1
|
Altieri C, Filippone A, Bevilacqua A, Corbo MR, Sinigaglia M. Lactobacilli and Bifidobacteria: A Parapostbiotic Approach to Study and Explain Their Mutual Bioactive Influence. Foods 2024; 13:2966. [PMID: 39335894 PMCID: PMC11431571 DOI: 10.3390/foods13182966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 09/12/2024] [Accepted: 09/16/2024] [Indexed: 09/30/2024] Open
Abstract
Three strains of Lactiplantibacillus plantarum and three bifidobacteria (Bifidobacterium animalis subsp. lactis, Bifidobacterium breve, and Bifidobacterium subtile) were used as target strains; in addition, for each microorganism, the cell-free supernatant (CFS) was produced and used as an ingredient of the growth medium. Namely CFSs from lactobacilli were used on bifidobacteria and CFSs from bifidobacteria were used on lactobacilli. The viable count was assessed, and the data were modelled through a reparametrized Gompertz equation cast both in the positive and negative form to evaluate the parameters t-7log, which is the time after which the viable count was 7 log CFU/mL, and the t-7log*, which is the time after which the viable count was below 7 log CFU/mL; the difference between the t-7log* and t-7log defines the stability time. Statistics through a multiparametric ANOVA (analysis of variance) provided evidence for the presence of a bifidogenic and/or bioactive factor produced by bifidobacteria and active on lactobacilli, and vice versa (bioactive factor of lactobacilli with a functional effect on bifidobacteria), although further studies are required to better explain the mechanisms beyond the positive effects. In addition, the influence on the target strains can be found during the growth phase (stimulation), as well as during senescence and death phase (protective effect), with a strong strain/species dependence on both CFS production and target strain.
Collapse
Affiliation(s)
- Clelia Altieri
- Department of Agriculture, Food, Natural Resources and Engineering, University of Foggia, 71122 Foggia, Italy; (C.A.); (A.F.); (M.R.C.); (M.S.)
| | - Alfonso Filippone
- Department of Agriculture, Food, Natural Resources and Engineering, University of Foggia, 71122 Foggia, Italy; (C.A.); (A.F.); (M.R.C.); (M.S.)
- Department of Psychology and Education, Pegaso University, 80143 Napoli, Italy
| | - Antonio Bevilacqua
- Department of Agriculture, Food, Natural Resources and Engineering, University of Foggia, 71122 Foggia, Italy; (C.A.); (A.F.); (M.R.C.); (M.S.)
| | - Maria Rosaria Corbo
- Department of Agriculture, Food, Natural Resources and Engineering, University of Foggia, 71122 Foggia, Italy; (C.A.); (A.F.); (M.R.C.); (M.S.)
| | - Milena Sinigaglia
- Department of Agriculture, Food, Natural Resources and Engineering, University of Foggia, 71122 Foggia, Italy; (C.A.); (A.F.); (M.R.C.); (M.S.)
| |
Collapse
|
2
|
Ukai K, Okamoto K, Ichinose A, Yoshida M, Higurashi Y, Yoneda R, Yamamoto S, Asahara T. Lacticaseibacillus paracasei Bacteremia Associated With Probiotic Use in a Child With Hypoganglionosis: Case Report and Literature Review. Pediatr Infect Dis J 2024:00006454-990000000-01004. [PMID: 39259865 DOI: 10.1097/inf.0000000000004548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/13/2024]
Abstract
Probiotics are generally considered safe and used to improve intestinal function. Here, we report a Lacticaseibacillus paracasei bacteremia case in an 8-month-old girl administered probiotics containing L. paracasei after jejunostomy for hypoganglionosis. The patient had persistent bacteremia with L. paracasei and was treated with antimicrobial therapy. The isolate from the blood culture was genetically indistinguishable to the strain contained in probiotics. Bacteremia risk should be considered before probiotic use in patients with underlying compromised immune systems or intestinal integrity.
Collapse
Affiliation(s)
- Kohei Ukai
- From the Department of Infectious Diseases, The University of Tokyo Hospital
| | - Koh Okamoto
- From the Department of Infectious Diseases, The University of Tokyo Hospital
- Department of Infectious Diseases, Tokyo Medical Dental University Hospital
| | - Akinori Ichinose
- Department of Pediatric Surgery, The University of Tokyo Hospital
| | - Mariko Yoshida
- Department of Pediatric Surgery, The University of Tokyo Hospital
| | - Yoshimi Higurashi
- Department of Infection Control and Prevention, The University of Tokyo Hospital, Bunkyo City, Tokyo, Japan
| | - Ryu Yoneda
- Department of Infection Control and Prevention, The University of Tokyo Hospital, Bunkyo City, Tokyo, Japan
| | - Shuta Yamamoto
- Yakult Central Institute, Yakult Honsha Co., Ltd, Kunitachi City, Tokyo, Japan
| | - Takashi Asahara
- Yakult Central Institute, Yakult Honsha Co., Ltd, Kunitachi City, Tokyo, Japan
| |
Collapse
|
3
|
Lin P, Zhuang J, Lai J, Cui J, Jiang D, Huang J. Efficacy of probiotics in the treatment of oral mucositis in head and neck cancer patients: A systematic review and meta-analysis. Microb Pathog 2024; 193:106785. [PMID: 38971507 DOI: 10.1016/j.micpath.2024.106785] [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: 03/05/2024] [Revised: 05/12/2024] [Accepted: 07/03/2024] [Indexed: 07/08/2024]
Abstract
OBJECTIVES To assess the effect of probiotics in oral mucositis induced by chemotherapy or radiotherapy on patients with head and neck cancer (HNC). METHODS The PubMed, Embase, Cochrane Library, Clinical trials were screened from January 2010 to April 2024. Randomized clinical trials (RCTs) comparing the efficacy of probiotics in treatment of oral mucositis in HNC were eligible. Outcomes of interest were incidence of oral mucositis and severe oral mucositis. The PROSPERO registration number was 42 022 384 685. The Cochrane risk-of-bias tool (RoB2) was used to assess methodological quality of studies and GRADE criteria (GRADEpro) was applied for rating the certainty of evidence. Meta-analysis was performed by using RevMan 5.4. RESULTS A total of eight RCTs comprising 691 patients with HNC were included in this meta-analysis. Probiotics administration significantly reduced the incidence of SOM (RR = 0.60, 95%CI: 0.46-0.78, P = 0.0002). However, it showed no distinct advantage in reducing the overall incidence of oral mucositis (RR = 0.88, 95%CI: 0.76-1.02, P = 0.08). Subgroup analysis found more benefit for reducing SOM in multi-bacterial treated group (RR = 0.35, 95%CI: 0.17-0.73, P = 0.005) than mono-bacterial treated group (RR = 0.69, 95%CI: 0.58-0.82, P < 0.0001). In Addition, probiotics could reduce the incidence of SOM in nasopharyngeal carcinoma (NPC) treated with intensity-modulated radiotherapy (RR = 0.43, 95%CI: 0.26-0.70, P = 0.0006). CONCLUSION Probiotics reduced the incidence of SOM caused by chemotherapy or radiotherapy for HNC. The multi-bacterial combination therapy was more efficacious than the mono-bacterial therapy. Moreover, probiotics also reduced the incidence of SOM in nasopharyngeal carcinoma. However, the advantage of probiotics had not been established in the overall incidence of OM.
Collapse
Affiliation(s)
- Peixin Lin
- Department of Head and Neck Oncology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, 524001, China
| | - Jiafeng Zhuang
- Department of Head and Neck Oncology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, 524001, China
| | - Jing Lai
- Department of Head and Neck Oncology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, 524001, China
| | - Ji Cui
- Department of Head and Neck Oncology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, 524001, China
| | - Danxian Jiang
- Department of Head and Neck Oncology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, 524001, China
| | - Jing Huang
- Department of Head and Neck Oncology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, 524001, China.
| |
Collapse
|
4
|
Wang Y, Zhang Z, Chen Q, Chen T. Simultaneous application of oral and intravaginal probiotics for Helicobacter pylori and its antibiotic-therapy-induced vaginal dysbacteriosis. NPJ Biofilms Microbiomes 2024; 10:49. [PMID: 38902244 PMCID: PMC11190290 DOI: 10.1038/s41522-024-00521-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Accepted: 06/07/2024] [Indexed: 06/22/2024] Open
Abstract
Helicobacter pylori is a prevalent bacterial pathogen globally, implicated in various gastrointestinal disorders. Current recommended antibiotic therapies for H. pylori infection have been proven to be therapeutically insufficient, with low eradication rates and high recurrence rates. Emerging evidence suggests that antibiotic therapy for H. pylori can lead to gastrointestinal and subsequent vaginal dysbiosis, posing challenges for conventional antibiotic approaches. Thus, this article proposes a novel probiotic therapy involving simultaneous oral and intra-vaginal probiotic administration alongside antibiotics for H. pylori treatment, aiming to enhance eradication rates and mitigate dysbiosis. We begin by providing an overview of gastrointestinal and vaginal microbiota and their interconnectedness through the vagina-gut axis. We then review the efficacy of current antibiotic regimens for H. pylori and discuss how antibiotic treatment impacts the vaginal microenvironment. To explore the feasibility of this approach, we evaluate the effectiveness of oral and intra-vaginal probiotics in restoring normal microbiota in the gastrointestinal and vaginal tracts, respectively. Additionally, we analyze the direct mechanisms by which oral and intra-vaginal probiotics act on their respective tracts and discuss potential cross-tract mechanisms. Considering the potential synergistic therapeutic effects of probiotics in both the gastrointestinal and vaginal tracts, dual-channel probiotic therapy holds promise as a more effective approach for H. pylori eradication and dysbiosis mitigation, presenting a novel concept in the collaborative treatment of gastrointestinal and genital disorders.
Collapse
Affiliation(s)
- Yufan Wang
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China
- Queen Mary School, Jiangxi Medical College, Nanchang University, Nanchang, 330031, China
- National Engineering Research Centre for Bioengineering Drugs and Technologies, Institute of Translational Medicine, Jiangxi Medical College, Nanchang University, Nanchang, 330031, China
| | - Zhenyu Zhang
- Department of Gastroenterology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China.
| | - Qi Chen
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China.
| | - Tingtao Chen
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China.
- National Engineering Research Centre for Bioengineering Drugs and Technologies, Institute of Translational Medicine, Jiangxi Medical College, Nanchang University, Nanchang, 330031, China.
- School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang, 330031, China.
| |
Collapse
|
5
|
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 2024:S2090-1232(24)00230-3. [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] [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
|
6
|
Wu WH, Lee CC, Chen YC, Chiang MC, Chiu CH. Invasive lactobacillus infection in pediatric patients in a tertiary center in Taiwan - 16 years' experience and literature review. Pediatr Neonatol 2024; 65:282-287. [PMID: 38007356 DOI: 10.1016/j.pedneo.2023.05.013] [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: 03/22/2023] [Revised: 05/06/2023] [Accepted: 05/19/2023] [Indexed: 11/27/2023] Open
Abstract
BACKGROUND Lactobacilli are common microorganisms in the human body. Some species were used as probiotics supplement for many purposes such as preventing necrotizing enterocolitis, or improving allergic diseases or diarrhea. Previously, Lactobacillus infection was thought of as contamination due to its low pathogenicity. However, there have been reports of invasive Lactobacillus infection in immunocompromised patients or patients with comorbidities. The purpose of this study was to analyze the clinical characteristics, antibiotic treatment and outcomes of pediatric patients with invasive Lactobacillus infection. METHODS We retrospectively reviewed pediatric patients diagnosed with invasive Lactobacillus infection between 2004 and 2020. Invasive Lactobacillus infection was diagnosed if sterile sites yielded Lactobacillus spp. Clinical manifestations, chronic diseases, potential predisposing factors, medical treatments, antimicrobial susceptibility tests and outcomes were recorded. RESULTS Fifteen pediatric patients were diagnosed with invasive Lactobacillus infection, accounting for 2.4% of total invasive Lactobacillus infections during the 16-year period. Eleven infections were bacteremia, two were intra-abdominal infections, and two were biliary tract infections. Fever was the most common symptom. Potential predisposing factors were immunocompromised status, central venous device, prolonged antibiotics use and receiving supplemented probiotics for at least one week. All patients survived with favorable outcomes. Most pathogens were identified as Lactobacillus spp, and two were Lactobacillus rhamnosus, which were related to supplemented probiotics. The antimicrobial susceptibility tests showed that Lactobacilli were all sensitive to ampicillin but resistant to glycopeptides. CONCLUSION Invasive Lactobacillus infections in pediatric patients were rare. Despite its low pathogenicity, Lactobacillus could cause invasive infection in those immunocompromised patients.
Collapse
Affiliation(s)
- Wei-Hung Wu
- Division of Neonatology, Department of Pediatrics, Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Chien-Chung Lee
- Division of Neonatology, Department of Pediatrics, Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Taoyuan, Taiwan; Graduate Institute of Clinical Medical Sciences, Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Yi-Ching Chen
- Molecular Infectious Disease Research Center, Department of Pediatrics, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan, Taiwan; Graduate Institute of Clinical Medical Sciences, Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Ming-Chou Chiang
- Division of Neonatology, Department of Pediatrics, Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Taoyuan, Taiwan; Division of Respiratory Therapy, Chang Gung Memorial Hospital, Taoyuan, Taiwan; Graduate Institute of Clinical Medical Sciences, Chang Gung University College of Medicine, Taoyuan, Taiwan.
| | - Cheng-Hsun Chiu
- Molecular Infectious Disease Research Center, Department of Pediatrics, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan, Taiwan; Graduate Institute of Clinical Medical Sciences, Chang Gung University College of Medicine, Taoyuan, Taiwan
| |
Collapse
|
7
|
Salahi A, Abd El-Ghany WA. Beyond probiotics, uses of their next-generation for poultry and humans: A review. J Anim Physiol Anim Nutr (Berl) 2024. [PMID: 38689488 DOI: 10.1111/jpn.13972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 04/03/2024] [Accepted: 04/13/2024] [Indexed: 05/02/2024]
Abstract
The production of healthy food is one of the basic requirements and challenges. Research efforts have been introduced in the human's food industry to reduce the microbial resistance and use safe and healthy alternatives with a high durability. However, the conducted work about these issues in the field of livestock animal production have been started since 2015. Inappropriate and extensive use of antibiotics has resulted in the increase of antimicrobial resistance, presence of drug residues in tissues, and destruction of the gut microbiome. Therefore, discovering and developing antibiotic substitutes were urgent demands. Probiotic compounds containing living micro-organisms are important antibiotic alternative that have been beneficially and extensively used in humans, animals, and poultry. However, some probiotics show some obstacles during production and applications. Accordingly, this review article proposes a comprehensive description of the next-generation of probiotics including postbiotics, proteobiotics, psychobiotics, immunobiotics and paraprobiotics and their effects on poultry production and human's therapy. These compounds proved great efficiency in terms of restoring gut health, improving performance and general health conditions, modulating the immune response and reducing the pathogenic micro-organisms. However, more future research work should be carried out regarding this issue.
