | | - Hania Szajewska
- Department of Pediatrics, Medical University of Warsaw, Warsaw, Poland
| | - Bradley C. Johnston
- Department of Community Health and Epidemiology, Dalhousie University, Halifax, Canada
| |
Collapse
|
30
|
Hui H, Wu Y, Zheng T, Zhou S, Tan Z. Bacterial Characteristics in Intestinal Contents of Antibiotic-Associated Diarrhea Mice Treated with Qiweibaizhu Powder. Med Sci Monit 2020; 26:e921771. [PMID: 32398636 PMCID: PMC7245059 DOI: 10.12659/msm.921771] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Accepted: 02/18/2020] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Qiweibaizhu powder (QWBZP) is a classical prescription of traditional Chinese medicine (TCM) to treat diarrhea in pediatric patients. Its use in health care practices and interventions has shown its effect on antibiotic-associated diarrhea (AAD). It is known that the occurrence of AAD is related to an imbalance of intestinal micro-ecology. Previous studies found that QWBZP could regulate the amount of some cultured microbes and the activities of lactase and sucrase in AAD mice. In order to investigate the treatment mechanism of QWBZP on AAD, we studied the effect of QWBZP on intestinal bacteria in a community of AAD mice. MATERIAL AND METHODS AAD mice were established by administrating the mixture of gentamycin sulfate and cefradine at the dose of 23.33 mL·kg⁻¹·d⁻¹ for 5 days. Then the AAD mice were gavaged with QWBZP decoction for 4 days and gradually recovered to a normal status. On the tenth day, the intestinal contents of mice were collected, and then the DNA was extracted for 16S rRNA sequencing followed by analysis. RESULTS The analysis of bacterial 16S rRNA sequencing showed the Simpson index was decreased and the Shannon index was increased in AAD mice treated with QWBZP compared to the model group; there was no significant difference between the control group and the treatment group (P>0.05). Principle co-ordinates analysis (PCoA) indicated that there was a shorter distance between the control group and the treatment group than that between the control group and model group. At the phylum level, use of antibiotics decreased the relative abundance of Actinobacteria, Bacteroidetes, and Proteobacteria, but increased the abundance of Firmicutes and Verrucomicrobia, and the reverse changes occurred after treated with QWBZP. At the genus level, the abundance of Bacteroides and Ochrobacitrum increased in the model group, while an opposite result was observed in the treatment group. Moreover, the relative abundance of Osillospira decreased in the model group and increased in the treatment group. Genus Dorea, Coprococcus and Blautia in the model group were higher than those in the control group and further increased in the treatment group. CONCLUSIONS These results indicated that QWBZP improved the diarrhea syndrome with restoring the diversity and adjusting the structures of bacteria in mice intestine, which might reveal the therapeutic mechanism of QWBZP on treating AAD.
Collapse
|
31
|
Li X, Wu Y, Xu Z, Chen J, Li Y, Xing H, Zhang X, Yuan J. Effects of Hetiao Jianpi Decoction on Intestinal Injury and Repair in Rats with Antibiotic-Associated Diarrhea. Med Sci Monit 2020; 26:e921745. [PMID: 32062668 PMCID: PMC7043351 DOI: 10.12659/msm.921745] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Through observing the changes of indexes of the intestinal mucosal barrier and intestinal flora in rats, we explored the mechanism by which Hetiao Jianpi Decoction (HTJPD) treats antibiotic-associated diarrhea (AAD) by repairing intestinal mucosal injury and regulating intestinal flora. MATERIAL AND METHODS Samples of colon tissues were collected for HE staining. Enzyme-linked immunosorbent assay (ELISA) was used to assess levels of diamine oxidase (DAO) and D-lactic acid in rat plasma and the expression of secretory immunoglobulin A (SIgA) in colon tissue. We assessed the abundance of intestinal contents by high-throughput sequencing of the 16S rRNA gene. RESULTS Compared with the Model group, the muscle layer and intestinal mucosal edema were improved, and the continuity was restored; the levels of DAO and D-lactic acid in plasma decreased, and the SIgA level were increased in the HTJPD group. The structure of the intestinal flora changed, as indicated by increased levels of certain beneficial bacteria (Verrucomicrobia, Actinobacteria, CF231, and Akkermansia), decreased levels of pathogenic bacteria (Spirochaetes and Treponema), and increased species diversity. CONCLUSIONS By improving the permeability and immune function of the intestinal mucosa, Hetiao Jianpi decoction prevented the occurrence of AAD by repairing the intestinal mucosal damage and regulating the structure and diversity of intestinal flora.
