1
|
Fabiano GA, Oliveira RPS, Rodrigues S, Santos BN, Venema K, Antunes AEC. Evidence of synbiotic potential of oat beverage enriched with inulin and fermented by L. rhamnosus LR B in a dynamic in vitro model of human colon. Food Res Int 2025; 211:116489. [PMID: 40356187 DOI: 10.1016/j.foodres.2025.116489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Revised: 04/14/2025] [Accepted: 04/16/2025] [Indexed: 05/15/2025]
Abstract
Fermented dairy products are known for their efficiency in delivering and protecting probiotic microorganisms. However, there is a growing demand for diversification of the market with plant-based products. The aim of this study was to develop an oat beverage enriched with inulin and fermented with Lacticaseibacillus rhamnosus LR B and evaluate its synbiotic effects in vitro. For this purpose, the validated dynamic colon model (the TNO Intestinal Model TIM-2) was used with focus on the composition of the gut microbiota and its production of metabolites to evaluate the functionality. The fermentation kinetics, sugars, organic acids and inulin dosage in the fermented oat beverage were also evaluated. The acidification rate was 16.91 10-3 pH units.min-1, reaching the final pH of 4.5 in 2.38 ± 0.05 h. Dosages of sucrose, glucose and lactic acid were 23.35 ± 0.45 g.L-1, 21.37 ± 0.77 g.L-1, 0.94 ± 0.05 g.L-1, respectively. After simulated in vitro digestion, the inulin concentration was partially preserved with 20.11 ± 0.21 maltose equivalent (μg.mL-1). The fermented and pre-digested oat beverage (with 7.71 ± 0.44 log CFU.mL-1) was fed into TIM-2, which was previously inoculated with feces from healthy adults. The analysis identified nine bacterial taxa that were significantly modulated compared to the standard ileal effluent medium (SIEM) control. An increase in relative abundance of Lactobacillus and Catenibacterium, and reduction in Citrobacter, Escherichia-Shigella, and Klebsiella was observed. In addition, the cumulative means of short-chain fatty acids (SCFAs) increased, especially for acetate and butyrate. These findings suggest that the developed oat beverage can positively influence the gut microbiota and its activity, highlighting possible health benefits.
Collapse
Affiliation(s)
- G A Fabiano
- Faculdade de Ciências Aplicadas, Universidade Estadual de Campinas (UNICAMP), Limeira, São Paulo, Brazil
| | - R P S Oliveira
- Department of Biochemical-Pharmaceutical Technology, Faculty of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - S Rodrigues
- Department of Food Engineering, Federal University of Ceara, Fortaleza, Ceara, Brazil
| | - B N Santos
- Department of Chemical Engineering, Federal University of Ceara, Fortaleza, Ceara, Brazil
| | - K Venema
- Maastricht University, Centre for Healthy Eating & Food Innovation (HEFI), Venlo, the Netherlands
| | - A E C Antunes
- Faculdade de Ciências Aplicadas, Universidade Estadual de Campinas (UNICAMP), Limeira, São Paulo, Brazil.
| |
Collapse
|
2
|
Rahim MA, Seo H, Barman I, Hossain MS, Shuvo MSH, Song HY. Insights into Autophagy in Microbiome Therapeutic Approaches for Drug-Resistant Tuberculosis. Cells 2025; 14:540. [PMID: 40214493 PMCID: PMC11989032 DOI: 10.3390/cells14070540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Revised: 03/23/2025] [Accepted: 04/02/2025] [Indexed: 04/14/2025] Open
Abstract
Tuberculosis, primarily caused by Mycobacterium tuberculosis, is an airborne lung disease and continues to pose a significant global health threat, resulting in millions of deaths annually. The current treatment for tuberculosis involves a prolonged regimen of antibiotics, which leads to complications such as recurrence, drug resistance, reinfection, and a range of side effects. This scenario underscores the urgent need for novel therapeutic strategies to combat this lethal pathogen. Over the last two decades, microbiome therapeutics have emerged as promising next-generation drug candidates, offering advantages over traditional medications. In 2022, the Food and Drug Administration approved the first microbiome therapeutic for recurrent Clostridium infections, and extensive research is underway on microbiome treatments for various challenging diseases, including metabolic disorders and cancer. Research on microbiomes concerning tuberculosis commenced roughly a decade ago, and the scope of this research has broadened considerably over the last five years, with microbiome therapeutics now viewed as viable options for managing drug-resistant tuberculosis. Nevertheless, the understanding of their mechanisms is still in its infancy. Although autophagy has been extensively studied in other diseases, research into its role in tuberculosis is just beginning, with preliminary developments in progress. Against this backdrop, this comprehensive review begins by succinctly outlining tuberculosis' characteristics and assessing existing treatments' strengths and weaknesses, followed by a detailed examination of microbiome-based therapeutic approaches for drug-resistant tuberculosis. Additionally, this review focuses on establishing a basic understanding of microbiome treatments for tuberculosis, mainly through the lens of autophagy as a mechanism of action. Ultimately, this review aims to contribute to the foundational comprehension of microbiome-based therapies for tuberculosis, thereby setting the stage for the further advancement of microbiome therapeutics for drug-resistant tuberculosis.
Collapse
Affiliation(s)
- Md Abdur Rahim
- Department of Microbiology and Immunology, School of Medicine, Soonchunhyang University, 31, Suncheonhyang 6-gil, Dongnam-gu, Cheonan-si 31151, Republic of Korea
- Human Microbiome Medical Research Center (HM·MRC), School of Medicine, Soonchunhyang University, 22, Soonchunhyang-ro, Sinchang-myeon, Asan-si 31538, Republic of Korea
| | - Hoonhee Seo
- Human Microbiome Medical Research Center (HM·MRC), School of Medicine, Soonchunhyang University, 22, Soonchunhyang-ro, Sinchang-myeon, Asan-si 31538, Republic of Korea
- Probiotics Microbiome Commercialization Research Center (PMC), Soonchunhyang University, 22, Soonchunhyang-ro, Sinchang-myeon, Asan-si 31538, Republic of Korea
| | - Indrajeet Barman
- Department of Microbiology and Immunology, School of Medicine, Soonchunhyang University, 31, Suncheonhyang 6-gil, Dongnam-gu, Cheonan-si 31151, Republic of Korea
- Human Microbiome Medical Research Center (HM·MRC), School of Medicine, Soonchunhyang University, 22, Soonchunhyang-ro, Sinchang-myeon, Asan-si 31538, Republic of Korea
| | - Mohammed Solayman Hossain
- Department of Microbiology and Immunology, School of Medicine, Soonchunhyang University, 31, Suncheonhyang 6-gil, Dongnam-gu, Cheonan-si 31151, Republic of Korea
- Human Microbiome Medical Research Center (HM·MRC), School of Medicine, Soonchunhyang University, 22, Soonchunhyang-ro, Sinchang-myeon, Asan-si 31538, Republic of Korea
| | - Md Sarower Hossen Shuvo
- Department of Microbiology and Immunology, School of Medicine, Soonchunhyang University, 31, Suncheonhyang 6-gil, Dongnam-gu, Cheonan-si 31151, Republic of Korea
- Human Microbiome Medical Research Center (HM·MRC), School of Medicine, Soonchunhyang University, 22, Soonchunhyang-ro, Sinchang-myeon, Asan-si 31538, Republic of Korea
| | - Ho-Yeon Song
- Department of Microbiology and Immunology, School of Medicine, Soonchunhyang University, 31, Suncheonhyang 6-gil, Dongnam-gu, Cheonan-si 31151, Republic of Korea
- Human Microbiome Medical Research Center (HM·MRC), School of Medicine, Soonchunhyang University, 22, Soonchunhyang-ro, Sinchang-myeon, Asan-si 31538, Republic of Korea
- Probiotics Microbiome Commercialization Research Center (PMC), Soonchunhyang University, 22, Soonchunhyang-ro, Sinchang-myeon, Asan-si 31538, Republic of Korea
| |
Collapse
|
3
|
Zhu CY, Byun H, Do EA, Zhang Y, Tanchoco E, Beld J, Hsiao A, Zhu J. Music exposure enhances resistance to Salmonella infection by promoting healthy gut microbiota. Microbiol Spectr 2025; 13:e0237724. [PMID: 40130867 PMCID: PMC12054044 DOI: 10.1128/spectrum.02377-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Accepted: 02/28/2025] [Indexed: 03/26/2025] Open
Abstract
Music intervention is gaining recognition as a cost-effective therapeutic for improving human health. Despite its growing application, the mechanisms through which music exerts beneficial health effects remain largely unexplored. Here, we show that music can exert beneficial effects in mice through modulating gut microbiome composition. Adult mice were exposed to ambient noise, Mozart's Flute Quartet in D Major, K. 285, or white noise over a three-week period. Afterward, we observed treatment-specific changes in the community of gut commensal bacteria in these animals. Upon subsequent challenge with the bacterial pathogen Salmonella typhimurium, control groups exhibited significant weight loss and increased Salmonella colonization, whereas the Mozart-treated group did not. 16S ribosomal RNA gene sequencing revealed that the Mozart group showed a significant increase in Lactobacillus salivarius, a probiotic known for its antibacterial properties. Further experiments confirmed that L. salivarius mitigated Salmonella infection in mice and that L. salivarius acidified local environments in in vitro culture, thus inhibiting Salmonella growth. Additionally, mice exposed to Mozart consumed more food but showed similar body weight compared to the control groups. Behavioral assessments, including open field and object location tests, revealed that Mozart-treated mice were more active, less anxious, and exhibited enhanced spatial memory. Finally, Mozart exposure was shown to significantly boost colonization of administered L. salivarius and alter gut metabolite profiles. These findings suggest that music exposure fosters healthier gut microbiota, enhancing resistance to bacterial infections and highlighting the potential of music therapy as a novel strategy to combat drug-resistant pathogen infections. IMPORTANCE Music therapy is increasingly recognized as a low-cost approach to improving health, but how it works remains unclear. Our study demonstrates that music can positively influence health by altering the gut microbiome. In a mouse model, exposure to Mozart's Flute Quartet in D Major enhanced the gut microbiota, specifically increasing levels of the beneficial bacterium Lactobacillus salivarius. This probiotic protected mice from Salmonella infection by creating an acidic environment that inhibited pathogen growth. Mozart-treated mice also showed reduced anxiety, better spatial memory, and higher food intake without weight gain, suggesting the benefits of music exposure. These findings reveal a novel link between music, gut health, and disease resistance, suggesting that music therapy could be a promising strategy for enhancing gut microbiota and combating infections, including those caused by drug-resistant bacteria.