Collapse
Affiliation(s)
- Ahmad Salahi
- Department of Animal Science, Faculty of Agriculture, Zanjan University, Zanjan, Iran
| | - Wafaa A Abd El-Ghany
- Department of Poultry Diseases, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| |
Collapse
|
8
|
Wei J, Li Z, Fan Y, Feng L, Zhong X, Li W, Guo T, Ning X, Li Z, Ou C. Lactobacillus rhamnosus GG aggravates vascular calcification in chronic kidney disease: A potential role for extracellular vesicles. Life Sci 2023; 331:122001. [PMID: 37625519 DOI: 10.1016/j.lfs.2023.122001] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 07/24/2023] [Accepted: 08/01/2023] [Indexed: 08/27/2023]
Abstract
AIMS Lactobacillus rhamnosus GG (LGG) is a probiotic with great promise in future clinical application, which can significantly promote bone formation. However, the effect of LGG on CKD-related vascular calcification is unclear. In this study, we aimed to investigate the effect of LGG on CKD-related vascular calcification. MATERIALS AND METHODS After 2 weeks of 5/6 nephrectomy, CKD rats received a special diet (4 % calcium and 1.8 % phosphate) combined with 1,25-dihydroxyvitamin D3 to induce vascular calcification. Meanwhile, CKD rats in the LGG group were gavaged orally with LGG (1 × 109 CFU bacteria/day). 16S RNA amplicon sequencing was performed to analyze the effect of LGG treatment on gut microbiota composition. Furthermore, differential ultracentrifugation was utilized to extract EVs. The effects of EVs on vascular calcification were evaluated in rat VSMCs, rat aortic rings, and CKD rat calcification models. In this study, vascular calcification was assessed by microcomputed tomography analysis, alizarin red staining, calcium content determination, and the expression of osteogenic transcription factors RUNX2 and BMP2. KEY FINDINGS LGG remarkably aggravated vascular calcification. LGG supplementation significantly altered gut microbiota composition in CKD rats, particularly increasing Lactobacillus. Interestingly, EVs presented a significant promoting effect on the development of calcification. Finally, mechanistic analysis proved that EVs aggravated vascular calcification through PI3K/AKT signaling. SIGNIFICANCE These results do not support the supplementation of LGG in CKD-associated vascular calcification patients. Our study presented a fresh perspective on LGG with potential risks and adverse effects. CKD patients should use specific probiotic strains cautiously.
Collapse
Affiliation(s)
- Jintao Wei
- The Tenth Affiliated Hospital of Southern Medical University (Dongguan People's Hospital), Southern Medical University, Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Dongguan 523018, PR China
| | - Zehua Li
- Department of Cardiology, Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, PR China
| | - Ying Fan
- The Tenth Affiliated Hospital of Southern Medical University (Dongguan People's Hospital), Southern Medical University, Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Dongguan 523018, PR China
| | - Liyun Feng
- Department of Cardiology, Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, PR China
| | - Xinglong Zhong
- Department of Cardiology, The Fourth Affiliated Hospital of Guangxi Medical University/Liuzhou Workers' Hospital, Liuzhou, PR China
| | - Weirun Li
- The Tenth Affiliated Hospital of Southern Medical University (Dongguan People's Hospital), Southern Medical University, Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Dongguan 523018, PR China
| | - Tingting Guo
- Department of Cardiology, Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, PR China
| | - Xiaodong Ning
- The Tenth Affiliated Hospital of Southern Medical University (Dongguan People's Hospital), Southern Medical University, Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Dongguan 523018, PR China
| | - Zhenhua Li
- The Tenth Affiliated Hospital of Southern Medical University (Dongguan People's Hospital), Southern Medical University, Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Dongguan 523018, PR China.
| | - Caiwen Ou
- The Tenth Affiliated Hospital of Southern Medical University (Dongguan People's Hospital), Southern Medical University, Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Dongguan 523018, PR China.
| |
Collapse
|
9
|
Zhang J, Ren L, Zhang L, Gong Y, Xu T, Wang X, Guo C, Zhai L, Yu X, Li Y, Zhu P, Chen R, Jing X, Jing G, Zhou S, Xu M, Wang C, Niu C, Ge Y, Ma B, Shang G, Cui Y, Yao S, Xu J. Single-cell rapid identification, in situ viability and vitality profiling, and genome-based source-tracking for probiotics products. IMETA 2023; 2:e117. [PMID: 38867931 PMCID: PMC10989769 DOI: 10.1002/imt2.117] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 04/22/2023] [Accepted: 05/07/2023] [Indexed: 06/14/2024]
Abstract
Rapid expansion of the probiotics industry demands fast, sensitive, comprehensive, and low-cost strategies for quality assessment. Here, we introduce a culture-free, one-cell-resolution, phenome-genome-combined strategy called Single-Cell Identification, Viability and Vitality tests, and Source-tracking (SCIVVS). For each cell directly extracted from the product, the fingerprint region of D2O-probed single-cell Raman spectrum (SCRS) enables species-level identification with 93% accuracy, based on a reference SCRS database from 21 statutory probiotic species, whereas the C-D band accurately quantifies viability, metabolic vitality plus their intercellular heterogeneity. For source-tracking, single-cell Raman-activated Cell Sorting and Sequencing can proceed, producing indexed, precisely one-cell-based genome assemblies that can reach ~99.40% genome-wide coverage. Finally, we validated an integrated SCIVVS workflow with automated SCRS acquisition where the whole process except sequencing takes just 5 h. As it is >20-fold faster, >10-time cheaper, vitality-revealing, heterogeneity-resolving, and automation-prone, SCIVVS is a new technological and data framework for quality assessment of live-cell products.
Collapse
Affiliation(s)
- Jia Zhang
- Single‐Cell Center, CAS Key Laboratory of Biofuels, Shandong Key Laboratory of Energy Genetics, Qingdao Institute of Bioenergy and Bioprocess Technology, Chinese Academy of SciencesQingdaoShandongChina
- Shandong Energy InstituteQingdaoShandongChina
- Qingdao New Energy Shandong LaboratoryQingdaoShandongChina
- University of Chinese Academy of SciencesBeijingChina
| | - Lihui Ren
- Single‐Cell Center, CAS Key Laboratory of Biofuels, Shandong Key Laboratory of Energy Genetics, Qingdao Institute of Bioenergy and Bioprocess Technology, Chinese Academy of SciencesQingdaoShandongChina
- Shandong Energy InstituteQingdaoShandongChina
- Qingdao New Energy Shandong LaboratoryQingdaoShandongChina
- College of Information Science & EngineeringOcean University of ChinaQingdaoShandongChina
| | - Lei Zhang
- Single‐Cell Center, CAS Key Laboratory of Biofuels, Shandong Key Laboratory of Energy Genetics, Qingdao Institute of Bioenergy and Bioprocess Technology, Chinese Academy of SciencesQingdaoShandongChina
- Shandong Energy InstituteQingdaoShandongChina
- Qingdao New Energy Shandong LaboratoryQingdaoShandongChina
- Qingdao Branch of China United Network Communications Co., Ltd.QingdaoShandongChina
| | - Yanhai Gong
- Single‐Cell Center, CAS Key Laboratory of Biofuels, Shandong Key Laboratory of Energy Genetics, Qingdao Institute of Bioenergy and Bioprocess Technology, Chinese Academy of SciencesQingdaoShandongChina
- Shandong Energy InstituteQingdaoShandongChina
- Qingdao New Energy Shandong LaboratoryQingdaoShandongChina
- University of Chinese Academy of SciencesBeijingChina
| | - Teng Xu
- Single‐Cell Center, CAS Key Laboratory of Biofuels, Shandong Key Laboratory of Energy Genetics, Qingdao Institute of Bioenergy and Bioprocess Technology, Chinese Academy of SciencesQingdaoShandongChina
- Shandong Energy InstituteQingdaoShandongChina
- Qingdao New Energy Shandong LaboratoryQingdaoShandongChina
- University of Chinese Academy of SciencesBeijingChina
| | - Xiaohang Wang
- Single‐Cell Center, CAS Key Laboratory of Biofuels, Shandong Key Laboratory of Energy Genetics, Qingdao Institute of Bioenergy and Bioprocess Technology, Chinese Academy of SciencesQingdaoShandongChina
- Shandong Energy InstituteQingdaoShandongChina
- Qingdao New Energy Shandong LaboratoryQingdaoShandongChina
- University of Chinese Academy of SciencesBeijingChina
| | - Cheng Guo
- Eastsea Pharma Co., Ltd.QingdaoShandongChina
| | - Lei Zhai
- China National Research Institute of Food and Fermentation Industries Co., Ltd., China Center of Industrial Culture CollectionBeijingChina
| | - Xuejian Yu
- China National Research Institute of Food and Fermentation Industries Co., Ltd., China Center of Industrial Culture CollectionBeijingChina
| | - Ying Li
- Qingdao Single‐Cell Biotech. Co., Ltd.QingdaoShandongChina
| | - Pengfei Zhu
- Single‐Cell Center, CAS Key Laboratory of Biofuels, Shandong Key Laboratory of Energy Genetics, Qingdao Institute of Bioenergy and Bioprocess Technology, Chinese Academy of SciencesQingdaoShandongChina
- Qingdao Single‐Cell Biotech. Co., Ltd.QingdaoShandongChina
| | - Rongze Chen
- Single‐Cell Center, CAS Key Laboratory of Biofuels, Shandong Key Laboratory of Energy Genetics, Qingdao Institute of Bioenergy and Bioprocess Technology, Chinese Academy of SciencesQingdaoShandongChina
- Shandong Energy InstituteQingdaoShandongChina
- Qingdao New Energy Shandong LaboratoryQingdaoShandongChina
- University of Chinese Academy of SciencesBeijingChina
| | - Xiaoyan Jing
- Single‐Cell Center, CAS Key Laboratory of Biofuels, Shandong Key Laboratory of Energy Genetics, Qingdao Institute of Bioenergy and Bioprocess Technology, Chinese Academy of SciencesQingdaoShandongChina
- Shandong Energy InstituteQingdaoShandongChina
- Qingdao New Energy Shandong LaboratoryQingdaoShandongChina
- University of Chinese Academy of SciencesBeijingChina
| | - Gongchao Jing
- Single‐Cell Center, CAS Key Laboratory of Biofuels, Shandong Key Laboratory of Energy Genetics, Qingdao Institute of Bioenergy and Bioprocess Technology, Chinese Academy of SciencesQingdaoShandongChina
- Shandong Energy InstituteQingdaoShandongChina
- Qingdao New Energy Shandong LaboratoryQingdaoShandongChina
- University of Chinese Academy of SciencesBeijingChina
| | - Shiqi Zhou
- Single‐Cell Center, CAS Key Laboratory of Biofuels, Shandong Key Laboratory of Energy Genetics, Qingdao Institute of Bioenergy and Bioprocess Technology, Chinese Academy of SciencesQingdaoShandongChina
- Shandong Energy InstituteQingdaoShandongChina
- Qingdao New Energy Shandong LaboratoryQingdaoShandongChina
| | - Mingyue Xu
- Single‐Cell Center, CAS Key Laboratory of Biofuels, Shandong Key Laboratory of Energy Genetics, Qingdao Institute of Bioenergy and Bioprocess Technology, Chinese Academy of SciencesQingdaoShandongChina
- Shandong Energy InstituteQingdaoShandongChina
- Qingdao New Energy Shandong LaboratoryQingdaoShandongChina
| | - Chen Wang
- Single‐Cell Center, CAS Key Laboratory of Biofuels, Shandong Key Laboratory of Energy Genetics, Qingdao Institute of Bioenergy and Bioprocess Technology, Chinese Academy of SciencesQingdaoShandongChina
- Shandong Energy InstituteQingdaoShandongChina
- Qingdao New Energy Shandong LaboratoryQingdaoShandongChina
| | | | - Yuanyuan Ge
- China National Research Institute of Food and Fermentation Industries Co., Ltd., China Center of Industrial Culture CollectionBeijingChina
| | - Bo Ma
- Single‐Cell Center, CAS Key Laboratory of Biofuels, Shandong Key Laboratory of Energy Genetics, Qingdao Institute of Bioenergy and Bioprocess Technology, Chinese Academy of SciencesQingdaoShandongChina
- Shandong Energy InstituteQingdaoShandongChina
- Qingdao New Energy Shandong LaboratoryQingdaoShandongChina
- University of Chinese Academy of SciencesBeijingChina
| | | | - Yunlong Cui
- Eastsea Pharma Co., Ltd.QingdaoShandongChina
| | - Su Yao
- China National Research Institute of Food and Fermentation Industries Co., Ltd., China Center of Industrial Culture CollectionBeijingChina
| | - Jian Xu
- Single‐Cell Center, CAS Key Laboratory of Biofuels, Shandong Key Laboratory of Energy Genetics, Qingdao Institute of Bioenergy and Bioprocess Technology, Chinese Academy of SciencesQingdaoShandongChina
- Shandong Energy InstituteQingdaoShandongChina
- Qingdao New Energy Shandong LaboratoryQingdaoShandongChina
- University of Chinese Academy of SciencesBeijingChina
| |
Collapse
|
10
|
Zhai J, Sun S, Cheng J, Wang J, Jin G, Xu X, Liu X, Zhao J, Chen C, Zhong W, Wang B. Lactobacillus acidophilus supernatant alleviates osteoporosis by upregulating colonic SERT expression. Future Microbiol 2023; 18:581-593. [PMID: 37424511 DOI: 10.2217/fmb-2022-0211] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/11/2023] Open
Abstract
Aims: To investigate the involvement of serotonin transporter (SERT) in colonic epithelial cells in the anti-osteoporosis role of Lactobacillus acidophilus (LA) supernatant (LAS). Methods: The abundance of fecal LA and bone mineral density (BMD) in patients with osteoporosis (OP) or severe osteoporosis were assessed. The protective role of LA in osteoporosis and the expression of SERT and relative signaling were evaluated. Results: Abundance of fecal LA was decreased in patients with severe OP and was positively correlated with BMD. Supplementing LAS to mice alleviated senile osteoporosis. In vitro, NOD2/RIP2/NF-κB signaling was inhibited by LAS due to increased SERT expression. Conclusion: LAS alleviates OP in mice by producing protective metabolites and upregulating SERT expression and represents a promising therapeutic agent.