Collapse
Affiliation(s)
- Xiaoya Li
- College of Basic Medical Science, Yunnan University of Chinese Medicine, Kunming, Yunnan, China (mainland)
| | - Yueying Wu
- College of First Clinical Medical Science, Yunnan University of Chinese Medicine, Kunming, Yunnan, China (mainland)
| | - Zhenyuan Xu
- College of Basic Medical Science, Yunnan University of Chinese Medicine, Kunming, Yunnan, China (mainland)
| | - Jing Chen
- College of Basic Medical Science, Yunnan University of Chinese Medicine, Kunming, Yunnan, China (mainland).,Provincial Innovation Team of Yunnan University of Chinese Medicine for Traditional Chinese Medicine to Regulate Human Microecology, Kunming, Yunnan, China (mainland)
| | - Yuqing Li
- College of Basic Medical Science, Yunnan University of Chinese Medicine, Kunming, Yunnan, China (mainland).,Provincial Innovation Team of Yunnan University of Chinese Medicine for Traditional Chinese Medicine to Regulate Human Microecology, Kunming, Yunnan, China (mainland)
| | - Haijing Xing
- College of Basic Medical Science, Yunnan University of Chinese Medicine, Kunming, Yunnan, China (mainland).,Provincial Innovation Team of Yunnan University of Chinese Medicine for Traditional Chinese Medicine to Regulate Human Microecology, Kunming, Yunnan, China (mainland)
| | - Xiaomei Zhang
- College of Basic Medical Science, Yunnan University of Chinese Medicine, Kunming, Yunnan, China (mainland).,Provincial Innovation Team of Yunnan University of Chinese Medicine for Traditional Chinese Medicine to Regulate Human Microecology, Kunming, Yunnan, China (mainland)
| | - Jiali Yuan
- Provincial Innovation Team of Yunnan University of Chinese Medicine for Traditional Chinese Medicine to Regulate Human Microecology, Kunming, Yunnan, China (mainland)
| |
Collapse
|
32
|
Prophylactic use of probiotics for gastrointestinal disorders in children. THE LANCET CHILD & ADOLESCENT HEALTH 2019; 3:655-662. [PMID: 31279590 DOI: 10.1016/s2352-4642(19)30182-8] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 05/13/2019] [Accepted: 05/15/2019] [Indexed: 02/07/2023]
Abstract
The gastrointestinal microbiome is a hot topic in clinical research. Beneficial effects of selected probiotics in the prevention of gastrointestinal disorders are mainly restricted to acute gastroenteritis, antibiotic-associated diarrhoea, infantile colic, and necrotising enterocolitis. However, no broad consensus exists to recommend the use of probiotics in the prevention of these conditions, mainly because of the different design of the studies done so far, resulting in little evidence for specific strains, dosages, and indications. More well designed studies are needed before recommendations can be proposed. At this stage, the evidence is insufficient to recommend the routine use of probiotics in infants and children for the prevention of gastrointestinal disorders.
Collapse
|
33
|
Guo Q, Goldenberg JZ, Humphrey C, El Dib R, Johnston BC, Cochrane IBD Group. Probiotics for the prevention of pediatric antibiotic-associated diarrhea. Cochrane Database Syst Rev 2019; 4:CD004827. [PMID: 31039287 PMCID: PMC6490796 DOI: 10.1002/14651858.cd004827.pub5] [Citation(s) in RCA: 109] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND Antibiotics alter the microbial balance commonly resulting in antibiotic-associated diarrhea (AAD). Probiotics may prevent AAD via providing gut barrier, restoration of the gut microflora, and other potential mechanisms of action. OBJECTIVES The primary objectives were to assess the efficacy and safety of probiotics (any specified strain or dose) used for the prevention of AAD in children. SEARCH METHODS MEDLINE, Embase, CENTRAL, CINAHL, and the Web of Science (inception to 28 May 2018) were searched along with registers including the ISRCTN and Clinicaltrials.gov. We also searched the NICE Evidence Services database as well as reference lists from relevant articles. SELECTION CRITERIA Randomized, parallel, controlled trials in children (0 to 18 years) receiving antibiotics, that compare probiotics to placebo, active alternative prophylaxis, or no treatment and measure the incidence of diarrhea secondary to antibiotic use were considered for inclusion. DATA COLLECTION AND ANALYSIS Study selection, data extraction, and risk of bias assessment were conducted independently by two authors. Dichotomous data (incidence of AAD, adverse events) were combined using a pooled risk ratio (RR) or risk difference (RD), and continuous data (mean duration of diarrhea) as mean difference (MD), along with corresponding 95% confidence interval (95% CI). We calculated the number needed to treat for an additional beneficial outcome (NNTB) where appropriate. For studies reporting on microbiome characteristics using heterogeneous outcomes, we describe the results narratively. The certainty of the evidence was evaluated using GRADE. MAIN RESULTS Thirty-three studies (6352 participants) were included. Probiotics assessed included Bacillus spp., Bifidobacterium spp., Clostridium butyricum, Lactobacilli spp., Lactococcus spp., Leuconostoc cremoris, Saccharomyces spp., orStreptococcus spp., alone