Collapse
Affiliation(s)
- Clara Y. Zhu
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Hyuntae Byun
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Elyza A. Do
- Department of Microbiology & Plant Pathology, University of California Riverside, Riverside, California, USA
| | - Yue Zhang
- Department of Microbiology & Plant Pathology, University of California Riverside, Riverside, California, USA
| | - Ethan Tanchoco
- Department of Microbiology & Plant Pathology, University of California Riverside, Riverside, California, USA
| | - Joris Beld
- Department of Microbiology & Immunology, College of Medicine, Drexel University, Philadelphia, Pennsylvania, USA
| | - Ansel Hsiao
- Department of Microbiology & Plant Pathology, University of California Riverside, Riverside, California, USA
| | - Jun Zhu
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
4
|
Wu X, Du J, Zhou X, Peng X, Jia C, Wang B, Wu B, Li Y, Yue M. Genomic epidemiology and public health implications of zoonotic monophasic Salmonella Typhimurium ST34. Front Cell Infect Microbiol 2025; 15:1490183. [PMID: 40134787 PMCID: PMC11933091 DOI: 10.3389/fcimb.2025.1490183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Accepted: 02/21/2025] [Indexed: 03/27/2025] Open
Abstract
Background Monophasic Salmonella Typhimurium sequence type 34 (mSTM ST34) has emerged as a significant global health threat, but our understanding of its genomic epidemiology and potential public health implications in international and regional contexts remains limited. This study aims to fill this crucial gap by assessing the genomic epidemiology of multidrug resistance (MDR) mSTM ST34, as well as its clinical characteristics and virulence. Methods To achieve the objectives of this study, we conducted a comprehensive genomic analysis of mSTM ST34 isolates. We obtained a global dataset comprising 13,844 strains from public databases, along with 339 strains from a regional surveillance collection in Zhejiang Province, China. This dataset aims to provide in-depth insights into antimicrobial resistance, mobile genetic elements, and pathogenicity. Additionally, we meticulously assessed the association between phenotypic profiles and clinical presentations. Results Our findings revealed that the prevalence of mSTM ST34 has surpassed that of the previously dominant ST19. In addition, we observed an increase in the detection of the IncQ1 plasmid, which is responsible for disseminating MDR. The prevalence of mSTM ST34 carriage was exceptionally high among children (≤12 years old) and elderly individuals (≥65 years old), with 92.6% of the isolates exhibiting MDR, including resistance to frontline antimicrobials such as third-generation cephalosporins and ciprofloxacin. Additionally, the human mSTM ST34 strain demonstrates a remarkable capacity for biofilm formation, which increases its virulence in animal models and complicates therapeutic interventions. Conclusions mSTM ST34 has surpassed the previously dominant ST19, and its ability to transmit across multi-species increases its potential for further human transmission. This study addresses critical gaps in our understanding of mSTM ST34 prevalence, highlighting the importance of whole genome sequencing in surveilling zoonotic pathogens.
Collapse
Affiliation(s)
- Xiaolei Wu
- Department of Veterinary Medicine, Zhejiang University College of Animal Sciences, Hangzhou, China
| | - Jiaxin Du
- Department of Veterinary Medicine, Zhejiang University College of Animal Sciences, Hangzhou, China
| | - Xiao Zhou
- Department of Veterinary Medicine, Zhejiang University College of Animal Sciences, Hangzhou, China
| | - Xianqi Peng
- Department of Veterinary Medicine, Zhejiang University College of Animal Sciences, Hangzhou, China
| | - Chenghao Jia
- Department of Veterinary Medicine, Zhejiang University College of Animal Sciences, Hangzhou, China
| | - Baikui Wang
- Department of Veterinary Medicine, Zhejiang University College of Animal Sciences, Hangzhou, China
| | - Beibei Wu
- Institute of Tuberculosis Control, Zhejiang Provincial Center for Disease Control and Prevention, Hangzhou, China
| | - Yan Li
- Department of Veterinary Medicine, Zhejiang University College of Animal Sciences, Hangzhou, China
- Hainan Institute of Zhejiang University, Sanya, China
| | - Min Yue
- Department of Veterinary Medicine, Zhejiang University College of Animal Sciences, Hangzhou, China
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, China
| |
Collapse
|
5
|
Chen M, Li Y, Zhai Z, Wang H, Lin Y, Chang F, Ge S, Sun X, Wei W, Wang D, Zhang M, Chen R, Yu H, Feng T, Huang X, Cheng D, Liu J, Di W, Hao Y, Yin P, Tang P. Bifidobacterium animalis subsp. lactis A6 ameliorates bone and muscle loss via modulating gut microbiota composition and enhancing butyrate production. Bone Res 2025; 13:28. [PMID: 40000617 PMCID: PMC11862215 DOI: 10.1038/s41413-024-00381-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 09/25/2024] [Accepted: 10/15/2024] [Indexed: 02/27/2025] Open
Abstract
Systematic bone and muscle loss is a complex metabolic disease, which is frequently linked to gut dysfunction, yet its etiology and treatment remain elusive. While probiotics show promise in managing diseases through microbiome modulation, their therapeutic impact on gut dysfunction-induced bone and muscle loss remains to be elucidated. Employing dextran sulfate sodium (DSS)-induced gut dysfunction model and wide-spectrum antibiotics (ABX)-treated mice model, our study revealed that gut dysfunction instigates muscle and bone loss, accompanied by microbial imbalances. Importantly, Bifidobacterium animalis subsp. lactis A6 (B. lactis A6) administration significantly ameliorated muscle and bone loss by modulating gut microbiota composition and enhancing butyrate-producing bacteria. This intervention effectively restored depleted butyrate levels in serum, muscle, and bone tissues caused by gut dysfunction. Furthermore, butyrate supplementation mitigated musculoskeletal loss by repairing the damaged intestinal barrier and enriching beneficial butyrate-producing bacteria. Importantly, butyrate inhibited the NF-κB pathway activation, and reduced the secretion of corresponding inflammatory factors in T cells. Our study highlights the critical role of dysbiosis in gut dysfunction-induced musculoskeletal loss and underscores the therapeutic potential of B. lactis A6. These discoveries offer new microbiome directions for translational and clinical research, providing promising strategies for preventing and managing musculoskeletal diseases.
Collapse
Affiliation(s)
- Ming Chen
- Senior Department of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing, China
- National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, Beijing, China
| | - Yi Li
- Senior Department of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing, China
- National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, Beijing, China
| | - Zhengyuan Zhai
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, China
| | - Hui Wang
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, China
| | - Yuan Lin
- The Department of Orthopedic Surgery, Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Feifan Chang
- Senior Department of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing, China
- National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, Beijing, China
| | - Siliang Ge
- Senior Department of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing, China
- National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, Beijing, China
| | - Xinyu Sun
- Senior Department of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing, China
- National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, Beijing, China
| | - Wei Wei
- Department of Clinical Nutrition, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Duanyang Wang
- The Department of Orthopedic Surgery, Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Mingming Zhang
- Senior Department of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing, China
- National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, Beijing, China
| | - Ruijing Chen
- Senior Department of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing, China
- National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, Beijing, China
| | - Haikuan Yu
- Senior Department of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing, China
- National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, Beijing, China
| | - Taojin Feng
- Senior Department of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing, China
- National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, Beijing, China
| | - Xiang Huang
- Senior Department of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing, China
- National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, Beijing, China
| | - Dongliang Cheng
- Senior Department of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing, China
- National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, Beijing, China
| | - Jiang Liu
- The Department of Orthopedic Surgery, Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Wenxuan Di
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, China
| | - Yanling Hao
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, China.
| | - Pengbin Yin
- Senior Department of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing, China.
- National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, Beijing, China.
| | - Peifu Tang
- Senior Department of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing, China
- National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, Beijing, China
| |
Collapse
|
6
|
Ibrahim M, Bednarczyk M, Stadnicka K, Grochowska E. Inter- and Transgenerational Effects of In Ovo Stimulation with Bioactive Compounds on Cecal Tonsils and Cecal Mucosa Transcriptomes in a Chicken Model. Int J Mol Sci 2025; 26:1174. [PMID: 39940944 PMCID: PMC11817890 DOI: 10.3390/ijms26031174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 01/14/2025] [Accepted: 01/17/2025] [Indexed: 02/16/2025] Open
Abstract
Exploring how early-life nutritional interventions may impact future generations, this study examines the inter- and transgenerational effects of in ovo injection of bioactive compounds on gene expression in the cecal tonsils and cecal mucosa using a chicken model. Synbiotic PoultryStar® (Biomin) and choline were injected in ovo on the 12th day of egg incubation. Three experimental groups were established in the generation F1: (1) a control group (C) receiving 0.9% physiological saline (NaCl), (2) a synbiotic group (SYN) receiving 2 mg/embryo, and (3) a combined synbiotic and choline group (SYNCH) receiving 2 mg synbiotic and 0.25 mg choline per embryo. For the generations F2 and F3, the SYN and SYNCH groups were each divided into two subgroups: (A) those injected solely in F1 (SYNs and SYNCHs) and (B) those injected in each generation (SYNr and SYNCHr). At 21 weeks posthatching, cecal tonsil and cecal mucosa samples were collected from F1, F2, and F3 birds for transcriptomic analysis. Gene expression profiling revealed distinct intergenerational and transgenerational patterns in both tissues. In cecal tonsils, a significant transgenerational impact on gene expression was noted in the generation F3, following a drop in F2. In contrast, cecal mucosa showed more gene expression changes in F2, indicating intergenerational effects. While some effects carried into F3, they were less pronounced, except in the SYNs group, which experienced an increase compared to F2. The study highlights that transgenerational effects of epigenetic modifications are dynamic and unpredictable, with effects potentially re-emerging in later generations under certain conditions or fading or intensifying over time. This study provides valuable insights into how epigenetic nutritional stimulation during embryonic development may regulate processes in the cecal tonsils and cecal mucosa across multiple generations. Our findings provide evidence supporting the phenomenon of epigenetic dynamics in a chicken model.