Collapse
Affiliation(s)
- Jianhua Zhai
- Department of Emergency, Tianjin Medical University General Hospital, Tianjin, China
| | - Siyuan Sun
- Department of Gastroenterology & Hepatology, Tianjin Medical University General Hospital, Tianjin, China
| | - Jing Cheng
- Department of Orthointernal, Tianjin Hospital, Tianjin, China
| | - Jing Wang
- Department of Gastroenterology & Hepatology, Tianjin Medical University General Hospital, Tianjin, China
| | - Ge Jin
- Department of Gastroenterology & Hepatology, Tianjin Medical University General Hospital, Tianjin, China
| | - Xiuxiu Xu
- Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, China
| | - Xiaotong Liu
- Department of Gastroenterology & Hepatology, Tianjin Medical University General Hospital, Tianjin, China
| | - Jingwen Zhao
- Department of Gastroenterology & Hepatology, Tianjin Medical University General Hospital, Tianjin, China
| | - Chen Chen
- Department of Geriatric Medicine, Tianjin Medical University General Hospital, Tianjin, China
| | - Weilong Zhong
- Department of Gastroenterology & Hepatology, Tianjin Medical University General Hospital, Tianjin, China
| | - Bangmao Wang
- Department of Gastroenterology & Hepatology, Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
11
|
Hefter Y, Powell L, Tabulov CE, Sadler ED, Campos J, Hanisch B. An 11-Year Review of Lactobacillus Bacteremia at a Pediatric Tertiary Care Center. Hosp Pediatr 2023; 13:e140-e143. [PMID: 37203378 DOI: 10.1542/hpeds.2022-006892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
OBJECTIVES To inform clinical decisions on the use of probiotics in a pediatric inpatient setting, we sought to determine the number of cases of Lactobacillus bacteremia as well as associated patient characteristics in a tertiary-care pediatric hospital over an 11-year period. METHODS Cases of Lactobacillus bacteremia among admitted patients were identified through positive blood culture reports. The clinical chart for each case was reviewed for presenting symptoms and risk factors such as probiotic use, presence of a central venous catheter, immunocompromised state, impaired intestinal function, and age below 3 months. Concurrent total inpatient probiotic administration was assessed. RESULTS Over an 11-year period, 8 cases of Lactobacillus bacteremia were identified among 127 845 hospital admissions. All cases were associated with systemic signs of infection. Lactobacillus bacteremia patients most frequently had underlying impaired intestinal function and a central venous catheter. Three cases had a history of probiotic use. The peak number of annual cases did not coincide with the peak number of inpatients who received probiotics. CONCLUSIONS Lactobacillus bacteremia is uncommon and did not correlate with doses of probiotics-administered in the hospital. However, certain populations may be at higher risk and require extra consideration in clinical decision-making regarding use of probiotics.
Collapse
Affiliation(s)
| | | | | | - Eleanor D Sadler
- Department of Pharmacy, The Mount Sinai Hospital, New York, New York
| | - Joseph Campos
- Laboratory Medicine, Children's National Hospital, Washington, District of Columbia
| | - Benjamin Hanisch
- Divisions of Infectious Diseases
- Department of Pediatrics, The George Washington University School of Medicine and Health Sciences, Washington, District of Columbia
| |
Collapse
|
12
|
Kullar R, Goldstein EJC, Johnson S, McFarland LV. Lactobacillus Bacteremia and Probiotics: A Review. Microorganisms 2023; 11:microorganisms11040896. [PMID: 37110319 PMCID: PMC10145752 DOI: 10.3390/microorganisms11040896] [Citation(s) in RCA: 28] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 03/26/2023] [Accepted: 03/29/2023] [Indexed: 04/29/2023] Open
Abstract
Lactobacilli are widely found in nature, are commensal microbes in humans, and are commonly used as probiotics. Concerns about probiotic safety have arisen due to reports of bacteremia and other Lactobacillus-associated infections. We reviewed the literature for articles on the pathogenicity of Lactobacillus spp. bacteremia and reports of probiotics in these patients. Our aim is to review these articles and update the present knowledge on the epidemiology of Lactobacillus spp. bacteremia and determine the role of probiotics in Lactobacillus bacteremia. Lactobacillus bacteremia is infrequent but has a higher risk of mortality and risk factors, including severe underlying diseases, immune system suppression, admission to intensive care units, and use of central venous catheters. A variety of Lactobacillus species may cause bacteremia and may or may not be associated with probiotic exposure. To determine if oral probiotics are the source of these infections, the blood isolates and the oral probiotic strain(s) must be compared by sensitive identification methods. The prevalence of Lactobacillus bacteremia is infrequent but is more common in patients taking probiotics compared to those not taking probiotics. Three probiotics (Lacticaseibacillus rhamnosus GG, Lactiplantibacillus plantarum, and Lacticaseibacillus paracasei) were directly linked with blood isolates from bacteremia patients using molecular identification assays.
Collapse
Affiliation(s)
- Ravina Kullar
- Expert Stewardship Inc., Newport Beach, CA 92663, USA
| | | | | | | |
Collapse
|
13
|
Nobre L, Fernandes C, Florêncio K, Alencar N, Wong D, Lima-Júnior R. Could paraprobiotics be a safer alternative to probiotics for managing cancer chemotherapy-induced gastrointestinal toxicities? Braz J Med Biol Res 2023; 55:e12522. [PMID: 36651453 PMCID: PMC9843735 DOI: 10.1590/1414-431x2022e12522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Accepted: 11/24/2022] [Indexed: 01/18/2023] Open
Abstract
Clinical oncology has shown outstanding progress improving patient survival due to the incorporation of new drugs. However, treatment success may be reduced by the emergency of dose-limiting side effects, such as intestinal mucositis and diarrhea. Mucositis and diarrhea management is symptomatic, and there is no preventive therapy. Bacterial and fungal-based compounds have been suggested as an alternative for preventing the development of diarrhea in cancer patients. Using probiotics is safe and effective in immunocompetent individuals, but concerns remain during immunosuppressive conditions. Paraprobiotics, formulations composed of non-viable microorganisms, have been proposed to overcome such limitation. The present literature review discusses current evidence regarding the possible use of paraprobiotics as an alternative to probiotics to prevent gastrointestinal toxicity of cancer chemotherapy.
Collapse
Affiliation(s)
- L.M.S. Nobre
- Núcleo de Pesquisa e Desenvolvimento de Medicamentos, Departamento de Fisiologia e Farmacologia, Faculdade de Medicina, Universidade Federal do Ceará, Fortaleza, CE, Brasil
| | - C. Fernandes
- Núcleo de Pesquisa e Desenvolvimento de Medicamentos, Departamento de Fisiologia e Farmacologia, Faculdade de Medicina, Universidade Federal do Ceará, Fortaleza, CE, Brasil
| | - K.G.D. Florêncio
- Núcleo de Pesquisa e Desenvolvimento de Medicamentos, Departamento de Fisiologia e Farmacologia, Faculdade de Medicina, Universidade Federal do Ceará, Fortaleza, CE, Brasil
| | - N.M.N. Alencar
- Núcleo de Pesquisa e Desenvolvimento de Medicamentos, Departamento de Fisiologia e Farmacologia, Faculdade de Medicina, Universidade Federal do Ceará, Fortaleza, CE, Brasil
| | - D.V.T. Wong
- Núcleo de Pesquisa e Desenvolvimento de Medicamentos, Departamento de Fisiologia e Farmacologia, Faculdade de Medicina, Universidade Federal do Ceará, Fortaleza, CE, Brasil
| | - R.C.P. Lima-Júnior
- Núcleo de Pesquisa e Desenvolvimento de Medicamentos, Departamento de Fisiologia e Farmacologia, Faculdade de Medicina, Universidade Federal do Ceará, Fortaleza, CE, Brasil
| |
Collapse
|
14
|
Yan XX, Wu D. Intestinal microecology-based treatment for inflammatory bowel disease: Progress and prospects. World J Clin Cases 2023; 11:47-56. [PMID: 36687179 PMCID: PMC9846986 DOI: 10.12998/wjcc.v11.i1.47] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 11/11/2022] [Accepted: 12/15/2022] [Indexed: 01/04/2023] Open
Abstract
Inflammatory bowel disease (IBD) is a chronic, recurrent, and debilitating disorder, and includes Crohn’s disease and ulcerative colitis. The pathogenesis of IBD is closely associated with intestinal dysbiosis, but has not yet been fully clarified. Genetic and environmental factors can influence IBD patients’ gut microbiota and metabolism, disrupt intestinal barriers, and trigger abnormal immune responses. Studies have reported the alteration of gut microbiota and metabolites in IBD, providing the basis for potential therapeutic options. Intestinal microbiota-based treatments such as pre/probiotics, metabolite supplementation, and fecal microbiota transplantation have been extensively studied, but their clinical efficacy remains controversial. Repairing the intestinal barrier and promoting mucosal healing have also been proposed. We here review the current clinical trials on intestinal microecology and discuss the prospect of research and practice in this field.
Collapse
Affiliation(s)
- Xia-Xiao Yan
- Department of Gastroenterology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100730, China
| | - Dong Wu
- Department of Gastroenterology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100730, China
| |
Collapse
|
15
|
van Lier YF, Vos J, Blom B, Hazenberg MD. Allogeneic hematopoietic cell transplantation, the microbiome, and graft-versus-host disease. Gut Microbes 2023; 15:2178805. [PMID: 36794370 PMCID: PMC9980553 DOI: 10.1080/19490976.2023.2178805] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/17/2023] Open
Abstract
Many patients with hematological malignancies, such as acute myeloid leukemia, receive an allogeneic hematopoietic cell transplantation (HCT) to cure their underlying condition. Allogeneic HCT recipients are exposed to various elements during the pre-, peri- and post-transplant period that can disrupt intestinal microbiota, including chemo- and radiotherapy, antibiotics, and dietary changes. The dysbiotic post-HCT microbiome is characterized by low fecal microbial diversity, loss of anaerobic commensals, and intestinal domination, particularly by Enterococcus species, and is associated with poor transplant outcomes. Graft-versus-host disease (GvHD) is a frequent complication of allogeneic HCT caused by immunologic disparity between donor and host cells and results in tissue damage and inflammation. Microbiota injury is particularly pronounced in allogeneic HCT recipients who go on to develop GvHD. At present, manipulation of the microbiome for example, via dietary interventions, antibiotic stewardship, prebiotics, probiotics, or fecal microbiota transplantation, is widely being explored to prevent or treat gastrointestinal GvHD. This review discusses current insights into the role of the microbiome in GvHD pathogenesis and summarizes interventions to prevent and treat microbiota injury.
Collapse
Affiliation(s)
- Yannouck F. van Lier
- Department of Hematology, Amsterdam UMC location AMC, Amsterdam, The Netherlands,Department of Experimental Immunology, Amsterdam Institute for Infection & Immunity Institute, Cancer Center Amsterdam, Amsterdam UMC location AMC, Amsterdam, The Netherlands
| | - Jaël Vos
- Department of Hematology, Amsterdam UMC location AMC, Amsterdam, The Netherlands,Department of Experimental Immunology, Amsterdam Institute for Infection & Immunity Institute, Cancer Center Amsterdam, Amsterdam UMC location AMC, Amsterdam, The Netherlands
| | - Bianca Blom
- Department of Experimental Immunology, Amsterdam Institute for Infection & Immunity Institute, Cancer Center Amsterdam, Amsterdam UMC location AMC, Amsterdam, The Netherlands
| | - Mette D. Hazenberg
- Department of Hematology, Amsterdam UMC location AMC, Amsterdam, The Netherlands,Department of Experimental Immunology, Amsterdam Institute for Infection & Immunity Institute, Cancer Center Amsterdam, Amsterdam UMC location AMC, Amsterdam, The Netherlands,Department of Hematopoiesis, Sanquin Research, Amsterdam, The Netherlands,CONTACT Mette D. Hazenberg Department of Hematology, Amsterdam UMC, Meibergdreef 9, Amsterdam1105 AZ, The Netherlands
| |
Collapse
|
16
|
Merenstein D, Pot B, Leyer G, Ouwehand AC, Preidis GA, Elkins CA, Hill C, Lewis ZT, Shane AL, Zmora N, Petrova MI, Collado MC, Morelli L, Montoya GA, Szajewska H, Tancredi DJ, Sanders ME. Emerging issues in probiotic safety: 2023 perspectives. Gut Microbes 2023; 15:2185034. [PMID: 36919522 PMCID: PMC10026873 DOI: 10.1080/19490976.2023.2185034] [Citation(s) in RCA: 71] [Impact Index Per Article: 71.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Accepted: 02/22/2023] [Indexed: 03/16/2023] Open
Abstract
Probiotics are used for both generally healthy consumers and in clinical settings. However, theoretical and proven adverse events from probiotic consumption exist. New probiotic strains and products, as well as expanding use of probiotics into vulnerable populations, warrants concise, and actionable recommendations on how to work toward their safe and effective use. The International Scientific Association for Probiotics and Prebiotics convened a meeting to discuss and produce evidence-based recommendations on potential acute and long-term risks, risks to vulnerable populations, the importance for probiotic product quality to match the needs of vulnerable populations, and the need for adverse event reporting related to probiotic use. The importance of whole genome sequencing, which enables determination of virulence, toxin, and antibiotic resistance genes, as well as clear assignment of species and strain identity, is emphasized. We present recommendations to guide the scientific and medical community on judging probiotic safety.
Collapse
Affiliation(s)
- Daniel Merenstein
- Department of Family Medicine, Georgetown University Medical Center, Washington, DCUSA
| | - Bruno Pot
- Yakult Europe BV, Almere, Netherlands
| | | | - Arthur C. Ouwehand
- Global Health & Nutrition Sciences, International Flavors & Fragrances, Kantvik, Finland
| | - Geoffrey A. Preidis
- Division of Gastroenterology, Hepatology & Nutrition, Department of Pediatrics, Baylor College of Medicine and Texas Children’s Hospital, Houston, TX, USA
| | - Christopher A. Elkins
- Clinical and Environmental Microbiology Branch, Division of Healthcare Quality Promotion, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Colin Hill
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | | | - Andi L. Shane
- Division of Infectious Diseases, Department of Pediatrics, Emory University School of Medicine and Children’s Healthcare of Atlanta, Emory Children’s Center, Atlanta, Georgia
| | - Niv Zmora
- Scientific consultant, Elinav Lab, Immunology Department, Weizmann Institute of Science, Department of Gastroenterology and Liver Diseases, Tel Aviv, Israel
| | | | - Maria Carmen Collado
- Institute of Agrochemistry and Food Technology-National Research Council (IATA-CSIC), Valencia, Spain
| | - Lorenzo Morelli
- Department of Food Science and Technology, Università Cattolica del Sacro Cuore, Piacenza, Italy
| | - Gina A. Montoya
- Department of Chemical Risk Assessment, Nestlé S.A., Lausanne, Switzerland
| | - Hania Szajewska
- Department of Paediatrics, The Medical University of Warsaw, Warsaw, Poland
| | - Daniel J. Tancredi
- Department of Pediatrics, UC Davis School of Medicine, Sacramento, CA, USA
| | - Mary Ellen Sanders
- International Scientific Association for Probiotics and Prebiotics, Centennial, CO, USA
| |
Collapse
|
17
|
Karbowiak M, Gałek M, Szydłowska A, Zielińska D. The Influence of the Degree of Thermal Inactivation of Probiotic Lactic Acid Bacteria and Their Postbiotics on Aggregation and Adhesion Inhibition of Selected Pathogens. Pathogens 2022; 11:1260. [PMID: 36365011 PMCID: PMC9692860 DOI: 10.3390/pathogens11111260] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 10/25/2022] [Accepted: 10/25/2022] [Indexed: 07/25/2023] Open
Abstract
The study aimed to evaluate the effect of thermal inactivation of potentially probiotic lactic acid bacteria (LAB) strains isolated from food on their ability to compete with pathogenic microorganisms. Five strains of LAB, previously isolated from food and characterized, one commercial reference strain of Lactiplantibacillus plantarum 299v, and two indicator strains of Staphylococcus aureus 25923 and Listeriamonocytogenes 15313 were used in the study. The experiment consisted in applying a stress factor (high temperature: 80 °C, at a different time: 5, 15, and 30 min) to the tested LAB cells to investigate the in vitro properties such as hydrophobicity abilities (against p-xylene and n-hexadecane), auto-aggregation, co-aggregation with pathogens, and inhibition of pathogens adhesion to the porcine gastric mucin. The bacterial strains showed various hydrophobicity to p-xylene (36-73%) and n-hexadecane (11-25%). The affinity for solvents expanded with increasing thermal inactivation time. All LAB isolates were able to auto-aggregate (ranging from 17 to 49%). Bacterial strains subjected to 5 and 15 min of thermal inactivation had the highest auto-aggregation ability in comparison to viable and heat-killed cells for 30 min. The LAB strains co-aggregated with pathogens to different degrees; among them, the highest scores of co-aggregation were observed for L. monocytogenes, reaching 27% (with 15 min of heat-killed LAB cells). All LAB strains reduced the adherence of pathogenic bacteria in the competition test, moreover, heat-killed cells (especially 15 min inactivated) were more efficient than viable cells. The properties of selected LAB strains as moderately heat-stressed forms analyzed in the study increased the prevention of colonization and elimination of pathogenic bacteria in the in vitro model of gastrointestinal tract. The thermal inactivation process may therefore preserve and modifies some characteristics of bacterial cells.