or in combination. The risk of bias was determined to be high in 20 studies and low in 13 studies. Complete case (patients who did not complete the studies were not included in the analysis) results from 33 trials reporting on the incidence of diarrhea show a precise benefit from probiotics compared to active, placebo or no treatment control.After 5 days to 12 weeks of follow-up, the incidence of AAD in the probiotic group was 8% (259/3232) compared to 19% (598/3120) in the control group (RR 0.45, 95% CI 0.36 to 0.56; I² = 57%, 6352 participants; NNTB 9, 95% CI 7 to 13; moderate certainty evidence). Nineteen studies had loss to follow-up ranging from 1% to 46%. After making assumptions for those lost, the observed benefit was still statistically significant using an extreme plausible intention-to-treat (ITT) analysis, wherein the incidence of AAD in the probiotic group was 12% (436/3551) compared to 19% (664/3468) in the control group (7019 participants; RR 0.61; 95% CI 0.49 to 0.77; P <0.00001; I² = 70%). An a priori available case subgroup analysis exploring heterogeneity indicated that high dose (≥ 5 billion CFUs per day) is more effective than low probiotic dose (< 5 billion CFUs per day), interaction P value = 0.01. For the high dose studies the incidence of AAD in the probiotic group was 8% (162/2029) compared to 23% (462/2009) in the control group (4038 participants; RR 0.37; 95% CI 0.30 to 0.46; P = 0.06; moderate certainty evidence). For the low dose studies the incidence of AAD in the probiotic group was 8% (97/1155) compared to 13% (133/1059) in the control group (2214 participants; RR 0.68; 95% CI 0.46 to 1.01; P = 0.02). Again, assumptions for loss to follow-up using an extreme plausible ITT analysis was statistically significant. For high dose studies the incidence of AAD in the probiotic group was 13% (278/2218) compared to 23% (503/2207) in control group (4425 participants; RR 0.54; 95% CI 0.42 to 0.70; P <0.00001; I² = 68%; moderate certainty evidence).None of the 24 trials (4415 participants) that reported on adverse events reported any serious adverse events attributable to probiotics. Adverse event rates were low. After 5 days to 4 weeks follow-up, 4% (86/2229) of probiotics participants had an adverse event compared to 6% (121/2186) of control participants (RD 0.00; 95% CI -0.01 to 0.01; P < 0.00001; I² = 75%; low certainty evidence). Common adverse events included rash, nausea, gas, flatulence, abdominal bloating, and constipation.After 10 days to 12 weeks of follow-up, eight studies recorded data on our secondary outcome, the mean duration of diarrhea; with probiotics reducing diarrhea duration by almost one day (MD -0.91; 95% CI -1.38 to -0.44; P <0.00001; low certainty evidence). One study reported on microbiome characteristics, reporting no difference in changes with concurrent antibiotic and probiotic use. AUTHORS' CONCLUSIONS The overall evidence suggests a moderate protective effect of probiotics for preventing AAD (NNTB 9, 95% CI 7 to 13). Using five criteria to evaluate the credibility of the subgroup analysis on probiotic dose, the results indicate the subgroup effect based on high dose probiotics (≥ 5 billion CFUs per day) was credible. Based on high-dose probiotics, the NNTB to prevent one case of diarrhea is 6 (95% CI 5 to 9). The overall certainty of the evidence for the primary endpoint, incidence of AAD based on high dose probiotics was moderate due to the minor issues with risk of bias and inconsistency related to a diversity of probiotic agents used. Evidence also suggests that probiotics may moderately reduce the duration of diarrhea, a reduction by almost one day. The benefit of high dose probiotics (e.g. Lactobacillus rhamnosus orSaccharomyces boulardii) needs to be confirmed by a large well-designed multi-centered randomized trial. It is premature to draw firm conclusions about the efficacy and safety of 'other' probiotic agents as an adjunct to antibiotics in children. Adverse event rates were low and no serious adverse events were attributable to probiotics. Although no serious adverse events were observed among inpatient and outpatient children, including small studies conducted in the intensive care unit and in the neonatal unit, observational studies not included in this review have reported serious adverse events in severely debilitated or immuno-compromised children with underlying risk factors including central venous catheter use and disorders associated with bacterial/fungal translocation.
Collapse
Affiliation(s)
- Qin Guo
- West China Second University Hospital, West China Women's and Children's HospitalDepartment of PediatricsChengduChina
| | - Joshua Z Goldenberg
- National University of Natural MedicineHelfgott Research Institute2220 SW 1st AvePortlandORUSA97102
| | | | - Regina El Dib
- Institute of Science and Technology, UNESP ‐ Univ Estadual PaulistaDepartment of Biosciences and Oral DiagnosisSão José dos CamposSPBrazil
| | - Bradley C Johnston
- Dalhousie UniversityDepartment of Community Health and Epidemiology5790 University AvenueHalifaxNSCanadaB3H 1V7
| | | |
Collapse
|