Collapse
Affiliation(s)
- Mariam Ibrahim
- Faculty of Health Sciences, Collegium Medicum, Nicolaus Copernicus University, Łukasiewicza 1, 85-821 Bydgoszcz, Poland; (M.I.); (K.S.)
- PBS Doctoral School, Bydgoszcz University of Science and Technology, Aleje prof. S. Kaliskiego 7, 85-796 Bydgoszcz, Poland
| | - Marek Bednarczyk
- Department of Animal Biotechnology and Genetics, Bydgoszcz University of Science and Technology, Mazowiecka 28, 85-084 Bydgoszcz, Poland
| | - Katarzyna Stadnicka
- Faculty of Health Sciences, Collegium Medicum, Nicolaus Copernicus University, Łukasiewicza 1, 85-821 Bydgoszcz, Poland; (M.I.); (K.S.)
| | - Ewa Grochowska
- Faculty of Health Sciences, Collegium Medicum, Nicolaus Copernicus University, Łukasiewicza 1, 85-821 Bydgoszcz, Poland; (M.I.); (K.S.)
| |
Collapse
|
7
|
Zhong L, Zheng J, Wang Z, Lin L, Cong Q, Qiao L. Metabolomics and proteomics reveal the inhibitory effect of Lactobacillus crispatus on cervical cancer. Talanta 2025; 281:126839. [PMID: 39265423 DOI: 10.1016/j.talanta.2024.126839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 09/04/2024] [Accepted: 09/07/2024] [Indexed: 09/14/2024]
Abstract
Cervical cancer remains a significant global health issue due to its high morbidity and mortality rates. Recently, Lactobacillus crispatus has been recognized for its crucial role in maintaining cervical health. While some studies have explored the use of L. crispatus to mitigate cervical cancer, the underlying mechanisms remain largely unknown. In this study, we employed non-targeted proteomics and metabolomics to investigate how L. crispatus affects the growth of cervical cancer cells (SiHa) and normal cervical cells (Ect1/E6E7). Our findings indicated that the inhibitory effect of L. crispatus on SiHa cells was associated with various biological processes, notably the ferroptosis pathway. Specifically, L. crispatus was found to regulate the expression of proteins such as HMOX1, SLC39A14, VDAC2, ACSL4, and LPCAT3 by SiHa cells, which are closely related to ferroptosis. Additionally, it activated the tricarboxylic acid (TCA) cycle in SiHa cells, leading to increased levels of reactive oxygen species (ROS) and lipid peroxides (LPO). These results revealed the therapeutic potential of L. crispatus in targeting the ferroptosis pathway for cervical cancer treatment, opening new avenues for research and therapy in cervical cancer.
Collapse
Affiliation(s)
- Lingyan Zhong
- Department of Chemistry, Zhongshan Hospital, and Obstetrics and Gynecology Hospital, Fudan University, Shanghai, 200000, China
| | - Jianxujie Zheng
- Department of Chemistry, Zhongshan Hospital, and Obstetrics and Gynecology Hospital, Fudan University, Shanghai, 200000, China
| | - Zengyu Wang
- Department of Chemistry, Zhongshan Hospital, and Obstetrics and Gynecology Hospital, Fudan University, Shanghai, 200000, China
| | - Ling Lin
- Department of Chemistry, Zhongshan Hospital, and Obstetrics and Gynecology Hospital, Fudan University, Shanghai, 200000, China.
| | - Qing Cong
- Department of Chemistry, Zhongshan Hospital, and Obstetrics and Gynecology Hospital, Fudan University, Shanghai, 200000, China.
| | - Liang Qiao
- Department of Chemistry, Zhongshan Hospital, and Obstetrics and Gynecology Hospital, Fudan University, Shanghai, 200000, China.
| |
Collapse
|
8
|
Nguyen PT, Seo Y, Ahn JS, Oh SJ, Park HJ, Yu JH, Kim SH, Lee Y, Yang JW, Cho J, Kang MJ, Park JH, Kim HS. De novo interleukin-10 production primed by Lactobacillus sakei CVL-001 amplifies the immunomodulatory abilities of mesenchymal stem cells to alleviate colitis. Biomed Pharmacother 2025; 182:117745. [PMID: 39705909 DOI: 10.1016/j.biopha.2024.117745] [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: 09/26/2024] [Revised: 12/06/2024] [Accepted: 12/08/2024] [Indexed: 12/23/2024] Open
Abstract
Mesenchymal stem cells (MSCs) hold therapeutic promise for treating inflammatory bowel disease (IBD) owing to their immunomodulatory properties. Currently, pre-conditioning strategies with several beneficial agents have been applied to enhance the efficacy of MSCs in treating IBDs. Probiotics are increasingly acknowledged as supplemental therapy for IBD; however, their potential benefits in MSCs-based therapy remain largely unexplored. In this study, we hypothesized that pretreating MSCs with Lactobacillus sakei CVL-001 (L. sakei CVL-001), a representative probiotic strain, could improve their therapeutic effectiveness for IBD. In line with this hypothesis, we noted that pretreatment with L. sakei CVL-001 significantly induced IL-10 secretion in MSCs via the activation of the STAT3 signaling pathway. These primed MSCs reduced pro-inflammatory cytokine production in LPS/IFN-γ-treated macrophages and promoted an M2 phenotype, associated with immunoregulation and tissue repair, in undifferentiated macrophages. In addition, their conditioned media significantly reduced the proliferation capacity of Jurkat T cells and splenocytes, while the neutralization of IL-10 reversed these phenomena. Furthermore, MSCs treated with L. sakei CVL-001 mitigated inflammatory responses and promoted epithelial regeneration, leading to accelerated recovery from disease symptoms and improved survival rates compared to naive MSCs in a DSS-induced colitis mouse model. In conclusion, our findings suggest that probiotics, such as L. sakei CVL-001, can improve the therapeutic efficacy of MSCs for treating IBD.
Collapse
Affiliation(s)
- Phuong Thao Nguyen
- Department of Oral Biochemistry; Dental and Life Science Institute, School of Dentistry, Pusan National University, Yangsan 50612, Republic of Korea; Department of Life Science in Dentistry, School of Dentistry, Pusan National University, Yangsan 50612, Republic of Korea; Education and Research Team for Life Science on Dentistry, Pusan National University, Yangsan 50612, Republic of Korea
| | - Yoojin Seo
- Department of Oral Biochemistry; Dental and Life Science Institute, School of Dentistry, Pusan National University, Yangsan 50612, Republic of Korea
| | - Ji-Su Ahn
- Department of Oral Biochemistry; Dental and Life Science Institute, School of Dentistry, Pusan National University, Yangsan 50612, Republic of Korea
| | - Su-Jeong Oh
- Department of Oral Biochemistry; Dental and Life Science Institute, School of Dentistry, Pusan National University, Yangsan 50612, Republic of Korea; Department of Life Science in Dentistry, School of Dentistry, Pusan National University, Yangsan 50612, Republic of Korea; Education and Research Team for Life Science on Dentistry, Pusan National University, Yangsan 50612, Republic of Korea
| | - Hee-Jeong Park
- Department of Oral Biochemistry; Dental and Life Science Institute, School of Dentistry, Pusan National University, Yangsan 50612, Republic of Korea; Department of Life Science in Dentistry, School of Dentistry, Pusan National University, Yangsan 50612, Republic of Korea; Education and Research Team for Life Science on Dentistry, Pusan National University, Yangsan 50612, Republic of Korea
| | - Jeong Hyun Yu
- Department of Oral Biochemistry; Dental and Life Science Institute, School of Dentistry, Pusan National University, Yangsan 50612, Republic of Korea; Department of Life Science in Dentistry, School of Dentistry, Pusan National University, Yangsan 50612, Republic of Korea; Education and Research Team for Life Science on Dentistry, Pusan National University, Yangsan 50612, Republic of Korea
| | - Seong Hui Kim
- Department of Oral Biochemistry; Dental and Life Science Institute, School of Dentistry, Pusan National University, Yangsan 50612, Republic of Korea; Department of Life Science in Dentistry, School of Dentistry, Pusan National University, Yangsan 50612, Republic of Korea; Education and Research Team for Life Science on Dentistry, Pusan National University, Yangsan 50612, Republic of Korea
| | - Yunji Lee
- Department of Oral Biochemistry; Dental and Life Science Institute, School of Dentistry, Pusan National University, Yangsan 50612, Republic of Korea; Department of Life Science in Dentistry, School of Dentistry, Pusan National University, Yangsan 50612, Republic of Korea; Education and Research Team for Life Science on Dentistry, Pusan National University, Yangsan 50612, Republic of Korea
| | - Ji Won Yang
- Department of Oral Biochemistry; Dental and Life Science Institute, School of Dentistry, Pusan National University, Yangsan 50612, Republic of Korea
| | - Jaejin Cho
- Department of Dental Regenerative Biotechnology, School of Dentistry, Seoul National University, Seoul 03080, Republic of Korea; Dental Research Institute, Seoul National University, Seoul 03080, Republic of Korea
| | - Min-Jung Kang
- Department of Oral Biochemistry; Dental and Life Science Institute, School of Dentistry, Pusan National University, Yangsan 50612, Republic of Korea.