Collapse
|
18
|
Muratore E, Leardini D, Baccelli F, Venturelli F, Prete A, Masetti R. Nutritional modulation of the gut microbiome in allogeneic hematopoietic stem cell transplantation recipients. Front Nutr 2022; 9:993668. [PMID: 36337625 PMCID: PMC9632163 DOI: 10.3389/fnut.2022.993668] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Accepted: 10/04/2022] [Indexed: 11/23/2022] Open
Abstract
Allogeneic hematopoietic stem cell transplantation (allo-HSCT) represents a potentially curative strategy for many oncological and non-oncological diseases, but it is associated with marked morbidity and mortality. The disruption of gut microbiota (GM) eubiosis has been linked to major allo-HSCT complications, including infections and acute graft vs. host disease (aGvHD), and correlates with mortality. This increasing knowledge on the role of the GM in the allo-HSCT procedure has led to fascinating ideas for modulating the intestinal ecosystem in order to improve clinical outcomes. Nutritional strategies, either by changing the route of nutritional supplementation or by administering specific molecules, are increasingly being considered as cost- and risk-effective methods of modulating the GM. Nutritional support has also emerged in the past several years as a key feature in supportive care for allo-HSCT recipients, and deterioration of nutritional status is associated with decreased overall survival and higher complication rates during treatment. Herein we provide a complete overview focused on nutritional modulation of the GM in allo-HSCT recipients. We address how pre transplant diet could affect GM composition and its ability to withstand the upsetting events occurring during transplantation. We also provide a complete overview on the influence of the route of nutritional administration on the intestinal ecosystem, with a particular focus on the comparison between enteral and parenteral nutrition (PN). Moreover, as mounting evidence are showing how specific components of post-transplant diet, such as lactose, could drastically shape the GM, we will also summarize the role of prebiotic supplementation in the modulation of the intestinal flora and in allo-HSCT outcomes.
Collapse
Affiliation(s)
- Edoardo Muratore
- Pediatric Oncology and Hematology “Lalla Seràgnoli,” IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Davide Leardini
- Pediatric Oncology and Hematology “Lalla Seràgnoli,” IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Francesco Baccelli
- Pediatric Oncology and Hematology “Lalla Seràgnoli,” IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
- *Correspondence: Francesco Baccelli,
| | - Francesco Venturelli
- Pediatric Oncology and Hematology “Lalla Seràgnoli,” IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy
| | - Arcangelo Prete
- Pediatric Oncology and Hematology “Lalla Seràgnoli,” IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Riccardo Masetti
- Pediatric Oncology and Hematology “Lalla Seràgnoli,” IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
| |
Collapse
|
19
|
Bacteraemia Caused by Probiotic Strains of Lacticaseibacillus rhamnosus—Case Studies Highlighting the Need for Careful Thought before Using Microbes for Health Benefits. Pathogens 2022; 11:pathogens11090977. [PMID: 36145409 PMCID: PMC9504050 DOI: 10.3390/pathogens11090977] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 08/16/2022] [Accepted: 08/24/2022] [Indexed: 11/16/2022] Open
Abstract
Lactic acid bacteria belonging to Lactobacillus spp. and Lacticaseibacillus spp. are a natural part of fermented milk and other food products, probiotic supplements and human microbiota. They mainly belong to mucosal microflora, especially oral, vaginal and intestinal. Lacticaseibacillus spp. strains included in probiotics are generally characterised as safe microorganisms, and the species are concerned bacteria with very low pathogenic potential. However, infections caused by Lactobacillus spp. and Lacticaseibacillus spp., including bacteraemia and endocarditis, occur occasionally. The aim of the study was to present two cases of bacteraemia due to Lacticaseibacillus rhamnosus associated with the use of a probiotic product. It afflicted patients in intensive care units. The investigation was preliminarily based on clinical and microbiological recognition of the cases. The initial observation was laboratory confirmed with the application of pulsed-field gel electrophoresis (PFGE) results. Identical PFGE patterns were obtained for the evaluated strains and the strains derived from a commercially available probiotic that was administered to those patients. The increasing number of studies describing opportunistic infections due to probiotic strains of Lacticaseibacillus spp. should result in verifying the safety of probiotic formulations used in immunocompromised patients and forming detailed guidelines for the use of probiotics among patients from several risk groups.
Collapse
|
20
|
Safety Assessment of Lactiplantibacillus plantarum TWK10 Based on Whole-Genome Sequencing, Phenotypic, and Oral Toxicity Analysis. Microorganisms 2022; 10:microorganisms10040784. [PMID: 35456834 PMCID: PMC9031848 DOI: 10.3390/microorganisms10040784] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 04/03/2022] [Accepted: 04/05/2022] [Indexed: 12/04/2022] Open
Abstract
Lactiplantibacillus plantarum TWK10 (TWK10), isolated from Taiwanese pickled cabbage, has been demonstrated to exert beneficial probiotic effects in both mice and humans. Here, we comprehensively assessed the safety of TWK10 using both in vivo and in vitro approaches, including whole-genome sequence analysis, an assessment of hemolytic activity, and performing an antimicrobial susceptibility test, the Ames bacterial reverse mutation assay, the chromosomal aberration test, a rodent peripheral blood micronucleus test, and the 28-day subacute oral toxicity assay. The results showed that there was no significant increase in the incidence of reverse mutations or chromosomal aberrations following exposure to TWK10. Moreover, no significant changes were detected either in the number of reticulocytes or the incidence of micronuclei in ICR mice, and no subacute toxicity was recorded in SD rats at the oral TWK10 dosage of 2000 mg/kg body weight/day repeated for 28 days. Additionally, TWK10 exhibited no hemolytic activity and was susceptible to all the antibiotics tested, except kanamycin. However, no antimicrobial resistance genes, virulence factors, or genes involved in biogenic amine synthesis were found in the genome of TWK10. Our findings demonstrated that TWK10 has high potential of being safe for human consumption as a probiotic.
Collapse
|
21
|
Wang Y, Dong J, Wang J, Chi W, Zhou W, Tian Q, Hong Y, Zhou X, Ye H, Tian X, Hu R, Wong A. Assessing the drug resistance profiles of oral probiotic lozenges. J Oral Microbiol 2022; 14:2019992. [PMID: 35024089 PMCID: PMC8745366 DOI: 10.1080/20002297.2021.2019992] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Background Probiotic lozenges have been developed to harvest the benefits of probiotics for oral health, but their long-term consumption may encourage the transfer of resistance genes from probiotics to commensals, and eventually to disease-causing bacteria. Aim To screen commercial probiotic lozenges for resistance to antibiotics, characterize the resistance determinants, and examine their transferability in vitro. Results Probiotics of all lozenges were resistant to glycopeptide, sulfonamide, and penicillin antibiotics, while some were resistant to aminoglycosides and cephalosporins. High minimum inhibitory concentrations (MICs) were detected for streptomycin (>128 µg/mL) and chloramphenicol (> 512 µg/mL) for all probiotics but only one was resistant to piperacillin (MIC = 32 µg/mL). PCR analysis detected erythromycin (erm(T), ermB or mefA) and fluoroquinolone (parC or gyr(A)) resistance genes in some lozenges although there were no resistant phenotypes. The dfrD, cat-TC, vatE, aadE, vanX, and aph(3")-III or ant(2")-I genes conferring resistance to trimethoprim, chloramphenicol, quinupristin/dalfopristin, vancomycin, and streptomycin, respectively, were detected in resistant probiotics. The rifampicin resistance gene rpoB was also present. We found no conjugal transfer of streptomycin resistance genes in our co-incubation experiments. Conclusion Our study represents the first antibiotic resistance profiling of probiotics from oral lozenges, thus highlighting the health risk especially in the prevailing threat of drug resistance globally.
Collapse
Affiliation(s)
- Yi Wang
- Department of Orthodontics, School and Hospital of Stomatology, Wenzhou Medical University, University Town, Wenzhou, Zhejiang Province, China
| | - Jingya Dong
- Department of Orthodontics, School and Hospital of Stomatology, Wenzhou Medical University, University Town, Wenzhou, Zhejiang Province, China
| | - Junyi Wang
- Department of Biology, College of Science and Technology, Wenzhou-Kean University, Wenzhou, Zhejiang Province, China
| | - Wei Chi
- Department of Biology, College of Science and Technology, Wenzhou-Kean University, Wenzhou, Zhejiang Province, China
| | - Wei Zhou
- Department of Biology, College of Science and Technology, Wenzhou-Kean University, Wenzhou, Zhejiang Province, China
| | - Qiwen Tian
- Department of Biology, College of Science and Technology, Wenzhou-Kean University, Wenzhou, Zhejiang Province, China
| | - Yue Hong
- Department of Orthodontics, School and Hospital of Stomatology, Wenzhou Medical University, University Town, Wenzhou, Zhejiang Province, China
| | - Xuan Zhou
- Department of Biology, College of Science and Technology, Wenzhou-Kean University, Wenzhou, Zhejiang Province, China
| | - Hailv Ye
- Department of Biology, College of Science and Technology, Wenzhou-Kean University, Wenzhou, Zhejiang Province, China
| | - Xuechen Tian
- Zhejiang Bioinformatics International Science and Technology Cooperation Center, Wenzhou, Zhejiang Province, China.,Wenzhou Municipal Key Lab for Applied Biomedical and Biopharmaceutical Informatics, Wenzhou, Zhejiang Province, China
| | - Rongdang Hu
- Department of Orthodontics, School and Hospital of Stomatology, Wenzhou Medical University, University Town, Wenzhou, Zhejiang Province, China
| | - Aloysius Wong
- Department of Biology, College of Science and Technology, Wenzhou-Kean University, Wenzhou, Zhejiang Province, China.,Zhejiang Bioinformatics International Science and Technology Cooperation Center, Wenzhou, Zhejiang Province, China.,Wenzhou Municipal Key Lab for Applied Biomedical and Biopharmaceutical Informatics, Wenzhou, Zhejiang Province, China
| |
Collapse
|
22
|
Martyniak A, Medyńska-Przęczek A, Wędrychowicz A, Skoczeń S, Tomasik PJ. Prebiotics, Probiotics, Synbiotics, Paraprobiotics and Postbiotic Compounds in IBD. Biomolecules 2021; 11:biom11121903. [PMID: 34944546 PMCID: PMC8699341 DOI: 10.3390/biom11121903] [Citation(s) in RCA: 72] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Revised: 12/09/2021] [Accepted: 12/14/2021] [Indexed: 12/12/2022] Open
Abstract
The increasing incidence of inflammatory bowel diseases (IBD) and the increasing severity of the course of these diseases create the need for developing new methods of therapy. The gut microbiome is extensively studied as a factor influencing the development and course of IBD. The composition of intestinal microbiota can be relatively easily modified by diet (i.e., prebiotics, mainly dietary fibers) and bacterial supplementation using beneficial bacteria strains called probiotics. Additionally, the effects of the improved microbiome could be enhanced or gained by using paraprobiotics (non-viable, inactivated bacteria or their components) and/or postbiotics (products of bacterial metabolism or equal synthetic products that beneficially modulate immunological response and inflammation). This study summarizes the recent works on prebiotics, probiotics, synbiotics (products merging pre- and probiotics), paraprobiotics and postbiotics in IBD.
Collapse
Affiliation(s)
- Adrian Martyniak
- Department of Clinical Biochemistry, Pediatric Institute, Faculty of Medicine, Jagiellonian University Medical College, 30-663 Krakow, Poland;
| | - Aleksandra Medyńska-Przęczek
- Department of Paediatrics, Gastroenterology and Nutrition, Pediatric Institute, Faculty of Medicine, Jagiellonian University Medical College, 30-663 Krakow, Poland; (A.M.-P.); (A.W.)
| | - Andrzej Wędrychowicz
- Department of Paediatrics, Gastroenterology and Nutrition, Pediatric Institute, Faculty of Medicine, Jagiellonian University Medical College, 30-663 Krakow, Poland; (A.M.-P.); (A.W.)
| | - Szymon Skoczeń
- Department of Pediatric Oncology and Hematology, Faculty of Medicine, Jagiellonian University Medical College, 30-663 Krakow, Poland;
| | - Przemysław J. Tomasik
- Department of Clinical Biochemistry, Pediatric Institute, Faculty of Medicine, Jagiellonian University Medical College, 30-663 Krakow, Poland;
- Correspondence:
| |
Collapse
|
23
|
Guo X, Yang X, Li Q, Shen X, Zhong H, Yang Y. The Microbiota in Systemic Lupus Erythematosus: An Update on the Potential Function of Probiotics. Front Pharmacol 2021; 12:759095. [PMID: 34887760 PMCID: PMC8650621 DOI: 10.3389/fphar.2021.759095] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Accepted: 11/01/2021] [Indexed: 12/18/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is a kind of chronic diffuse connective tissue illness characterized by multisystem and multiorgan involvement, repeated recurrence and remission, and the presence of a large pool of autoantibodies in the body. Although the exact cause of SLE is not thoroughly revealed, accumulating evidence has manifested that intake of probiotics alters the composition of the gut microbiome, regulating the immunomodulatory and inflammatory response, which may be linked to the disease pathogenesis. Particularly, documented experiments demonstrated that SLE patients have remarkable changes in gut microbiota compared to healthy controls, indicating that the alteration of microbiota may be implicated in different phases of SLE. In this review, the alteration of microbiota in the development of SLE is summarized, and the mechanism of intestinal microbiota on the progression of immune and inflammatory responses in SLE is also discussed. Due to limited reports on the effects of probiotics supplementation in SLE patients, we emphasize advancements made in the last few years on the function and mechanisms of probiotics in the development of SLE animal models. Besides, we follow through literature to survey whether probiotics supplements can be an adjuvant therapy for comprehensive treatment of SLE. Research has indicated that intake of probiotics alters the composition of the gut microbiome, contributing to prevent the progression of SLE. Adjustment of the gut microbiome through probiotics supplementation seems to alleviate SLE symptoms and their cardiovascular and renal complications in animal models, marking this treatment as a potentially novel approach.