| | - Jong-Hwan Park
- Laboratory Animal Medicine, College of Veterinary Medicine, Animal Medical Institute, Chonnam National University, Gwangju 61186, Republic of Korea; NODCURE, Inc, 77 Yongbong-ro, Buk-gu, Gwangju 61186, Republic of Korea.
| | - Hyung-Sik Kim
- Department of Oral Biochemistry; Dental and Life Science Institute, School of Dentistry, Pusan National University, Yangsan 50612, Republic of Korea; Department of Life Science in Dentistry, School of Dentistry, Pusan National University, Yangsan 50612, Republic of Korea; Education and Research Team for Life Science on Dentistry, Pusan National University, Yangsan 50612, Republic of Korea.
| |
Collapse
|
9
|
Zang J, Yin Z, Ouyang H, Liu Y, Liu Z, Yin Z. Advances in the preparation, applications, challenges, and future trends of polysaccharide-based gels as food-grade delivery systems for probiotics: A review. Compr Rev Food Sci Food Saf 2025; 24:e70111. [PMID: 39865632 DOI: 10.1111/1541-4337.70111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 12/27/2024] [Accepted: 12/29/2024] [Indexed: 01/28/2025]
Abstract
Probiotics are highly regarded for their multiple functions, such as regulating gut health, enhancing the immune system, and preventing chronic diseases. However, their stability in harsh environments and targeted release remain significant challenges. Therefore, exploring effective protection and delivery strategies to ensure targeted release of probiotics is critically important. Polysaccharides, known for their non-toxicity, excellent biocompatibility, and superior biodegradability, show broad prospects in probiotic delivery by forming physical barriers to protect the probiotics. Particularly, polysaccharide-based gels (PBGs), with their unique "spider-web" like structure, capture and ensure the targeted release of probiotics, significantly enhancing their efficacy. This review discusses common polysaccharides used in PBG preparation, their classification and synthesis in food applications, and the advantages of PBGs as probiotic delivery systems. Despite their potential, challenges such as inconsistent gel properties and the need for improved stability remain. Future research should focus on developing novel PBG materials with higher biodegradability and mechanical strength, optimizing the physicochemical properties and cross-linking methods, as well as designing multilayered structures for more precise release control. Additionally, exploring the co-delivery of probiotics with prebiotics, active ingredients, or multi-strain systems could further enhance the efficacy of probiotic delivery.
Collapse
Affiliation(s)
- Jianwei Zang
- Jiangxi Key Laboratory of Natural Products and Functional Foods, College of Food Science and Engineering, Jiangxi Agricultural University, Nanchang, China
| | - Zelin Yin
- Jiangxi Key Laboratory of Natural Products and Functional Foods, College of Food Science and Engineering, Jiangxi Agricultural University, Nanchang, China
| | - Huidan Ouyang
- Nanchang Key Laboratory of Egg Safety Production and Processing Engineering, Jiangxi Agricultural University, Nanchang, China
- Vocational Teachers College, Jiangxi Agricultural University, Nanchang, China
| | - Yuanzhi Liu
- Nanchang Key Laboratory of Egg Safety Production and Processing Engineering, Jiangxi Agricultural University, Nanchang, China
| | - Zebo Liu
- Jiangxi Key Laboratory of Natural Products and Functional Foods, College of Food Science and Engineering, Jiangxi Agricultural University, Nanchang, China
| | - Zhongping Yin
- Jiangxi Key Laboratory of Natural Products and Functional Foods, College of Food Science and Engineering, Jiangxi Agricultural University, Nanchang, China
- Nanchang Key Laboratory of Egg Safety Production and Processing Engineering, Jiangxi Agricultural University, Nanchang, China
| |
Collapse
|
10
|
Duduyemi OP, Potapenko K, Limanska N, Kotsyuda S, Petriv N, Suo H, Gudzenko T, Ivanytsia V, Yevsa T. Lactiplantibacillus plantarum inhibited the growth of primary liver cancer by inducing early apoptosis and senescence, in vitro. Front Microbiol 2024; 15:1451170. [PMID: 39600571 PMCID: PMC11590124 DOI: 10.3389/fmicb.2024.1451170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 08/27/2024] [Indexed: 11/29/2024] Open
Abstract
Primary liver cancer (PLC), comprising hepatocellular carcinoma (HCC) and cholangiocarcinoma (CCA), is a severe form of cancer associated with a high mortality and morbidity rate and increasing incidence worldwide. Current treatment options are limited and chemotherapeutics demonstrate strong side effects. New therapies are highly required. Lactobacilli represent the most diverse lactic acid-producing bacteria group and a prominent example of probiotics. Several studies have highlighted the anticancer efficacy of probiotics, especially of Lactiplantibacillus plantarum. However, there are limited studies on its activity on two PLC types, hepatocellular carcinoma (HCC) and cholangiocarcinoma (CCA). This study evaluated the inhibitory mechanism and properties of L. plantarum ONU 12 (Lp 12) and L. plantarum ONU 355 (Lp 355), isolated from grapes in Ukraine and France, in murine PLC cell lines, in vitro. Strain Lacticaseibacillus casei ATCC 393 (Lc 393) has been taken for a direct comparison, as the most studied probiotic strain. The three Lactobacillus species were used in three forms: as live and heat-killed suspensions, and as sonicated extracts, and tested either as a monotherapy or in combination with standard chemotherapeutics (sorafenib for HCC and gemcitabine for CCA). Cell proliferation and viability were assessed via crystal violet staining assay and cell counting kit-8 assay. The induction of senescence was investigated by senescence-associated β-galactosidase assay. Fluorescence-activated cell sorting analysis was used to determine the apoptotic mechanism behind the inhibitory property of lactobacilli. The results showed that the live suspensions and sonicated extracts of Lp 12, Lp 355, and Lc 393 demonstrated inhibitory properties in CCA and HCC cells after 48 h of incubation. In combinations with standard chemotherapeutics, lactobacilli treatments have shown strong synergistic effects. The combination therapy allowed to reduce the chemotherapeutic doses of gemcitabine from 50 μM to 0.1 and 0.05 μM and sorafenib from 13.8 μM to 6.9 and 3.45 μM. Successful treatment regimes induced early apoptosis and cellular senescence in PLC, as the mechanism of inhibition. Heat-killed suspensions showed no inhibitory effect in none of the cell lines. Both strains, Lp 12 and Lp 355, showed successful results and need further testing in vivo, using autochthonous HCC and CCA models.
Collapse
Affiliation(s)
- Oladimeji Paul Duduyemi
- Department of Gastroenterology, Hepatology, Infectious Diseases, and Endocrinology, Hannover Medical School, Hanover, Germany
| | - Kateryna Potapenko
- Department of Gastroenterology, Hepatology, Infectious Diseases, and Endocrinology, Hannover Medical School, Hanover, Germany
- Department of Microbiology, Virology, and Biotechnology, Odesa I. I. Mechnykov National University, Odesa, Ukraine
| | - Nataliia Limanska
- Department of Gastroenterology, Hepatology, Infectious Diseases, and Endocrinology, Hannover Medical School, Hanover, Germany
- Department of Microbiology, Virology, and Biotechnology, Odesa I. I. Mechnykov National University, Odesa, Ukraine
| | - Sofiya Kotsyuda
- Department of Gastroenterology, Hepatology, Infectious Diseases, and Endocrinology, Hannover Medical School, Hanover, Germany
| | - Nataliia Petriv
- Department of Gastroenterology, Hepatology, Infectious Diseases, and Endocrinology, Hannover Medical School, Hanover, Germany
| | - Huizhen Suo
- Department of Gastroenterology, Hepatology, Infectious Diseases, and Endocrinology, Hannover Medical School, Hanover, Germany
| | - Tetyana Gudzenko
- Department of Microbiology, Virology, and Biotechnology, Odesa I. I. Mechnykov National University, Odesa, Ukraine
| | - Volodymyr Ivanytsia
- Department of Microbiology, Virology, and Biotechnology, Odesa I. I. Mechnykov National University, Odesa, Ukraine
| | - Tetyana Yevsa
- Department of Gastroenterology, Hepatology, Infectious Diseases, and Endocrinology, Hannover Medical School, Hanover, Germany
| |
Collapse
|
11
|
Kang A, Kwak MJ, Choi HJ, Son SH, Lim SH, Eor JY, Song M, Kim MK, Kim JN, Yang J, Lee M, Kang M, Oh S, Kim Y. Integrative Analysis of Probiotic-Mediated Remodeling in Canine Gut Microbiota and Metabolites Using a Fermenter for an Intestinal Microbiota Model. Food Sci Anim Resour 2024; 44:1080-1095. [PMID: 39246539 PMCID: PMC11377207 DOI: 10.5851/kosfa.2024.e41] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 05/10/2024] [Accepted: 05/10/2024] [Indexed: 09/10/2024] Open
Abstract
In contemporary society, the increasing number of pet-owning households has significantly heightened interest in companion animal health, expanding the probiotics market aimed at enhancing pet well-being. Consequently, research into the gut microbiota of companion animals has gained momentum, however, ethical and societal challenges associated with experiments on intelligent and pain-sensitive animals necessitate alternative research methodologies to reduce reliance on live animal testing. To address this need, the Fermenter for Intestinal Microbiota Model (FIMM) is being investigated as an in vitro tool designed to replicate gastrointestinal conditions of living animals, offering a means to study gut microbiota while minimizing animal experimentation. The FIMM system explored interactions between intestinal microbiota and probiotics within a simulated gut environment. Two strains of commercial probiotic bacteria, Enterococcus faecium IDCC 2102 and Bifidobacterium lactis IDCC 4301, along with a newly isolated strain from domestic dogs, Lactobacillus acidophilus SLAM AK001, were introduced into the FIMM system with gut microbiota from a beagle model. Findings highlight the system's capacity to mirror and modulate the gut environment, evidenced by an increase in beneficial bacteria like Lactobacillus and Faecalibacterium and a decrease in the pathogen Clostridium. The study also verified the system's ability to facilitate accurate interactions between probiotics and commensal bacteria, demonstrated by the production of short-chain fatty acids and bacterial metabolites, including amino acids and gamma-aminobutyric acid precursors. Thus, the results advocate for FIMM as an in vitro system that authentically simulates the intestinal environment, presenting a viable alternative for examining gut microbiota and metabolites in companion animals.