Collapse
Affiliation(s)
- Xirui Guo
- Department of Pharmacy, Chengdu Second People's Hospital, Chengdu, China
| | - Xuerong Yang
- Department of Pharmacy, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Qi Li
- Department of Pharmacy, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Xiaoyan Shen
- Department of Pharmacy, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Huiyun Zhong
- Department of Pharmacy, Sichuan Vocational College of Health and Rehabilitation, Zigong, China.,Department of Pharmacy, The First People's Hospital of Zigong, Zigong, China
| | - Yong Yang
- Department of Pharmacy, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
24
|
Masetti R, Muratore E, Leardini D, Zama D, Turroni S, Brigidi P, Esposito S, Pession A. Gut microbiome in pediatric acute leukemia: from predisposition to cure. Blood Adv 2021; 5:4619-4629. [PMID: 34610115 PMCID: PMC8759140 DOI: 10.1182/bloodadvances.2021005129] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 08/25/2021] [Indexed: 01/02/2023] Open
Abstract
The gut microbiome (GM) has emerged as a key factor in the genesis and progression of many diseases. The intestinal bacterial composition also influences treatment-related side effects and even the efficacy of oncological therapies. Acute leukemia (AL) is the most common cancer among children and the most frequent cause of cancer-related death during childhood. Outcomes have improved considerably over the past 4 decades, with the current long-term survival for acute lymphoblastic leukemia being ∼90%. However, several acute toxicities and long-term sequelae are associated with the multimodal therapy protocols applied in these patients. Specific GM configurations could contribute to the multistep developmental hypothesis for leukemogenesis. Moreover, GM alterations occur during the AL therapeutic course and are associated with treatment-related complications, especially during hematopoietic stem cell transplantation. The GM perturbation could last even after the removal of microbiome-modifying factors, like antibiotics, chemotherapeutic drugs, or alloimmune reactions, contributing to several health-related issues in AL survivors. The purpose of this article is to provide a comprehensive review of the chronological changes of GM in children with AL, from predisposition to cure. The underpinning biological processes and the potential interventions to modulate the GM toward a potentially health-promoting configuration are also highlighted.
Collapse
Affiliation(s)
- Riccardo Masetti
- Pediatric Oncology and Hematology Unit “Lalla Seràgnoli,” Pediatric Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Edoardo Muratore
- Pediatric Oncology and Hematology Unit “Lalla Seràgnoli,” Pediatric Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Davide Leardini
- Pediatric Oncology and Hematology Unit “Lalla Seràgnoli,” Pediatric Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Daniele Zama
- Pediatric Oncology and Hematology Unit “Lalla Seràgnoli,” Pediatric Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Silvia Turroni
- Unit of Microbial Ecology of Health, Department of Pharmacy and Biotechnology, and
| | - Patrizia Brigidi
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy; and
| | - Susanna Esposito
- Pediatric Clinic, Pietro Barilla Children's Hospital, University of Parma, Parma, Italy
| | - Andrea Pession
- Pediatric Oncology and Hematology Unit “Lalla Seràgnoli,” Pediatric Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| |
Collapse
|
25
|
Kumar H, Schütz F, Bhardwaj K, Sharma R, Nepovimova E, Dhanjal DS, Verma R, Kumar D, Kuča K, Cruz-Martins N. Recent advances in the concept of paraprobiotics: Nutraceutical/functional properties for promoting children health. Crit Rev Food Sci Nutr 2021:1-16. [PMID: 34748444 DOI: 10.1080/10408398.2021.1996327] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Probiotics consumption has been associated with various health promoting benefits, including disease prevention and even treatment by modulating gut microbiota. Contrary to this, probiotics may also overstimulate the immune system, trigger systemic infections, harmful metabolic activities, and promote gene transfer. In children, the fragile immune system and impaired intestinal barrier may boost the occurrence of adverse effects following probiotics' consumption. To overcome these health challenges, the key focus has been shifted toward non-viable probiotics, also called paraprobiotics. Cell wall polysaccharides, peptidoglycans, surface proteins and teichoic acid present on cell's surface are involved in the interaction of paraprobiotics with the host, ultimately providing health benefits. Among other benefits, paraprobiotics possess the ability to regulate innate and adaptive immunity, exert anti-adhesion, anti-biofilm, anti-hypertensive, anti-inflammatory, antioxidant, anti-proliferative, and antagonistic effects against pathogens, while also enhance clinical impact and general safety when administered in children in comparison to probiotics. Clinical evidence have underlined the paraprobiotics impact in children and young infants against atopic dermatitis, respiratory and gastrointestinal infections, in addition to be useful for immunocompromised individuals. Therefore, this review focuses on probiotics-related issues in children's health and also discusses the Lactobacillus and Bifidobacterium spp. qualities for qualifying as paraprobiotics and their role in promoting the children's health.
Collapse
Affiliation(s)
- Harsh Kumar
- School of Bioengineering & Food Technology, Shoolini University of Biotechnology and Management Sciences, Solan, India
| | - Francine Schütz
- Department of Medicine/Biomedicine, Faculty of Medicine, University of Porto, Porto, Portugal
| | - Kanchan Bhardwaj
- School of Biological and Environmental Sciences, Shoolini University of Biotechnology and Management Sciences, Solan, India
| | - Ruchi Sharma
- School of Bioengineering & Food Technology, Shoolini University of Biotechnology and Management Sciences, Solan, India
| | - Eugenie Nepovimova
- Department of Chemistry, Faculty of Science, University of Hradec Kralove, Hradec Kralove, Czech Republic
| | - Daljeet Singh Dhanjal
- School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, Punjab, India
| | - Rachna Verma
- School of Biological and Environmental Sciences, Shoolini University of Biotechnology and Management Sciences, Solan, India
| | - Dinesh Kumar
- School of Bioengineering & Food Technology, Shoolini University of Biotechnology and Management Sciences, Solan, India
| | - Kamil Kuča
- Department of Chemistry, Faculty of Science, University of Hradec Kralove, Hradec Kralove, Czech Republic
| | - Natália Cruz-Martins
- Department of Medicine/Biomedicine, Faculty of Medicine, University of Porto, Porto, Portugal.,Institute for Research and Innovation in Health (i3S), University of Porto, Porto, Portugal.,Institute of Research and Advanced Training in Health Sciences and Technologies (CESPU), Rua Central de Gandra, Gandra, PRD, Portugal
| |
Collapse
|
26
|
D’Agostin M, Squillaci D, Lazzerini M, Barbi E, Wijers L, Da Lozzo P. Invasive Infections Associated with the Use of Probiotics in Children: A Systematic Review. CHILDREN-BASEL 2021; 8:children8100924. [PMID: 34682189 PMCID: PMC8534463 DOI: 10.3390/children8100924] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 10/09/2021] [Accepted: 10/13/2021] [Indexed: 11/16/2022]
Abstract
Although the effectiveness of probiotics has only been proven in specific conditions, their use in children is massively widespread because of their perception as harmless products. Recent evidence raises concerns about probiotics' safety, especially but not only in the paediatric population due to severe opportunistic infections after their use. This review aimed at summarising available case reports on invasive infections related to probiotics' use in children. For this purpose, we assessed three electronic databases to identify papers describing paediatric patients with documented probiotic-derived invasive infections, with no language restrictions. A total of 49 case reports from 1995 to June 2021 were identified. The infections were caused by Lactobacillus spp. (35%), Saccharomyces spp. (29%), Bifidobacterium spp. (31%), Bacillus clausii (4%), and Escherichia coli (2%). Most (80%) patients were younger than 2 years old and sepsis was the most observed condition (69.4%). All the patients except one had at least one condition facilitating the development of invasive infection, with prematurity (55%) and intravenous catheter use (51%) being the most frequent. Three (6%) children died. Given the large use of probiotics, further studies aiming at evaluating the real incidence of probiotic-associated systemic infections are warranted.
Collapse
Affiliation(s)
- Martina D’Agostin
- Department of Medicine, Surgery and Health Sciences, University of Trieste, 34127 Trieste, Italy; (M.D.); (E.B.); (P.D.L.)
| | - Domenica Squillaci
- Department of Medicine, Surgery and Health Sciences, University of Trieste, 34127 Trieste, Italy; (M.D.); (E.B.); (P.D.L.)
- Correspondence:
| | - Marzia Lazzerini
- Institute of Maternal and Child Health—IRCCS Burlo Garofolo, Via dell’Istria 65/1, 34137 Trieste, Italy;
| | - Egidio Barbi
- Department of Medicine, Surgery and Health Sciences, University of Trieste, 34127 Trieste, Italy; (M.D.); (E.B.); (P.D.L.)
- Institute of Maternal and Child Health—IRCCS Burlo Garofolo, Via dell’Istria 65/1, 34137 Trieste, Italy;
| | - Lotte Wijers
- Faculty of Health, Medicine and Life Sciences, Maastricht University, 6229 ER Maastricht, The Netherlands;
| | - Prisca Da Lozzo
- Department of Medicine, Surgery and Health Sciences, University of Trieste, 34127 Trieste, Italy; (M.D.); (E.B.); (P.D.L.)
| |
Collapse
|
27
|
Fan L, Lee JH. Enteral feeding and the microbiome in critically ill children: a narrative review. Transl Pediatr 2021; 10:2778-2791. [PMID: 34765500 PMCID: PMC8578772 DOI: 10.21037/tp-20-349] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Accepted: 04/09/2021] [Indexed: 01/10/2023] Open
Abstract
OBJECTIVE This narrative review summarizes our current knowledge on the interplay between enteral nutrition (EN) and gut microbiota in critically ill children, using examples from two commonly encountered diagnoses in the pediatric intensive care unit (PICU): severe sepsis and acute respiratory distress syndrome (ARDS). This review will also highlight potential areas of therapeutic interventions that should be explored in future studies. BACKGROUND Critically ill children display extreme dysbiosis in their gut microbiome. Factors within the PICU that are often associated with dysbiosis include the use of broad-spectrum antibiotics, proton-pump inhibitors (PPIs), intravenous morphine, and fasting. Dysbiosis can potentially lead to adverse clinical outcomes (e.g., nosocomial infection, and prolonged hospitalization). EN may modulate dysbiosis. The gut microbiota is involved in the breaking down of macronutrients, mainly carbohydrates and proteins. Fermentation of undigestible carbohydrate (e.g., inulin and oligosaccharides), and amino acids by large intestine microbiota produces short chain fatty acids (SCFAs). SCFAs serve as the main fuel source for enterocytes and help to maintain healthy gut lining. Changes to selected components of macronutrients can result in alterations in gut microbiome and have potentially beneficial effects in patients in the PICU. METHODS A comprehensive search of the MEDLINE, Cochrane Library and Google Scholar databases was conducted using appropriate MESH terms and keywords. In this narrative review, we provide a summary of current knowledge on effect of EN on gut microbiota in pediatric studies, but also describes animal- and lab-based, as well as adult studies where relevant. CONCLUSIONS The gut microbiome can be altered by dietary modifications and common PICU practices and treatment. Although there are strong associations in restoring eubiosis and improvement in clinical outcomes, proving causality remains challenging. Further microbiome research is needed to provide mechanistic insights into the impact of the ever changing gut microbiome. In the future, new microbiota targeted therapies could potentially be the treatment of challenging PICU conditions and restore homeostasis in these children.
Collapse
Affiliation(s)
- Lijia Fan
- Division of Paediatric Critical Care, Department of Paediatrics, Khoo Teck Puat-National University Children's Medical Institute, National University Hospital, Singapore, Singapore
| | - Jan Hau Lee
- Children's Intensive Care Unit, KK Women's and Children's Hospital, Singapore, Singapore.,Duke-NUS Medical School, Singapore, Singapore
| |
Collapse
|
28
|
Su L, Su Y, An Z, Zhang P, Yue Q, Zhao C, Sun X, Zhang S, Liu X, Li K, Zhao L. Fermentation products of Danshen relieved dextran sulfate sodium-induced experimental ulcerative colitis in mice. Sci Rep 2021; 11:16210. [PMID: 34376708 PMCID: PMC8355158 DOI: 10.1038/s41598-021-94594-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 06/29/2021] [Indexed: 12/19/2022] Open
Abstract
With the increased incidence and recognition, ulcerative colitis (UC) has become a global public health problem in the world. Although many immunosuppressant and biological drugs have been used for UC treatment, the cure rate is still very low. It is necessary to find some safe and long-term used medicine for UC cure. Recently, the Chinese traditional herb Danshen has been investigated in the treatment of UC. However, it is a limitation of Danshen that many of the active components in Danshen are not easily absorbed by the human body. Probiotics could convert macromolecules into smaller molecules to facilitate absorption. Thus, Lactobacillus rhamnosus (F-B4-1) and Bacillus subtillis Natto (F-A7-1) were screened to ferment Danshen in this study. The fermented Danshen products were gavaged in the dextran sulfate sodium (DSS)-induced UC model mice. Danshen had better results to attenuate symptoms of DSS-induced UC after fermented with F-B4-1 and F-A7-1. Loss of body weight and disease activity index (DAI) were reduced. The abnormally short colon lengths and colonic damage were recovered. And fermented Danshen had the better inhibitory effect than Danshen itself on pro-inflammatory cytokine expression during DSS-induced UC. The results indicated that compared with Danshen, fermented Danshen relieved DSS-induced UC in mice more effectively. Danshen fermented by probiotics might be an effective treatment to UC in clinic stage in the future.