Collapse
Affiliation(s)
- Anna Kang
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Science, Seoul National University, Seoul 08826, Korea
| | - Min-Jin Kwak
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Science, Seoul National University, Seoul 08826, Korea
| | - Hye Jin Choi
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Science, Seoul National University, Seoul 08826, Korea
| | - Seon-Hui Son
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Science, Seoul National University, Seoul 08826, Korea
| | - Sei-Hyun Lim
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Science, Seoul National University, Seoul 08826, Korea
| | - Ju Young Eor
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Science, Seoul National University, Seoul 08826, Korea
| | - Minho Song
- Department of Animal Science and Biotechnology, Chungnam National University, Daejeon 34134, Korea
| | - Min Kyu Kim
- Department of Animal Science and Biotechnology, Chungnam National University, Daejeon 34134, Korea
| | - Jong Nam Kim
- Department of Food Science & Nutrition, Dongseo University, Busan 47011, Korea
| | - Jungwoo Yang
- IBS R&D Center, Ildong Bioscience, Pyeongtaek 17957, Korea
| | - Minjee Lee
- IBS R&D Center, Ildong Bioscience, Pyeongtaek 17957, Korea
| | - Minkyoung Kang
- Department of Functional Food and Biotechnology, Jeonju University, Jeonju 55069, Korea
| | - Sangnam Oh
- Department of Functional Food and Biotechnology, Jeonju University, Jeonju 55069, Korea
| | - Younghoon Kim
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Science, Seoul National University, Seoul 08826, Korea
| |
Collapse
|
12
|
Kim YC, Sohn KH, Kang HR. Gut microbiota dysbiosis and its impact on asthma and other lung diseases: potential therapeutic approaches. Korean J Intern Med 2024; 39:746-758. [PMID: 39252487 PMCID: PMC11384250 DOI: 10.3904/kjim.2023.451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2023] [Revised: 03/08/2024] [Accepted: 04/07/2024] [Indexed: 09/11/2024] Open
Abstract
The emerging field of gut-lung axis research has revealed a complex interplay between the gut microbiota and respiratory health, particularly in asthma. This review comprehensively explored the intricate relationship between these two systems, focusing on their influence on immune responses, inflammation, and the pathogenesis of respiratory diseases. Recent studies have demonstrated that gut microbiota dysbiosis can contribute to asthma onset and exacerbation, prompting investigations into therapeutic strategies to correct this imbalance. Probiotics and prebiotics, known for their ability to modulate gut microbial compositions, were discussed as potential interventions to restore immune homeostasis. The impact of antibiotics and metabolites, including short-chain fatty acids produced by the gut microbiota, on immune regulation was examined. Fecal microbiota transplantation has shown promise in various diseases, but its role in respiratory disorders is not established. Innovative approaches, including mucus transplants, inhaled probiotics, and microencapsulation strategies, have been proposed as novel therapeutic avenues. Despite challenges, including the sophisticated adaptability of microbial communities and the need for mechanistic clarity, the potential for microbiota-based interventions is considerable. Collaboration between researchers, clinicians, and other experts is essential to unravel the complexities of the gut-lung axis, paving a way for innovative strategies that could transform the management of respiratory diseases.
Collapse
Affiliation(s)
- Young-Chan Kim
- Division of Allergy and Clinical Immunology, Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
| | - Kyoung-Hee Sohn
- Division of Respiratory, Allergy and Critical Care Medicine, Department of Internal Medicine, Kyung Hee University Hospital, Seoul, Korea
| | - Hye-Ryun Kang
- Division of Allergy and Clinical Immunology, Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
- Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul National University College of Medicine, Seoul, Korea
| |
Collapse
|
13
|
Han C, Manners MT, Robinson SA. Sex differences in opioid response: a role for the gut microbiome? Front Pharmacol 2024; 15:1455416. [PMID: 39268474 PMCID: PMC11390522 DOI: 10.3389/fphar.2024.1455416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 08/14/2024] [Indexed: 09/15/2024] Open
Abstract
Opioid drugs have been long known to induce different responses in males compared to females, however, the molecular mechanisms underlying these effects are yet to be fully characterized. Recent studies have established a link between the gut microbiome and behavioral responses to opioids. Chronic opioid use is associated with gut dysbiosis, or microbiome disruptions, which is thought to contribute to altered opioid analgesia and reward processing. Gut microbiome composition and functioning have also been demonstrated to be influenced by sex hormones. Despite this, there is currently very little work investigating whether sex differences in the gut microbiome mediate sex-dependent responses to opioids, highlighting a critical gap in the literature. Here, we briefly review the supporting evidence implicating a potential role for the gut microbiome in regulating sexually dimorphic opioid response and identify areas for future research.
Collapse
Affiliation(s)
- Caitlin Han
- Department of Psychology, Williams College, Williamstown, MA, United States
| | - Melissa T. Manners
- Department of Biological and Biomedical Sciences, Rowan University, Glassboro, NJ, United States
| | - Shivon A. Robinson
- Department of Psychology, Williams College, Williamstown, MA, United States
| |
Collapse
|
14
|
Abbaszadeh SH, Yousefi M, Arefhosseini SR, Mahmoodpoor A, Mameghani ME. Effect of a seven-strain probiotic on dietary intake, inflammatory markers, and T-cells in severe traumatic brain injury patients: A randomized, double-blind, placebo-controlled trial. Sci Prog 2024; 107:368504241259299. [PMID: 39196597 PMCID: PMC11363228 DOI: 10.1177/00368504241259299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/29/2024]
Abstract
BACKGROUND Inflammatory processes are key factors in pathological events associated with severe traumatic brain injury (STBI). The aim of this trial was to determine the effect of probiotics on anthropometric measures, disease severity, inflammatory markers, and T cells in patients with STBI. METHODS Forty adult patients with STBI were enrolled in this parallel randomized, double-blind, placebo-controlled trial. Energy and protein status, Acute Physiology and Chronic Health Evaluation (APACHE II) score, Sequential Organ Failure Assessment (SOFA), interleukin 10 (IL-10), interleukin 1β (IL-1β), tumor necrosis factor-alpha (TNF-α), transforming growth factor beta (TGF-β), T-helper 17 (Th17), and T- Regulator (T-reg) cells were assessed at baseline (day 1), and week 2 (day 14) for each patient. RESULTS Probiotic supplementation led to a substantial reduction in the serum levels of TNF-α (from 10.15 ± 6.52 to 5.05 ± 3.27) (P = 0.034), IL-1β (from 11.84 ± 7.74 to 5.87 ± 3.77) (P < 0.001), and Th17 cells (from 5.19 ± 1.69 to 2.67 ± 1.89) (P < 0.001) and a substantial increase in the serum levels of IL-10 (from 3.35 ± 1.45 to 6.17 ± 2.04) (P = 0.038), TGF-β (from 30.5 ± 15.27 to 46.25 ± 21.05) (P < 0.001), and T-reg cells (from 2.83 ± 1.43 to 4.29 ± 1.89) (P < 0.001) compared with the placebo group. Furthermore, no notable changes were observed in energy and protein intake and also, terms of SOFA and APACHE II scores following probiotic treatment compared with the placebo. CONCLUSIONS Probiotics could reduce inflammation and improve cellular immunity and may be considered as an adjunctive therapy in STBI patients.
Collapse
Affiliation(s)
- Seyed Hamze Abbaszadeh
- Department of Biochemistry and Diet Therapy, School of Nutrition & Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mehdi Yousefi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Seyed Rafie Arefhosseini
- Department of Biochemistry and Diet Therapy, School of Nutrition & Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ata Mahmoodpoor
- Department of Anesthesiology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mehrangiz Ebrahimi Mameghani
- Department of Biochemistry and Diet Therapy, School of Nutrition & Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
15
|
Anwer M, Wei MQ. Harnessing the power of probiotic strains in functional foods: nutritive, therapeutic, and next-generation challenges. Food Sci Biotechnol 2024; 33:2081-2095. [PMID: 39130669 PMCID: PMC11315846 DOI: 10.1007/s10068-024-01630-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 05/28/2024] [Accepted: 06/03/2024] [Indexed: 08/13/2024] Open
Abstract
Functional foods have become an essential element of the diet in developed nations, due to their health benefits and nutritive values. Such food products are only called functional if they, "In addition to basic nutrition, have valuable effects on one or multiple functions of the human body, thereby enhancing general and physical conditions and/or reducing the risk of disease progression". Functional foods are currently one of the most extensively researched areas in the food and nutrition sciences. They are fortified and improved food products. Presently, probiotics are regarded as the most significant and commonly used functional food product. Diverse probiotic food products and supplements are used according to the evidence that supports their strength, functionality, and recommended dosage. This review provides an overview of the current functional food market, with a particular focus on probiotic microorganisms as pivotal functional ingredients. It offers insights into current research endeavors and outlines potential future directions in the field.