Collapse
Affiliation(s)
- Le Su
- State Key Laboratory of Biobased Material and Green Papermaking, School of Bioengineering, Qilu University of Technology, Shandong Academy of Sciences, Jinan, 250353, China
| | - Yue Su
- State Key Laboratory of Biobased Material and Green Papermaking, School of Bioengineering, Qilu University of Technology, Shandong Academy of Sciences, Jinan, 250353, China
| | - Zaiyong An
- State Key Laboratory of Biobased Material and Green Papermaking, School of Bioengineering, Qilu University of Technology, Shandong Academy of Sciences, Jinan, 250353, China
| | - Ping Zhang
- State Key Laboratory of Biobased Material and Green Papermaking, School of Bioengineering, Qilu University of Technology, Shandong Academy of Sciences, Jinan, 250353, China
| | - Qiulin Yue
- State Key Laboratory of Biobased Material and Green Papermaking, School of Bioengineering, Qilu University of Technology, Shandong Academy of Sciences, Jinan, 250353, China
| | - Chen Zhao
- Shandong Provincial Key Laboratory of Food and Fermentation Engineering, Shandong Food Ferment Industry Research and Design Institute, Qilu University of Technology, Shandong Academy of Sciences, Jinan, 250013, China
| | - Xin Sun
- State Key Laboratory of Biobased Material and Green Papermaking, School of Bioengineering, Qilu University of Technology, Shandong Academy of Sciences, Jinan, 250353, China
| | - Song Zhang
- State Key Laboratory of Biobased Material and Green Papermaking, School of Bioengineering, Qilu University of Technology, Shandong Academy of Sciences, Jinan, 250353, China
| | - Xinli Liu
- State Key Laboratory of Biobased Material and Green Papermaking, School of Bioengineering, Qilu University of Technology, Shandong Academy of Sciences, Jinan, 250353, China
| | - Kunlun Li
- Jinan Hangchen Biotechnology Co., Ltd., Jinan, 250353, China
| | - Lin Zhao
- State Key Laboratory of Biobased Material and Green Papermaking, School of Bioengineering, Qilu University of Technology, Shandong Academy of Sciences, Jinan, 250353, China.
| |
Collapse
|
29
|
Van Lier YF, Van den Brink MRM, Hazenberg MD, Markey KA. The post-hematopoietic cell transplantation microbiome: relationships with transplant outcome and potential therapeutic targets. Haematologica 2021; 106:2042-2053. [PMID: 33882637 PMCID: PMC8327718 DOI: 10.3324/haematol.2020.270835] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Indexed: 01/16/2023] Open
Abstract
Microbiota injury occurs in many patients undergoing allogeneic hematopoietic cell transplantation, likely as a consequence of conditioning regimens involving chemo- and radiotherapy, the widespread use of both prophylactic and therapeutic antibiotics, and profound dietary changes during the peri-transplant period. Peri-transplant dysbiosis is characterized by a decrease in bacterial diversity, loss of commensal bacteria and single-taxon domination (e.g., with Enterococcal strains). Clinically, deviation of the post-transplant microbiota from a normal, high-diversity, healthy state has been associated with increased risk of bacteremia, development of graft-versus-host disease and decreases in overall survival. A number of recent clinical trials have attempted to target the microbiota in allogeneic hematopoietic cell transplantation patients via dietary interventions, selection of therapeutic antibiotics, administration of pre- or pro-biotics, or by performing fecal microbiota transplantation. These strategies have yielded promising results but the mechanisms by which these interventions influence transplant-related complications remain largely unknown. In this review we summarize the current approaches to targeting the microbiota, discuss potential underlying mechanisms and highlight the key outstanding areas that require further investigation in order to advance microbiota- targeting therapies.
Collapse
Affiliation(s)
- Yannouck F Van Lier
- Department of Hematology, Amsterdam UMC, Amsterdam, the Netherlands; Department of Experimental Immunology, Amsterdam Institute for Infection and Immunity (AII), Cancer Center Amsterdam, Amsterdam UMC, Amsterdam
| | - Marcel R M Van den Brink
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Medicine, Weill Cornell Medical College, New York, NY
| | - Mette D Hazenberg
- Department of Hematology, Amsterdam UMC, Amsterdam, the Netherlands; Department of Experimental Immunology, Amsterdam Institute for Infection and Immunity (AII), Cancer Center Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands; Department of Hematopoiesis, Sanquin Research, Amsterdam
| | - Kate A Markey
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Medicine, Weill Cornell Medical College, New York, NY.
| |
Collapse
|
30
|
Probiotic Bacterial Application in Pediatric Critical Illness as Coadjuvants of Therapy. ACTA ACUST UNITED AC 2021; 57:medicina57080781. [PMID: 34440989 PMCID: PMC8399162 DOI: 10.3390/medicina57080781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2021] [Revised: 07/24/2021] [Accepted: 07/26/2021] [Indexed: 11/17/2022]
Abstract
The use of probiotics in critically ill adult and children patients has been growing exponentially over the last 20 years. Numerous factors in pediatriac intensive care unit (PICU) patients may contribute to intestinal dysbiosis, which subsequently promotes the pathobiota's growth. Currently, lactobacillus and bifidobacterium species are mainly used to prevent the development of systemic diseases due to the subverted microbiome, followed by streptococcus, enterococcus, propionibacterium, bacillus and Escherichia coli, Lactobacillus rhamnosus GG, and Lactobacillus reuteri DSM 17938. The aim of this article is to review the scientific literature for further confirmation of the importance of the usage of probiotics in intensive care unit (ICU) patients, especially in the pediatric population. A progressive increase in nosocomial infections, especially nosocomial bloodstream infections, has been observed over the last 30 years. The World Health Organization (WHO) reported that the incidence of nosocomial infections in PICUs was still high and ranged between 5% and 10%. Petrof et al. was one of the first to demonstrate the efficacy of probiotics for preventing systemic diseases in ICU patients. Recently, however, the use of probiotics with different lactobacillus spp. has been shown to cause a decrease of pro-inflammatory cytokines and an increase in anti-inflammatory cytokines. In addition, in some studies, the use of probiotics, in particular the mix of Lactobacillus and Bifidobacterium reduces the incidence of ventilator-associated pneumonia (VAP) in PICU patients requiring mechanical ventilation. In abdominal infections, there is no doubt at all about the usefulness of using Lactobacillus spp probiotics, which help to treat ICU-acquired diarrhoea episodes as well as in positive blood culture for candida spp. Despite the importance of using probiotics being supported by various studies, their use is not yet part of the standard protocols to which all doctors must adhere. In the meantime, while waiting for protocols to be drawn up as soon as possible for use in PICUs, routine use could certainly stimulate the intestine's immune defences. Though it is still too early to say, they could be considered the drugs of the future.
Collapse
|
31
|
Probiotics for the Management of Sepsis: Advances in Animal Models and Intensive Care Unit Environments. MICROBIOLOGY RESEARCH 2021. [DOI: 10.3390/microbiolres12030039] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Sepsis frequently leads to multiple organ failure and is a major cause of morbidity and mortality in critically ill patients. Although intensive care protocols and antibiotic therapy have improved sepsis treatment, specific management is lacking with respect to efficient protection from tissue damage and long-term outcomes. Probiotics are live microbes that modulate the immune system and inflammation and colonize the gut. In this narrative review, we have traced the evolution of the administration of probiotics in an animal model of sepsis and treatment alternatives in the intensive care unit setting. First, probiotics are categorized by species before describing their modulation of the microbiota, repair of tissue-specific damage, immune response, and molecular pathways to prevent complications. The impact on therapy for infant and adult patients is also addressed. Finally, we have emphasized the challenges and gaps in current studies as well as future perspectives for further investigation. The present review can open up avenues for new strategies that employ promising probiotic strains for the treatment of sepsis and discusses their ability to prevent disease-associated long-term complications.
Collapse
|
32
|
Ailioaie LM, Litscher G. Probiotics, Photobiomodulation, and Disease Management: Controversies and Challenges. Int J Mol Sci 2021; 22:ijms22094942. [PMID: 34066560 PMCID: PMC8124384 DOI: 10.3390/ijms22094942] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 04/26/2021] [Accepted: 04/29/2021] [Indexed: 02/06/2023] Open
Abstract
In recent decades, researchers around the world have been studying intensively how micro-organisms that are present inside living organisms could affect the main processes of life, namely health and pathological conditions of mind or body. They discovered a relationship between the whole microbial colonization and the initiation and development of different medical disorders. Besides already known probiotics, novel products such as postbiotics and paraprobiotics have been developed in recent years to create new non-viable micro-organisms or bacterial-free extracts, which can provide benefits to the host with additional bioactivity to probiotics, but without the risk of side effects. The best alternatives in the use of probiotics and postbiotics to maintain the health of the intestinal microbiota and to prevent the attachment of pathogens to children and adults are highlighted and discussed as controversies and challenges. Updated knowledge of the molecular and cellular mechanisms involved in the balance between microbiota and immune system for the introspection on the gut-lung-brain axis could reveal the latest benefits and perspectives of applied photobiomics for health. Multiple interconditioning between photobiomodulation (PBM), probiotics, and the human microbiota, their effects on the human body, and their implications for the management of viral infectious diseases is essential. Coupled complex PBM and probiotic interventions can control the microbiome, improve the activity of the immune system, and save the lives of people with immune imbalances. There is an urgent need to seek and develop innovative treatments to successfully interact with the microbiota and the human immune system in the coronavirus crisis. In the near future, photobiomics and metabolomics should be applied innovatively in the SARS-CoV-2 crisis (to study and design new therapies for COVID-19 immediately), to discover how bacteria can help us through adequate energy biostimulation to combat this pandemic, so that we can find the key to the hidden code of communication between RNA viruses, bacteria, and our body.
Collapse
Affiliation(s)
- Laura Marinela Ailioaie
- Department of Medical Physics, Alexandru Ioan Cuza University, 11 Carol I Boulevard, 700506 Iasi, Romania;
- Ultramedical & Laser Clinic, 83 Arcu Street, 700135 Iasi, Romania
| | - Gerhard Litscher
- Research Unit of Biomedical Engineering in Anesthesia and Intensive Care Medicine, Research Unit for Complementary and Integrative Laser Medicine, and Traditional Chinese Medicine (TCM) Research Center Graz, Medical University of Graz, Auenbruggerplatz 39, 8036 Graz, Austria
- Correspondence: ; Tel.: +43-316-385-83907
| |
Collapse
|
33
|
Hong T, Wang R, Wang X, Yang S, Wang W, Gao Q, Zhang X. Interplay Between the Intestinal Microbiota and Acute Graft-Versus-Host Disease: Experimental Evidence and Clinical Significance. Front Immunol 2021; 12:644982. [PMID: 33815399 PMCID: PMC8010685 DOI: 10.3389/fimmu.2021.644982] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Accepted: 02/26/2021] [Indexed: 12/23/2022] Open
Abstract
Allogeneic hematopoietic stem cell transplantation (allo-HSCT) is a potentially curative therapy for many hematological disorders and autoimmune diseases, but acute graft-versus-host disease (aGVHD) has remained a major obstacle that limits allo-HSCT and exhibits a daunting mortality rate. The gastrointestinal system is among the most common sites affected by aGVHD. Experimental advances in the field of intestinal microbiota research enhanced our understanding - not only of the quantity and diversity of intestinal microbiota - but also their association with homeostasis of the immune system and disease pathogenesis, including that of aGVHD. Meanwhile, ever-growing clinical evidence suggest that the intestinal microbiota is dysregulated in patients who develop aGVHD and that the imbalance may affect clinical outcomes, indicating a potential predictive role for microbiota dysregulation in aGVHD severity and prognosis. The current animal and human studies investigating the intestinal microbiota in aGVHD and the understanding of the influence and management of the microbiota in the clinic are reviewed herein. Taken together, monitoring and remodeling the intestinal microecology following allo-HSCT may provide us with promising avenues for diagnosing, preventing or treating aGVHD in the clinic.
Collapse
Affiliation(s)
- Tao Hong
- Medical Center of Hematology, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Rui Wang
- Medical Center of Hematology, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Xiaoqi Wang
- Medical Center of Hematology, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Shijie Yang
- Medical Center of Hematology, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Weihao Wang
- Medical Center of Hematology, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Qiangguo Gao
- Department of Cell Biology, College of Basic Medicine, Third Military Medical University (Army Medical University), Chongqing, China
| | - Xi Zhang
- Medical Center of Hematology, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, China.,State Key Laboratory of Trauma, Burns and Combined Injury, Third Military Medical University (Army Medical University), Chongqing, China
| |
Collapse
|
34
|
Abbasi A, Rad AH, Ghasempour Z, Sabahi S, Kafil HS, Hasannezhad P, Rahbar Saadat Y, Shahbazi N. The biological activities of postbiotics in gastrointestinal disorders. Crit Rev Food Sci Nutr 2021; 62:5983-6004. [PMID: 33715539 DOI: 10.1080/10408398.2021.1895061] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
According to outcomes from clinical studies, an intricate relationship occurs between the beneficial microbiota, gut homeostasis, and the host's health status. Numerous studies have confirmed the health-promoting effects of probiotics, particularly in gastrointestinal diseases. On the other hand, the safety issues regarding the consumption of some probiotics are still a matter of debate, thus to overcome the problems related to the application of live probiotic cells in terms of clinical, technological, and economic aspects, microbial-derived biomolecules (postbiotics) were introducing as a potential alternative agent. Presently scientific literature confirms that the postbiotic components can be used as promising tools for both prevention and treatment strategies in gastrointestinal disorders with less undesirable side-effects, particularly in infants and children. Future head-to-head trials are required to distinguish appropriate strains of parent cells, optimal dosages of postbiotics, and assessment of the cost-effectiveness of postbiotics compared to alternative drugs. This review provides an overview of the concept and safety issues regarding postbiotics, with emphasis on their biological role in the treatment of some important gastrointestinal disorders.
Collapse
Affiliation(s)
- Amin Abbasi
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Food Science and Technology, Faculty of Nutrition & Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Aziz Homayouni Rad
- Department of Food Science and Technology, Faculty of Nutrition & Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Zahra Ghasempour
- Department of Food Science and Technology, Faculty of Nutrition & Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sahar Sabahi
- Department of Nutritional Sciences, School of Paramedical Sciences, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Hossein Samadi Kafil
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Paniz Hasannezhad
- Department of Medical Engineering Science, University College of Rouzbahan, Sari, Iran
| | - Yalda Rahbar Saadat
- Nutrition Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nayyer Shahbazi
- Faculty of Agriculture Engineering, Department of Food Science, Shahrood University of Technology, Shahrood, Iran
| |
Collapse
|
35
|
Abstract
A number of diseases and conditions have been associated with prolonged or persistent exposure to non-physiological levels of reactive oxygen species (ROS). Similarly, ROS underproduction due to loss-of-function mutations in superoxide or hydrogen peroxide (H2O2)-generating enzymes is a risk factor or causative for certain diseases. However, ROS are required for basic cell functions; in particular the diffusible second messenger H2O2 that serves as signaling molecule in redox processes. This activity sets H2O2 apart from highly reactive oxygen radicals and influences the approach to drug discovery, clinical utility, and therapeutic intervention. Here we review the chemical and biological fundamentals of ROS with emphasis on H2O2 as a signaling conduit and initiator of redox relays and propose an integrated view of physiological versus non-physiological reactive species. Therapeutic interventions that target persistently altered ROS levels should include both selective inhibition of a specific source of primary ROS and careful consideration of a targeted pro-oxidant approach, an avenue that is still underdeveloped. Both strategies require attention to redox dynamics in complex cellular systems, integration of the overall spatiotemporal cellular environment, and target validation to yield effective and safe therapeutics. The only professional primary ROS producers are NADPH oxidases (NOX1-5, DUOX1-2). Many other enzymes, e.g., xanthine oxidase (XO), monoamine oxidases (MAO), lysyl oxidases (LO), lipoxygenase (LOX), and cyclooxygenase (COX), produce superoxide and H2O2 secondary to their primary metabolic function. Superoxide is too reactive to disseminate, but H2O2 is diffusible, only limited by adjacent PRDXs or GPXs, and can be apically secreted and imported into cells through aquaporin (AQP) channels. H2O2 redox signaling includes oxidation of the active site thiol in protein tyrosine phosphatases, which will inhibit their activity and thereby increase tyrosine phosphorylation on target proteins. Essential functions include the oxidative burst by NOX2 as antimicrobial innate immune response; gastrointestinal NOX1 and DUOX2 generating low H2O2 concentrations sufficient to trigger antivirulence mechanisms; and thyroidal DUOX2 essential for providing H2O2 reduced by TPO to oxidize iodide to an iodinating form which is then attached to tyrosyls in TG. Loss-of-function (LoF) variants in TPO or DUOX2 cause congenital hypothyroidism and LoF variants in the NOX2 complex chronic granulomatous disease.