Collapse
Affiliation(s)
- Muneera Anwer
- Menzies Health Institute Queensland and School of Medical Science, Griffith University, Gold Coast Campus, Parklands Drive, Southport, QLD 4215 Australia
- Atta-ur-Rahman School of Applied Biosciences (ASAB), National University of Sciences and Technology (NUST), Islamabad, Pakistan
| | - Ming Q. Wei
- Menzies Health Institute Queensland and School of Medical Science, Griffith University, Gold Coast Campus, Parklands Drive, Southport, QLD 4215 Australia
| |
Collapse
|
16
|
Sagmeister T, Gubensäk N, Buhlheller C, Grininger C, Eder M, Ðordić A, Millán C, Medina A, Murcia PAS, Berni F, Hynönen U, Vejzović D, Damisch E, Kulminskaya N, Petrowitsch L, Oberer M, Palva A, Malanović N, Codée J, Keller W, Usón I, Pavkov-Keller T. The molecular architecture of Lactobacillus S-layer: Assembly and attachment to teichoic acids. Proc Natl Acad Sci U S A 2024; 121:e2401686121. [PMID: 38838019 PMCID: PMC11181022 DOI: 10.1073/pnas.2401686121] [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: 01/26/2024] [Accepted: 04/26/2024] [Indexed: 06/07/2024] Open
Abstract
S-layers are crystalline arrays found on bacterial and archaeal cells. Lactobacillus is a diverse family of bacteria known especially for potential gut health benefits. This study focuses on the S-layer proteins from Lactobacillus acidophilus and Lactobacillus amylovorus common in the mammalian gut. Atomic resolution structures of Lactobacillus S-layer proteins SlpA and SlpX exhibit domain swapping, and the obtained assembly model of the main S-layer protein SlpA aligns well with prior electron microscopy and mutagenesis data. The S-layer's pore size suggests a protective role, with charged areas aiding adhesion. A highly similar domain organization and interaction network are observed across the Lactobacillus genus. Interaction studies revealed conserved binding areas specific for attachment to teichoic acids. The structure of the SlpA S-layer and the suggested incorporation of SlpX as well as its interaction with teichoic acids lay the foundation for deciphering its role in immune responses and for developing effective treatments for a variety of infectious and bacteria-mediated inflammation processes, opening opportunities for targeted engineering of the S-layer or lactobacilli bacteria in general.
Collapse
Affiliation(s)
- Theo Sagmeister
- Institute of Molecular Biosciences, University of Graz, Graz, Austria8010
| | - Nina Gubensäk
- Institute of Molecular Biosciences, University of Graz, Graz, Austria8010
| | | | | | - Markus Eder
- Institute of Molecular Biosciences, University of Graz, Graz, Austria8010
| | - Anđela Ðordić
- Institute of Molecular Biosciences, University of Graz, Graz, Austria8010
| | - Claudia Millán
- Structural Biology Unit, Institute of Molecular Biology of Barcelona, Spanish National Research Council, Barcelona08028, Spain
| | - Ana Medina
- Structural Biology Unit, Institute of Molecular Biology of Barcelona, Spanish National Research Council, Barcelona08028, Spain
| | - Pedro Alejandro Sánchez Murcia
- Laboratory of Computer-Aided Molecular Design, Division of Medicinal Chemistry, Otto-Loewi Research Center, Medical University of Graz, Graz, Austria8010
| | - Francesca Berni
- Department of Bio-Organic Synthesis, Leiden Institute of Chemistry, Leiden University, Leiden2333, The Netherlands
| | - Ulla Hynönen
- Department of Basic Veterinary Sciences, Division of Microbiology and Epidemiology, University of Helsinki, Helsinki00100, Finland
| | - Djenana Vejzović
- Institute of Molecular Biosciences, University of Graz, Graz, Austria8010
| | - Elisabeth Damisch
- Institute of Molecular Biosciences, University of Graz, Graz, Austria8010
| | | | - Lukas Petrowitsch
- Institute of Molecular Biosciences, University of Graz, Graz, Austria8010
| | - Monika Oberer
- Institute of Molecular Biosciences, University of Graz, Graz, Austria8010
- Field of Excellence BioHealth, University of Graz, Graz8010, Austria
- BioTechMed-Graz, University of Graz, Graz8010, Austria
| | - Airi Palva
- Department of Basic Veterinary Sciences, Division of Microbiology and Epidemiology, University of Helsinki, Helsinki00100, Finland
| | - Nermina Malanović
- Institute of Molecular Biosciences, University of Graz, Graz, Austria8010
- Field of Excellence BioHealth, University of Graz, Graz8010, Austria
- BioTechMed-Graz, University of Graz, Graz8010, Austria
| | - Jeroen Codée
- Department of Bio-Organic Synthesis, Leiden Institute of Chemistry, Leiden University, Leiden2333, The Netherlands
| | - Walter Keller
- Institute of Molecular Biosciences, University of Graz, Graz, Austria8010
- Field of Excellence BioHealth, University of Graz, Graz8010, Austria
- BioTechMed-Graz, University of Graz, Graz8010, Austria
| | - Isabel Usón
- Structural Biology Unit, Institute of Molecular Biology of Barcelona, Spanish National Research Council, Barcelona08028, Spain
- Institució Catalana de Recerca i Estudis Avançats, Barcelona08003, Spain
| | - Tea Pavkov-Keller
- Institute of Molecular Biosciences, University of Graz, Graz, Austria8010
- Field of Excellence BioHealth, University of Graz, Graz8010, Austria
- BioTechMed-Graz, University of Graz, Graz8010, Austria
| |
Collapse
|
17
|
Tsuchiya M, Tsuchiya S, Momma H, Ikeda R, Suzuki J, Nagatomi R, Yaegashi N, Arima T, Igarashi K. Impact of Habitual Yogurt Intake in Mother-Child Dyads on Incidence of Childhood Otitis Media: The Japan Environment and Children's Study (JECS). Probiotics Antimicrob Proteins 2024; 16:884-893. [PMID: 37195509 DOI: 10.1007/s12602-023-10086-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/04/2023] [Indexed: 05/18/2023]
Abstract
Otitis media, one of the most common childhood diseases, is characterized by inflammation or infection of the middle ear. Due to their ease of access, daily probiotics are recommended for the prevention of early childhood otitis media. This study aimed to assess the impact of probiotics on the incidence of otitis media using a dataset (n = 95,380) from the Japan Environment and Children's Study, a nationwide birth cohort study. After multiple imputations, the association between the incidence of otitis media in early childhood and the daily frequency of yogurt intake in children and mothers was examined using a generalized linear model after adjusting for several confounders. Repeated incidence of otitis media during the 2 years after birth was found in 14,874 participants (15.6%). Based on participants with the lowest frequency of yogurt intake ("almost never") as the reference group, risk ratios for otitis media incidence decreased with higher frequencies of yogurt intake in children at one year of age, but also independently in mothers during pregnancy. The lowest risk ratio (95% confidence interval) for otitis media incidence at six months of age was observed with the most frequent yogurt intake (once/day or more) (0.54 [0.46-0.63]). Additionally, although a similar association was observed in the subgroup of those with cleft lip and/or palate (CL/P), a high-risk group for severe recurrent otitis media, no statistical significance was observed. Thus, increased regular yogurt intake in both children and mothers was associated with a decrease of otitis media during early childhood.
Collapse
Affiliation(s)
| | - Shinobu Tsuchiya
- Department of Orthodontics and Speech Therapy for Craniofacial Anomalies, Tohoku University Hospital, Sendai, Japan
| | - Haruki Momma
- Department of Medicine and Science in Sports and Exercise, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Ryoukichi Ikeda
- Department of Otolaryngology - Head and Neck Surgery, Iwate Medical School of University, Morioka, Japan
| | - Jun Suzuki
- Department of Otolaryngology - Head and Neck Surgery, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Ryoichi Nagatomi
- Department of Medicine and Science in Sports and Exercise, Tohoku University Graduate School of Medicine, Sendai, Japan
- Division of Biomedical Engineering for Health & Welfare, Tohoku University Graduate School of Biomedical Engineering, Sendai, Japan
| | - Nobuo Yaegashi
- Department of Obstetrics and Gynecology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Takahiro Arima
- Department of Informative Genetics, Environment and Genome Research Center, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Kaoru Igarashi
- Department of Orthodontics and Speech Therapy for Craniofacial Anomalies, Tohoku University Hospital, Sendai, Japan
- Division of Craniofacial Anomalies, Tohoku University Graduate School of Dentistry, Sendai, Japan
| |
Collapse
|
18
|
Wan W, Wu W, Amier Y, Li X, Yang J, Huang Y, Xun Y, Yu X. Engineered microorganisms: A new direction in kidney stone prevention and treatment. Synth Syst Biotechnol 2024; 9:294-303. [PMID: 38510204 PMCID: PMC10950756 DOI: 10.1016/j.synbio.2024.02.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 01/31/2024] [Accepted: 02/20/2024] [Indexed: 03/22/2024] Open
Abstract
Numerous studies have shown that intestinal and urinary tract flora are closely related to the formation of kidney stones. The removal of probiotics represented by lactic acid bacteria and the colonization of pathogenic bacteria can directly or indirectly promote the occurrence of kidney stones. However, currently existing natural probiotics have limitations. Synthetic biology is an emerging discipline in which cells or living organisms are genetically designed and modified to have biological functions that meet human needs, or even create new biological systems, and has now become a research hotspot in various fields. Using synthetic biology approaches of microbial engineering and biological redesign to enable probiotic bacteria to acquire new phenotypes or heterologous protein expression capabilities is an important part of synthetic biology research. Synthetic biology modification of microorganisms in the gut and urinary tract can effectively inhibit the development of kidney stones by a range of means, including direct degradation of metabolites that promote stone production or indirect regulation of flora homeostasis. This article reviews the research status of engineered microorganisms in the prevention and treatment of kidney stones, to provide a new and effective idea for the prevention and treatment of kidney stones.