Collapse
|
36
|
Shin M, Ban OH, Jung YH, Yang J, Kim Y. Genomic characterization and probiotic potential of Lactobacillus casei IDCC 3451 isolated from infant faeces. Lett Appl Microbiol 2021; 72:578-588. [PMID: 33421164 DOI: 10.1111/lam.13449] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Revised: 12/11/2020] [Accepted: 12/28/2020] [Indexed: 01/02/2023]
Abstract
Probiotics play an important role in health benefits on the host. However, they also possess potentials for infectivity or in situ toxin production; thus, requiring a comprehensive assessment of their safety. In this study, we report genomic characteristics of a newly isolated Lactobacillus casei IDCC 3451 from infant faeces. Phenotypic assays based on enzyme activities and carbohydrate fermentation profiles represented metabolic features of the strain. Safety evaluation for antimicrobial resistance, biogenic amines production and cytotoxicity to a murine mouse model suggested its safe use as a probiotic strain. Our findings on the genetic background of L. casei IDCC 3451 and its potential features provide a promising functional and safe probiotic strain for the human consumption.
Collapse
Affiliation(s)
- M Shin
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Science, Seoul National University, Seoul, Republic of Korea
| | - O-H Ban
- Ildong Bioscience, Gyeonggi-do, Republic of Korea
| | - Y H Jung
- School of Food Science and Biotechnology, Kyungpook National University, Daegu, Republic of Korea
| | - J Yang
- Ildong Bioscience, Gyeonggi-do, Republic of Korea
| | - Y Kim
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Science, Seoul National University, Seoul, Republic of Korea
| |
Collapse
|
37
|
Bossù G, Di Sario R, Argentiero A, Esposito S. Antimicrobial Prophylaxis and Modifications of the Gut Microbiota in Children with Cancer. Antibiotics (Basel) 2021; 10:antibiotics10020152. [PMID: 33546312 PMCID: PMC7913491 DOI: 10.3390/antibiotics10020152] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 01/27/2021] [Accepted: 01/29/2021] [Indexed: 12/12/2022] Open
Abstract
In children with cancer, chemotherapy can produce cytotoxic effects, resulting in immunosuppression and an augmented risk of febrile neutropenia and bloodstream infections. This has led to widespread use of antibiotic prophylaxis which, combined with intensive chemotherapy treatment, could have a long-term effect on the gastrointestinal microbiome. In this review, we aimed to analyze the current literature about the widespread use of antibiotic prophylaxis in children experiencing infectious complications induced by chemotherapy and its effects on the gut microbiome. Our review of the literature shows that antimicrobial prophylaxis in children with cancer is still a trending topic and, at the moment, there are not enough data to define universal guidelines. Children with cancer experience long and painful medical treatments and side effects, which are associated with great economic and social burdens, important psychological consequences, and dysbiosis induced by antibiotics and also by chemotherapy. Considering the importance of a healthy gut microbiota, studies are needed to understand the impact of dysbiosis in response to therapy in these children and to define how to modulate the microbiome to favor a positive therapeutic outcome.
Collapse
|
38
|
De Musis C, Granata L, Dallio M, Miranda A, Gravina AG, Romano M. Inflammatory Bowel Diseases: The Role of Gut Microbiota. Curr Pharm Des 2021; 26:2951-2961. [PMID: 32310042 DOI: 10.2174/1381612826666200420144128] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Accepted: 04/13/2020] [Indexed: 12/12/2022]
Abstract
Inflammatory bowel diseases (IBD) are chronic multifactorial diseases characterized by partially unclear pathogenic mechanisms including changes in intestinal microbiota. Despite the microbiota, alteration is well established in IBD patients, as reported by 16RNA sequencing analysis, an important goal is to define if it is just a consequence of the disease progression or a trigger factor of the disease itself. To date, gut microbiota composition and gut microbiota-related metabolites seem to affect the host healthy state both by modulating metabolic pathways or acting on the expression of different genes through epigenetic effects. Because of this, it has been suggested that intestinal microbiota might represent a promising therapeutic target for IBD patients. The aim of this review is to summarize both the most recent acquisitions in the field of gut microbiota and its involvement in intestinal inflammation together with the available strategies for the modulation of microbiota, such as prebiotics and/or probiotics administration or fecal microbiota transplantation.
Collapse
Affiliation(s)
- Cristiana De Musis
- Departments of Precision Medicine and Polyspecialistic Internal Medicine, University of Campania ''Luigi Vanvitelli'' and University Hospital, Naples, Italy
| | - Lucia Granata
- Departments of Precision Medicine and Polyspecialistic Internal Medicine, University of Campania ''Luigi Vanvitelli'' and University Hospital, Naples, Italy
| | - Marcello Dallio
- Departments of Precision Medicine and Polyspecialistic Internal Medicine, University of Campania ''Luigi Vanvitelli'' and University Hospital, Naples, Italy
| | - Agnese Miranda
- Departments of Precision Medicine and Polyspecialistic Internal Medicine, University of Campania ''Luigi Vanvitelli'' and University Hospital, Naples, Italy
| | - Antonietta G Gravina
- Departments of Precision Medicine and Polyspecialistic Internal Medicine, University of Campania ''Luigi Vanvitelli'' and University Hospital, Naples, Italy
| | - Marco Romano
- Departments of Precision Medicine and Polyspecialistic Internal Medicine, University of Campania ''Luigi Vanvitelli'' and University Hospital, Naples, Italy
| |
Collapse
|
39
|
Angurana S, Mehta A. Probiotics in critically ill children: An updated review. JOURNAL OF PEDIATRIC CRITICAL CARE 2021. [DOI: 10.4103/jpcc.jpcc_73_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
|
40
|
Abstract
The pandemic of Coronavirus disease 2019 (COVID-19) is rapidly progressing, causing significant morbidity and mortality. Various antiviral drugs, anti-inflammatory drugs and immunomodulators have been tried without substantial clinical benefits. The severe and critical cases of COVID-19 disease are characterised by gut microbiome dysbiosis, immune dysregulation, hyper-inflammation and hypercytokinaemia (cytokine storm). Therefore, the strategies which target these pathophysiological processes may be beneficial. Probiotics are one such strategy that exerts beneficial effects by manipulation of the gut microbiota, suppression of opportunistic pathogens in the gut, decreasing translocation of opportunistic organisms, activation of mucosal immunity and modulation of the innate and adaptive immune response. Probiotics are the potential candidates to be tested in moderate and severe cases of COVID-19 due to several beneficial effects, including easy availability, easy to administer, safe and economical to use.
Collapse
|
41
|
Pace F, Macchini F, Massimo Castagna V. Safety of probiotics in humans: A dark side revealed? Dig Liver Dis 2020; 52:981-985. [PMID: 32563720 DOI: 10.1016/j.dld.2020.04.029] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 04/21/2020] [Accepted: 04/23/2020] [Indexed: 12/11/2022]
Affiliation(s)
- Fabio Pace
- Gastroenterology Unit, Bolognini Hospital, ASST Bergamo Est, Seriate, BG, Italy
| | - Federico Macchini
- Gastroenterology Unit, Bolognini Hospital, ASST Bergamo Est, Seriate, BG, Italy.
| | | |
Collapse
|
42
|
A critical review of antibiotic resistance in probiotic bacteria. Food Res Int 2020; 136:109571. [PMID: 32846610 DOI: 10.1016/j.foodres.2020.109571] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 07/16/2020] [Accepted: 07/17/2020] [Indexed: 12/12/2022]
Abstract
Probiotics are defined as live microorganisms that, when administered in adequate amounts, confer a health benefit upon the host. At present, probiotics are gaining popularity worldwide and are widely used in food and medicine. Consumption of probiotics is increasing with further in-depth research on the relationship between intestinal flora and host health. Most people pay more attention to the function of probiotics but ignore their potential risks, such as infection and antibiotic resistance transfer to pathogenic microbes. Physiological functions, effects and mechanisms of action of probiotics were covered in this review, as well as the antibiotic resistance phenotypes, mechanisms and genes found in probiotics. Typical cases of antibiotic resistance of probiotics were also highlighted, as well as the potential risks (including pathogenicity, infectivity and excessive immune response) and corresponding strategies (dosage, formulation, and administration route). This timely study provides an avenue for further research, development and application of probiotics.
Collapse
|
43
|
Sadeghi-Bojd S, Naghshizadian R, Mazaheri M, Ghane Sharbaf F, Assadi F. Efficacy of Probiotic Prophylaxis After The First Febrile Urinary Tract Infection in Children With Normal Urinary Tracts. J Pediatric Infect Dis Soc 2020; 9:305-310. [PMID: 31100124 DOI: 10.1093/jpids/piz025] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2019] [Accepted: 04/29/2019] [Indexed: 12/19/2022]
Abstract
BACKGROUND Growing antibiotic resistance and debates over their efficacy for urinary tract infection (UTI) recurrence warrants studying nonantibiotic prophylaxis for preventing UTI recurrences. METHODS We randomly assigned 181 children, aged 4 months to 5 years, with a normal urinary tract after recovery from their first febrile UTI in a 1:1 ratio to receive a probiotic mixture of Lactobacillus acidophilus, Lactobacillus rhamnosus, Bifidobacterium bifidum, and Bifidobacterium lactis (n = 91) or placebo (n = 90) for a total of 18 months of therapy. The primary objective was to show the superiority of probiotic prophylaxis to placebo. The primary end point was composite cure (UTI-free survival) at 18 months, and the secondary end point was the median time to first UTI recurrence. RESULTS The probiotics were superior to placebo with respect to the primary efficacy end point. At 18 months, composite cure was observed in 96.7% (3 of 91) of the patients in the probiotic group and 83.3% (15 of 90) of those in the placebo group (P = .02). The median time to the first incidence of UTI recurrence was 3.5 months (range, 1-4 months) and 6.5 months (range, 2-14 months) in the probiotic and placebo groups, respectively (P = .04). The main microorganism that caused recurrent UTI was Escherichia coli, followed by Klebsiella pneumoniae, and these results were not significantly different between the 2 groups. We found no specific adverse events among the participants who received the probiotic mixture during the course of therapy. CONCLUSIONS The probiotics were more effective than placebo at reducing the risk of recurrent UTI in children with a normal urinary tract after their first episode of febrile UTI.
Collapse
Affiliation(s)
- S Sadeghi-Bojd
- Department of Pediatrics, Division of Nephrology, Zahedan University of Medical Sciences, Zahedan, Iran
| | - R Naghshizadian
- Department of Pediatrics, Section of Nephrology, Kurdistan University of Medical Science, Sanandaj, Iran
| | - M Mazaheri
- Department of Pediatrics, Section of Nephrology, Semnan University of Medical Science1 Semnan, Iran
| | - F Ghane Sharbaf
- Department of Pediatrics, Division of Nephrology, Dr Sheikh Hospital, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - F Assadi
- Department of Pediatrics, Division of Nephrology, Rush University Medical Center, Chicago, Illinois
| |
Collapse
|
44
|
Park IJ, Lee JH, Kye BH, Oh HK, Cho YB, Kim YT, Kim JY, Sung NY, Kang SB, Seo JM, Sim JH, Lee JL, Lee IK. Effects of PrObiotics on the Symptoms and Surgical ouTComes after Anterior REsection of Colon Cancer (POSTCARE): A Randomized, Double-Blind, Placebo-Controlled Trial. J Clin Med 2020; 9:jcm9072181. [PMID: 32664289 PMCID: PMC7408996 DOI: 10.3390/jcm9072181] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 06/25/2020] [Accepted: 07/02/2020] [Indexed: 01/08/2023] Open
Abstract
We investigated microbiota changes following surgical colon cancer resection and evaluate effects of probiotics on microbiota and surgical recovery. This randomized double-blind trial was performed at four medical centers in South Korea. Of 68 patients expected to undergo anterior sigmoid colon cancer resection, 60 were eligible, of whom 29 and 31 received probiotics and placebo, respectively, for four weeks, starting at one week preoperatively. Third- and/or fourth-week information on anterior resection syndrome (ARS), inflammatory markers, and quality of life was obtained. Stool sample analysis was conducted after randomization and bowel preparation and at three and four postoperative weeks. Bacteria were categorized into Set I (with probiotic effects) and II (colon cancer-associated). The probiotic group’s ARS score showed an improving trend (p = 0.063), particularly for flatus control (p = 0.030). Serum zonulin levels significantly decreased with probiotics. Probiotic ingestion resulted in compositional changes in gut microbiota; greater increases and decreases in Set I and II bacteria, respectively, occurred with probiotics. Compositional increase in Set I bacteria was associated with reduced white blood cells, neutrophils, neutrophil-lymphocyte ratio, and zonulin. Bifidobacterium composition was negatively correlated with zonulin levels in the probiotic group. Probiotics improved postoperative flatus control and modified postoperative changes in microbiota and inflammatory markers.