Collapse
Affiliation(s)
- Wenlong Wan
- Department of Urology, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Weisong Wu
- Department of Urology, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Yirixiatijiang Amier
- Department of Urology, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Xianmiao Li
- Department of Urology, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Junyi Yang
- Department of Urology, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Yisheng Huang
- Department of Urology, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Yang Xun
- Department of Urology, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Xiao Yu
- Department of Urology, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| |
Collapse
|
19
|
Bani Saeid A, De Rubis G, Williams KA, Yeung S, Chellappan DK, Singh SK, Gupta G, Hansbro PM, Shahbazi MA, Gulati M, Kaur IP, Santos HA, Paudel KR, Dua K. Revolutionizing lung health: Exploring the latest breakthroughs and future prospects of synbiotic nanostructures in lung diseases. Chem Biol Interact 2024; 395:111009. [PMID: 38641145 DOI: 10.1016/j.cbi.2024.111009] [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: 03/10/2024] [Revised: 04/04/2024] [Accepted: 04/15/2024] [Indexed: 04/21/2024]
Abstract
The escalating prevalence of lung diseases underscores the need for innovative therapies. Dysbiosis in human body microbiome has emerged as a significant factor in these diseases, indicating a potential role for synbiotics in restoring microbial equilibrium. However, effective delivery of synbiotics to the target site remains challenging. Here, we aim to explore suitable nanoparticles for encapsulating synbiotics tailored for applications in lung diseases. Nanoencapsulation has emerged as a prominent strategy to address the delivery challenges of synbiotics in this context. Through a comprehensive review, we assess the potential of nanoparticles in facilitating synbiotic delivery and their structural adaptability for this purpose. Our review reveals that nanoparticles such as nanocellulose, starch, and chitosan exhibit high potential for synbiotic encapsulation. These offer flexibility in structure design and synthesis, making them promising candidates for addressing delivery challenges in lung diseases. Furthermore, our analysis highlights that synbiotics, when compared to probiotics alone, demonstrate superior anti-inflammatory, antioxidant, antibacterial and anticancer activities. This review underscores the promising role of nanoparticle-encapsulated synbiotics as a targeted and effective therapeutic approach for lung diseases, contributing valuable insights into the potential of nanomedicine in revolutionizing treatment strategies for respiratory conditions, ultimately paving the way for future advancements in this field.
Collapse
Affiliation(s)
- Ayeh Bani Saeid
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Ultimo, NSW, 2007, Australia; Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW, 2007, Australia
| | - Gabriele De Rubis
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Ultimo, NSW, 2007, Australia; Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW, 2007, Australia
| | - Kylie A Williams
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Ultimo, NSW, 2007, Australia
| | - Stewart Yeung
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Ultimo, NSW, 2007, Australia; Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW, 2007, Australia
| | - Dinesh Kumar Chellappan
- Department of Life Sciences, School of Pharmacy, International Medical University, Bukit Jalil, 57000, Kuala Lumpur, Malaysia
| | - Sachin Kumar Singh
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW, 2007, Australia; School of Pharmaceutical Sciences, Lovely Professional University, Jalandhar-Delhi G.T Road, Phagwara, 144411, India
| | - Gaurav Gupta
- School of Pharmacy, Graphic Era Hill University, Dehradun, 248007, India; Department of Clinical Sciences, College of Pharmacy and Health Sciences, Ajman University, Ajman, United Arab Emirates
| | - Philip M Hansbro
- Centre for Inflammation, Faculty of Science, School of Life Sciences, Centenary Institute and University of Technology Sydney, Sydney, NSW, 2007, Australia
| | - Mohammad-Ali Shahbazi
- Department of Biomaterials and Biomedical Technology, University Medical Center Groningen, University of Groningen, AV, 9713, Groningen, the Netherlands
| | - Monica Gulati
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW, 2007, Australia; School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, India
| | - Indu Pal Kaur
- University Institute of Pharmaceutical Sciences, Punjab University Chandigarh, India
| | - Hélder A Santos
- Department of Biomaterials and Biomedical Technology, University Medical Center Groningen, University of Groningen, AV, 9713, Groningen, the Netherlands; Drug Research Program Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, FI-00014, Finland
| | - Keshav Raj Paudel
- Centre for Inflammation, Faculty of Science, School of Life Sciences, Centenary Institute and University of Technology Sydney, Sydney, NSW, 2007, Australia; Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India.
| | - Kamal Dua
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Ultimo, NSW, 2007, Australia; Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW, 2007, Australia; Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India.
| |
Collapse
|
20
|
Govindaraj D, Jensen GB, Rahman Qazi K, Sverremark‐Ekström E, Abrahamsson T, Jenmalm MC. Effects of extremely preterm birth on cytokine and chemokine responses induced by T-cell activation during infancy. Clin Transl Immunology 2024; 13:e1510. [PMID: 38737447 PMCID: PMC11087183 DOI: 10.1002/cti2.1510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 04/17/2024] [Accepted: 04/22/2024] [Indexed: 05/14/2024] Open
Abstract
Objectives Extremely preterm (EPT; gestational week < 28 + 0, < 1000 g) neonates are vulnerable to infections and necrotising enterocolitis, important contributors to mortality and morbidity. However, knowledge regarding their immune maturation remains limited. We here investigated the longitudinal development of functional T-cell capacity in EPT infants. Methods Peripheral blood mononuclear cells were isolated at 14th and 28th day (D) and at gestational week 36 + 0 (Gw36) from EPT infants, participated in a randomised, double-blind, placebo-controlled study of Lactobacillus reuteri DSM 17938 probiotic supplementation. Blood collected from 25 full-term (FT) infants at D14 was used as control. The secretion of immune mediators was determined through comprehensive Luminex panels after stimulation with human T-cell activator CD3/CD28 beads. Results The levels of many mediators were low in EPT infants at D14, whereas the secretion of several chemokines was higher in EPT than in FT infants. Furthermore, Th2:Th1 cytokine ratios were higher in EPT than in FT infants. Progressively elevated secretion of, for example, IFN-γ, TNF and IL-17A in EPT infants was observed from D14 to D28 and then at Gw36. Elevated levels were observed for many proinflammatory mediators at D28. Probiotic supplementation or perinatal factors (e.g. clinical chorioamnionitis, preeclampsia and delivery mode) did not influence the cytokine and chemokine responses. Conclusions Immune mediators induced by T-cell activation in EPT infants were mainly reduced at D14 and Th2 skewed compared to those in FT infants, but mostly recovered at Gw36, indicating immune maturation. Increased proinflammatory responses at D28 may be related to the heightened risk of severe immune-associated complications seen in EPT infants.
Collapse
Affiliation(s)
- Dhanapal Govindaraj
- Division of Inflammation and Infection (II), Department of Biomedical and Clinical Sciences (BKV)Linköping UniversityLinköpingSweden
| | - Georg Bach Jensen
- Division of Inflammation and Infection (II), Department of Biomedical and Clinical Sciences (BKV)Linköping UniversityLinköpingSweden
- Crown Princess Victoria Children's HospitalLinköpingCounty of ÖstergötlandSweden
| | - Khaleda Rahman Qazi
- Department of Molecular Biosciences, The Wenner‐Gren InstituteStockholm UniversityStockholmSweden
| | - Eva Sverremark‐Ekström
- Department of Molecular Biosciences, The Wenner‐Gren InstituteStockholm UniversityStockholmSweden
| | - Thomas Abrahamsson
- Division of Inflammation and Infection (II), Department of Biomedical and Clinical Sciences (BKV)Linköping UniversityLinköpingSweden
- Crown Princess Victoria Children's HospitalLinköpingCounty of ÖstergötlandSweden
| | - Maria C Jenmalm
- Division of Inflammation and Infection (II), Department of Biomedical and Clinical Sciences (BKV)Linköping UniversityLinköpingSweden
| |
Collapse
|
21
|
Rau S, Gregg A, Yaceczko S, Limketkai B. Prebiotics and Probiotics for Gastrointestinal Disorders. Nutrients 2024; 16:778. [PMID: 38542689 PMCID: PMC10975713 DOI: 10.3390/nu16060778] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 03/05/2024] [Accepted: 03/06/2024] [Indexed: 01/03/2025] Open
Abstract
The complex role of the gut microbiome in the pathogenesis of gastrointestinal (GI) disorders is an emerging area of research, and there is considerable interest in understanding how diet can alter the composition and function of the microbiome. Prebiotics and probiotics have been shown to beneficially modulate the gut microbiome, which underlies their potential for benefit in GI conditions. Formulating specific recommendations for the public regarding these dietary supplements has been difficult due to the significant heterogeneity between strains, doses, and duration of treatment investigated across studies, as well as safety concerns with administering live organisms. This review aims to summarize the existing evidence for the use of prebiotics and probiotics in various GI disorders, paying special attention to strain-specific effects that emerged and any adverse effects noted.