Collapse
Affiliation(s)
- In Ja Park
- Department of Colon and Rectal Surgery, Asan Medical Centre and University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul 05505, Korea;
| | - Ju-Hoon Lee
- Department of Food Science and Biotechnology, Graduate School of Biotechnology, Kyung Hee University, 1732, Deogyeong-daero, Giheung-gu, Yongin-si, Gyeonggi-do 17104, Korea; (J.-H.L.); (Y.-T.K.)
| | - Bong-Hyeon Kye
- Department of Surgery, St. Vincent’s Hospital, College of Medicine, The Catholic University of Korea, 93, Jungbu-daero, Paldal-gu, Suwon-si, Gyeonggi-do 16247, Korea;
| | - Heung-Kwon Oh
- Department of Surgery, Seoul National University Bundang Hospital, 300 Gumi-dong Bundang-gu, Seongnam-si, Gyeonggi-do 13620, Korea; (H.-K.O.); (S.-B.K.)
| | - Yong Beom Cho
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul 06351, Korea;
| | - You-Tae Kim
- Department of Food Science and Biotechnology, Graduate School of Biotechnology, Kyung Hee University, 1732, Deogyeong-daero, Giheung-gu, Yongin-si, Gyeonggi-do 17104, Korea; (J.-H.L.); (Y.-T.K.)
| | - Joo Yun Kim
- R&BD Centre, Korea Yakult Co. Ltd., 577, Gangnam-daero, Seocho-gu, Seoul 05505, Korea; (J.Y.K.); (J.-H.S.); (J.-L.L.)
| | - Na Young Sung
- National Cancer Control Institute, National Cancer Centre, 323 Ilsan-ro, Ilsandong-gu, Goyang-si Gyeonggi-do 10408, Korea;
| | - Sung-Bum Kang
- Department of Surgery, Seoul National University Bundang Hospital, 300 Gumi-dong Bundang-gu, Seongnam-si, Gyeonggi-do 13620, Korea; (H.-K.O.); (S.-B.K.)
| | - Jeong-Meen Seo
- Division of Pediatric Surgery, Samsung Medical Centre, Sungkyunkwan University School of Medicine, 81 Ilwon-ro, Gangnam-gu, Seoul 06351, Korea;
| | - Jae-Hun Sim
- R&BD Centre, Korea Yakult Co. Ltd., 577, Gangnam-daero, Seocho-gu, Seoul 05505, Korea; (J.Y.K.); (J.-H.S.); (J.-L.L.)
| | - Jung-Lyoul Lee
- R&BD Centre, Korea Yakult Co. Ltd., 577, Gangnam-daero, Seocho-gu, Seoul 05505, Korea; (J.Y.K.); (J.-H.S.); (J.-L.L.)
| | - In Kyu Lee
- Department of Surgery, Division of Colorectal Surgery, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul 06591, Korea
- Correspondence: ; Tel.: +82-2-2258-6104
| |
Collapse
|
45
|
Yeo S, Park H, Seo E, Kim J, Kim BK, Choi IS, Huh CS. Anti-Inflammatory and Gut Microbiota Modulatory Effect of Lactobacillus rhamnosus Strain LDTM 7511 in a Dextran Sulfate Sodium-Induced Colitis Murine Model. Microorganisms 2020; 8:E845. [PMID: 32512895 PMCID: PMC7356973 DOI: 10.3390/microorganisms8060845] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 05/27/2020] [Accepted: 06/02/2020] [Indexed: 12/14/2022] Open
Abstract
Inflammatory bowel disease (IBD) is a group of conditions involving chronic relapsing-remitting inflammation of the gastrointestinal tract with an unknown etiology. Although the cause-effect relationship between gut microbiota and IBD has not been clearly established, emerging evidence from experimental models supports the idea that gut microbes play a fundamental role in the pathogenesis of IBD. As microbiome-based therapeutics for IBD, the beneficial effects of probiotics have been found in animal colitis models and IBD patients. In this study, based on the dextran sulfate sodium (DSS)-induced colitis mouse model, we investigated Lactobacillus rhamnosus strain LDTM 7511 originating from Korean infant feces as a putative probiotic strain for IBD. The strain LDTM 7511 not only alleviated the release of inflammatory mediators, but also induced the transition of gut microbiota from dysbiotic conditions, exhibiting the opposite pattern in the abundance of DSS colitis-associated bacterial taxa to the DSS group. Our findings suggest that the strain LDTM 7511 has the potential to be used as a probiotic treatment for IBD patients in comparison to L. rhamnosus GG (ATCC 53103), which has been frequently used for IBD studies.
Collapse
Affiliation(s)
- Soyoung Yeo
- WCU Biomodulation Major, Department of Agricultural Biotechnology, College of Agriculture and Life Sciences, Seoul National University, Seoul 08826, Korea; (S.Y.); (E.S.); (J.K.)
| | - Hyunjoon Park
- Research Institute of Eco-Friendly Livestock Science, Institute of Green-Bio Science and Technology, Seoul National University, Pyeongchang 25354, Korea;
| | - Eunsol Seo
- WCU Biomodulation Major, Department of Agricultural Biotechnology, College of Agriculture and Life Sciences, Seoul National University, Seoul 08826, Korea; (S.Y.); (E.S.); (J.K.)
| | - Jihee Kim
- WCU Biomodulation Major, Department of Agricultural Biotechnology, College of Agriculture and Life Sciences, Seoul National University, Seoul 08826, Korea; (S.Y.); (E.S.); (J.K.)
- Chong Kun Dang Bio Research Institute, Chong Kun Dang Bio Research Institute, Ansan 15604, Korea; (B.K.K.); (I.S.C.)
| | - Byoung Kook Kim
- Chong Kun Dang Bio Research Institute, Chong Kun Dang Bio Research Institute, Ansan 15604, Korea; (B.K.K.); (I.S.C.)
| | - In Suk Choi
- Chong Kun Dang Bio Research Institute, Chong Kun Dang Bio Research Institute, Ansan 15604, Korea; (B.K.K.); (I.S.C.)
| | - Chul Sung Huh
- Research Institute of Eco-Friendly Livestock Science, Institute of Green-Bio Science and Technology, Seoul National University, Pyeongchang 25354, Korea;
- Graduate School of International Agricultural Technology, Seoul National University, Pyeongchang 25354, Korea
| |
Collapse
|
46
|
Sendil S, Shrimanker I, Mansoora Q, Goldman J, Nookala VK. Lactobacillus rhamnosus Bacteremia in an Immunocompromised Renal Transplant Patient. Cureus 2020; 12:e6887. [PMID: 32190450 PMCID: PMC7058391 DOI: 10.7759/cureus.6887] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Conventionally, Lactobacillus species are considered as low virulence organisms and rarely cause infection in immunocompetent individuals. However, it might be an opportunistic infection source in immunocompromised patients and can cause invasive serious infections. To our knowledge, there are only a handful of cases in the literature reporting primary bloodstream infection caused by Lactobacilli spp. in transplant recipients. Here, we report a case of a kidney transplant recipient with Lactobacillus rhamnosus bacteremia.
Collapse
Affiliation(s)
- Selin Sendil
- Internal Medicine, University of Pittsburgh Medical Center Pinnacle, Harrisburg, USA
| | - Isha Shrimanker
- Internal Medicine, University of Pittsburgh Medical Center Pinnacle, Harrisburg, USA
| | - Qurat Mansoora
- Internal Medicine, University of Pittsburgh Medical Center Pinnacle, Harrisburg, USA
| | - John Goldman
- Internal Medicine: Infectious Disease, University of Pittsburgh Medical Center Pinnacle, Harrisburg, USA
| | - Vinod K Nookala
- Internal Medicine, University of Pittsburgh Medical Center Pinnacle, Harrisburg, USA
| |
Collapse
|
47
|
Pradhan D, Mallappa RH, Grover S. Comprehensive approaches for assessing the safety of probiotic bacteria. Food Control 2020. [DOI: 10.1016/j.foodcont.2019.106872] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
48
|
Zama D, Bossù G, Leardini D, Muratore E, Biagi E, Prete A, Pession A, Masetti R. Insights into the role of intestinal microbiota in hematopoietic stem-cell transplantation. Ther Adv Hematol 2020; 11:2040620719896961. [PMID: 32010434 PMCID: PMC6974760 DOI: 10.1177/2040620719896961] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Accepted: 11/20/2019] [Indexed: 12/11/2022] Open
Abstract
The gut microbiota (GM) is able to modulate the human immune system. The development of novel investigation methods has provided better characterization of the GM, increasing our knowledge of the role of GM in the context of hematopoietic stem-cell transplantation (HSCT). In particular, the GM influences the development of the major complications seen after HSCT, having an impact on overall survival. In fact, this evidence highlights the possible therapeutic implications of modulation of the GM during HSCT. Insights into the complex mechanisms and functions of the GM are essential for the rational design of these therapeutics. To date, preemptive and curative approaches have been tested. The current state of understanding of the impact of the GM on HSCT, and therapies targeting the GM balance is reviewed herein.
Collapse
Affiliation(s)
- Daniele Zama
- Pediatric Oncology and Hematology Unit ‘Lalla
Seràgnoli,’ Sant’Orsola-Malpighi Hospital, University of Bologna, Via
Massarenti 11, Bologna, 40137, Italy
| | - Gianluca Bossù
- Department of Pediatrics, ‘Lalla Seràgnoli,’
Hematology-Oncology Unit, University of Bologna, Italy
| | - Davide Leardini
- Department of Pediatrics, ‘Lalla Seràgnoli,’
Hematology-Oncology Unit, University of Bologna, Italy
| | - Edoardo Muratore
- Department of Pediatrics, ‘Lalla Seràgnoli,’
Hematology-Oncology Unit, University of Bologna, Italy
| | - Elena Biagi
- Department of Pharmacy and Biotechnology,
University of Bologna, Bologna, Italy
| | - Arcangelo Prete
- Department of Pediatrics, ‘Lalla Seràgnoli,’
Hematology-Oncology Unit, University of Bologna, Italy
| | - Andrea Pession
- Department of Pediatrics, ‘Lalla Seràgnoli,’
Hematology-Oncology Unit, University of Bologna, Italy
| | - Riccardo Masetti
- Department of Pediatrics, ‘Lalla Seràgnoli,’
Hematology-Oncology Unit, University of Bologna, Italy
| |
Collapse
|
49
|
Bischoff SC, Escher J, Hébuterne X, Kłęk S, Krznaric Z, Schneider S, Shamir R, Stardelova K, Wierdsma N, Wiskin AE, Forbes A. ESPEN practical guideline: Clinical Nutrition in inflammatory bowel disease. Clin Nutr 2020; 39:632-653. [PMID: 32029281 DOI: 10.1016/j.clnu.2019.11.002] [Citation(s) in RCA: 187] [Impact Index Per Article: 46.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Accepted: 11/01/2019] [Indexed: 02/06/2023]
Abstract
The present guideline is the first of a new series of "practical guidelines" based on more detailed scientific guidelines produced by ESPEN during the last few years. The guidelines have been shortened and now include flow charts that connect the individual recommendations to logical care pathways and allow rapid navigation through the guideline. The purpose of the present practical guideline is to provide an easy-to-use tool to guide nutritional support and primary nutritional therapy in inflammatory bowel disease (IBD). The guideline is aimed at professionals working in clinical practice, either in hospitals or in outpatient medicine, and treating patients with IBD. In 40 recommendations, general aspects of care in patients with IBD, and specific aspects during active disease and in remission are addressed. All recommendations are equipped with evidence grades, consensus rates, short commentaries and links to cited literature.
Collapse
Affiliation(s)
- Stephan C Bischoff
- University of Hohenheim, Institute of Nutritional Medicine, Stuttgart, Germany.
| | - Johanna Escher
- Erasmus Medical Center - Sophia Children's Hospital, Rotterdam, the Netherlands
| | - Xavier Hébuterne
- Gastroentérologie et Nutrition Clinique, CHU de Nice, Université Côte d'Azur, Nice, France
| | - Stanisław Kłęk
- General and Oncology Surgery Unit, Stanley Dudrick's Memorial Hospital, Krakow, Poland
| | - Zeljko Krznaric
- Clinical Hospital Centre Zagreb, University of Zagreb, Zagreb, Croatia
| | - Stéphane Schneider
- Gastroentérologie et Nutrition Clinique, CHU de Nice, Université Côte d'Azur, Nice, France
| | - Raanan Shamir
- Tel-Aviv University, Schneider Children's Medical Center of Israel, Petach-Tikva, Israel
| | - Kalina Stardelova
- University Clinic for Gasrtroenterohepatology, Clinal Centre "Mother Therese", Skopje, Macedonia
| | | | - Anthony E Wiskin
- Pediatric Gastroenterology & Nutrition Unit, Bristol Royal Hospital for Children, Bristol, United Kingdom
| | - Alastair Forbes
- Norwich Medical School, University of East Anglia, Norwich, United Kingdom
| |
Collapse
|
50
|
Kaban RK, Wardhana, Hegar B, Rohsiswatmo R, Handryastuti S, Amelia N, Muktiarti D, Indrio F, Vandenplas Y. Lactobacillus reuteri DSM 17938 Improves Feeding Intolerance in Preterm Infants. Pediatr Gastroenterol Hepatol Nutr 2019; 22:545-553. [PMID: 31777720 PMCID: PMC6856506 DOI: 10.5223/pghn.2019.22.6.545] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2019] [Accepted: 06/15/2019] [Indexed: 01/29/2023] Open
Abstract
PURPOSE Feeding tolerance is extremely important in preterm infants. This study aimed to evaluate whether preterm infants receiving Lactobacillus reuteri DSM 17938 would develop fewer symptoms of feeding intolerance. Secondary outcomes were duration of parenteral nutrition, time to reach full feeding, length of hospital stay, sepsis, necrotizing enterocolitis (NEC), diarrhea, and mortality. METHODS This double-blind randomized controlled trial of L. reuteri DSM 17938 versus placebo included 94 neonates with a gestational age of 28-34 weeks and birth weight of 1,000-1,800 g. RESULTS Feeding intolerance (vomiting and/or distension) was less common in the probiotic group than in the placebo group (8.5% vs. 25.5%; relative risk, 0.33; 95% confidence interval, 0.12-0.96; p=0.03). No significant intergroup differences were found in proven sepsis, time to reach full feeding, length of hospital stay, or diarrhea. The prevalence of NEC (stages 2 and 3) was 6.4% in the placebo group vs. 0% in the probiotic group (relative risk, 1.07; 95% confidence interval, 0.99-1.15; p=0.24). Mortality rates were 2.1% in the probiotic group and 8.5% in the placebo group, p=0.36). CONCLUSION The administration of L. reuteri DSM 17938 to preterm infants was safe and significantly reduced feeding intolerance. No significant differences were found in any other secondary outcomes.
Collapse
Affiliation(s)
- Risma K. Kaban
- Department of Pediatric Health, Faculty of Medicine, Universitas Indonesia–Dr. Cipto Mangunkusumo Hospital, Jakarta, Indonesia
| | - Wardhana
- Department of Pediatric Health, Faculty of Medicine, Universitas Indonesia–Dr. Cipto Mangunkusumo Hospital, Jakarta, Indonesia
| | - Badriul Hegar
- Department of Pediatric Health, Faculty of Medicine, Universitas Indonesia–Dr. Cipto Mangunkusumo Hospital, Jakarta, Indonesia
| | - Rinawati Rohsiswatmo
- Department of Pediatric Health, Faculty of Medicine, Universitas Indonesia–Dr. Cipto Mangunkusumo Hospital, Jakarta, Indonesia
| | - Setyo Handryastuti
- Department of Pediatric Health, Faculty of Medicine, Universitas Indonesia–Dr. Cipto Mangunkusumo Hospital, Jakarta, Indonesia
| | - Novie Amelia
- Department of Pediatric Health, Faculty of Medicine, Universitas Indonesia–Dr. Cipto Mangunkusumo Hospital, Jakarta, Indonesia
| | - Dina Muktiarti
- Department of Pediatric Health, Faculty of Medicine, Universitas Indonesia–Dr. Cipto Mangunkusumo Hospital, Jakarta, Indonesia
| | - Flavia Indrio
- Department of Paediatrics, University of Bari, Bari, Italy
| | - Yvan Vandenplas
- KidZ Health Castle, UZ Brussel, Vrije Universiteit Brussel, Brussels, Belgium
| |
Collapse
|