Collapse
Affiliation(s)
| | | | | | - Berkeley Limketkai
- Vatche & Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, CA 90095, USA; (S.R.); (A.G.); (S.Y.)
| |
Collapse
|
22
|
Nessim Kostandy E, Suh JH, Tian X, Okeugo B, Rubin E, Shirai S, Luo M, Taylor CM, Kim KH, Rhoads JM, Liu Y. Probiotic Limosilactobacillus reuteri DSM 17938 Changes Foxp3 Deficiency-Induced Dyslipidemia and Chronic Hepatitis in Mice. Nutrients 2024; 16:511. [PMID: 38398835 PMCID: PMC10892585 DOI: 10.3390/nu16040511] [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: 12/30/2023] [Revised: 01/28/2024] [Accepted: 02/07/2024] [Indexed: 02/25/2024] Open
Abstract
The probiotic Limosilactobacillus reuteri DSM 17938 produces anti-inflammatory effects in scurfy (SF) mice, a model characterized by immune dysregulation, polyendocrinopathy, enteropathy, and X-linked inheritance (called IPEX syndrome in humans), caused by regulatory T cell (Treg) deficiency and is due to a Foxp3 gene mutation. Considering the pivotal role of lipids in autoimmune inflammatory processes, we investigated alterations in the relative abundance of lipid profiles in SF mice (± treatment with DSM 17938) compared to normal WT mice. We also examined the correlation between plasma lipids and gut microbiota and circulating inflammatory markers. We noted a significant upregulation of plasma lipids associated with autoimmune disease in SF mice, many of which were downregulated by DSM 17938. The upregulated lipids in SF mice demonstrated a significant correlation with gut bacteria known to be implicated in the pathogenesis of various autoimmune diseases. Chronic hepatitis in SF livers responded to DSM 17938 treatment with a reduction in hepatic inflammation. Altered gene expression associated with lipid metabolism and the positive correlation between lipids and inflammatory cytokines together suggest that autoimmunity leads to dyslipidemia with impaired fatty acid oxidation in SF mice. Probiotics are presumed to contribute to the reduction of lipids by reducing inflammatory pathways.
Collapse
Affiliation(s)
- Erini Nessim Kostandy
- Department of Pediatrics, Division of Gastroenterology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; (E.N.K.); (B.O.)
| | - Ji Ho Suh
- Department of Anesthesiology, Critical Care and Pain Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; (J.H.S.); (K.H.K.)
| | - Xiangjun Tian
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Center, Houston, TX 77030, USA;
| | - Beanna Okeugo
- Department of Pediatrics, Division of Gastroenterology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; (E.N.K.); (B.O.)
| | - Erin Rubin
- Department of Pathology and Laboratory Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; (E.R.); (S.S.)
| | - Sara Shirai
- Department of Pathology and Laboratory Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; (E.R.); (S.S.)
| | - Meng Luo
- Department of Microbiology, Immunology and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA; (M.L.)
| | - Christopher M. Taylor
- Department of Microbiology, Immunology and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA; (M.L.)
| | - Kang Ho Kim
- Department of Anesthesiology, Critical Care and Pain Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; (J.H.S.); (K.H.K.)
| | - J. Marc Rhoads
- Department of Pediatrics, Division of Gastroenterology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; (E.N.K.); (B.O.)
| | - Yuying Liu
- Department of Pediatrics, Division of Gastroenterology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; (E.N.K.); (B.O.)
| |
Collapse
|
23
|
Naeem H, Hassan HU, Shahbaz M, Imran M, Memon AG, Hasnain A, Murtaza S, Alsagaby SA, Al Abdulmonem W, Hussain M, Abdelgawad MA, Ghoneim MM, Al Jbawi E. Role of Probiotics against Human Cancers, Inflammatory Diseases, and Other Complex Malignancies. J Food Biochem 2024; 2024:1-23. [DOI: 10.1155/2024/6632209] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
Abstract
Probiotics have growing medical importance as a result of their potential in the prevention and therapeutic support of several complex diseases, including different types of cancers. The anticarcinogenic properties of probiotics are attributed to various mechanisms, including alterations in the composition of the intestinal microbiota, suppression of cell proliferation, stimulation of apoptosis, inhibition of NF-kB, reduction in levels of H2AX, 8-hydroxy-deoxyguanosine, RIG-I, downregulation of IL-17, and TNF signaling pathway. Furthermore, probiotics have demonstrated significant advantages in the prevention and management of other complex diseases, including diabetes, obesity, and cardiovascular diseases. Probiotics had a considerable effect in reducing inflammatory infiltration and the occurrence of precancerous lesions. Additionally, the administration of probiotics led to a decrease in the appearance level of genes related to proinflammatory pathways, including NF-κB, IL-17, and TNF signaling pathways. However, further research studies are required to comprehend the processes via which probiotics exert their effects and to authenticate their potential as alternative therapeutic interventions.
Collapse
Affiliation(s)
- Hammad Naeem
- Department of Food Science and Technology, Muhammad Nawaz Shareef University of Agriculture, Multan, Pakistan
| | - Hammad Ul Hassan
- Department of Food Science and Technology, Muhammad Nawaz Shareef University of Agriculture, Multan, Pakistan
| | - Muhammad Shahbaz
- Department of Food Science and Technology, Muhammad Nawaz Shareef University of Agriculture, Multan, Pakistan
| | - Muhammad Imran
- Department of Food Science and Technology, University of Narowal, Narowal, Pakistan
| | - Anjuman Gul Memon
- Department of Biochemistry, College of Medicine, Qassim University, Buraydah, Saudi Arabia
| | - Ammarah Hasnain
- Department of Biotechnology, Lahore University of Biological & Applied Sciences, Lahore, Pakistan
| | - Shamas Murtaza
- Department of Food Science and Technology, Muhammad Nawaz Shareef University of Agriculture, Multan, Pakistan
| | - Suliman A. Alsagaby
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Majmaah University, Al-Majmaah 11932, Saudi Arabia
| | - Waleed Al Abdulmonem
- Department of Pathology, College of Medicine, Qassim University, P.O. Box 6655, Buraidah 51452, Saudi Arabia
| | - Muzzamal Hussain
- Department of Food Sciences, Government College University Faisalabad, Faisalabad, Pakistan
| | - Mohamed A. Abdelgawad
- Department of Pharmaceutical Chemistry, College of Pharmacy, Jouf University, Sakaka 72341, Saudi Arabia
| | - Mohammed M. Ghoneim
- Department of Pharmacy Practice, College of Pharmacy, AlMaarefa University, Al Diriyah 13713, Saudi Arabia
| | | |
Collapse
|
24
|
Kadia BM, Allen SJ. Effect of Pre-, Pro-, and Synbiotics on Biomarkers of Systemic Inflammation in Children: A Scoping Review. Nutrients 2024; 16:336. [PMID: 38337621 PMCID: PMC10856957 DOI: 10.3390/nu16030336] [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: 12/22/2023] [Revised: 01/19/2024] [Accepted: 01/19/2024] [Indexed: 02/12/2024] Open
Abstract
Systemic inflammation plays a central role in many diseases and is, therefore, an important therapeutic target. In a scoping review, we assessed the evidence base for the anti-inflammatory effects of pre-, pro-, and synbiotics in children. Of the 1254 clinical trials published in English in Ovid Medline and Cochrane Library PubMed from January 2003 to September 2022, 29 were included in the review. In six studies of healthy children (n = 1552), one reported that fructo-oligosaccharides added to infant formula significantly reduced pro-inflammatory biomarkers, and one study of a single-strain probiotic reported both anti- and pro-inflammatory effects. No effects were seen in the remaining two single-strain studies, one multi-strain probiotic, and one synbiotic study. In 23 studies of children with diseases (n = 1550), prebiotics were tested in 3, single-strain in 16, multi-strain probiotics in 6, and synbiotics in 2 studies. Significantly reduced inflammatory biomarkers were reported in 7/10 studies of atopic/allergic conditions, 3/5 studies of autoimmune diseases, 1/2 studies of preterm infants, 1 study of overweight/obesity, 2/2 studies of severe illness, and 2/3 studies of other diseases. However, only one or two of several biomarkers were often improved; increased pro-inflammatory biomarkers occurred in five of these studies, and a probiotic increased inflammatory biomarkers in a study of newborns with congenital heart disease. The evidence base for the effects of pre-, pro-, and synbiotics on systemic inflammation in children is weak. Further research is needed to determine if anti-inflammatory effects depend on the specific pre-, pro-, and synbiotic preparations, health status, and biomarkers studied.
Collapse
Affiliation(s)
| | - Stephen John Allen
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool L3 5QA, UK;
| |
Collapse
|
25
|
Wiegers C, van Beek EHT, Larsen OFA. Clinical research with probiotics as an indicator of global valorization since the year 2000. Front Microbiol 2023; 14:1323920. [PMID: 38163072 PMCID: PMC10756236 DOI: 10.3389/fmicb.2023.1323920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 11/30/2023] [Indexed: 01/03/2024] Open
Abstract
Probiotics are becoming increasingly popular due to their potential health benefits. With this rise in popularity and demand as indicated by ever-growing market prospects, it seems evident that innovation and valorization are on the rise as well. However, an increasing body of literature shows that innovation is stagnating, which may be detrimental to the exploitation of the benefits of probiotics, for example the development of alternative therapies to manage the increasing prevalence of metabolic and autoimmune disorders. To this end, this study investigated global clinical trials that have been executed since the year 2000 as a first indicator of the status of probiotic valorization. The cumulative number of clinical trials has indeed increased significantly from 0 at the start of the century up to 2,517 registered trials in 2023. However, in Asia, Europe, and North America, the continents with the highest numbers of clinical trials, stagnating or declining trends have been found. In these locations, most clinical trials were funded by non-industry sponsors and targeting probiotic supplements or undefined products. Considering the overall stagnation in clinical trials and viewing these trends in the context of developments in local markets and regulations, the global valorization of probiotics appears to slow down. This could impact the transition from academic research to the development of products that are beneficial and accessible for consumers, either to maintain a healthy lifestyle or to treat medical conditions.
Collapse
Affiliation(s)
- Cato Wiegers
- Athena Institute, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | | | | |
Collapse
|
26
|
Suthar P, Kumar S, Kumar V, Sharma V, Dhiman A. Postbiotics: an exposition on next generation functional food compounds- opportunities and challenges. Crit Rev Food Sci Nutr 2023; 65:1163-1182. [PMID: 38063352 DOI: 10.1080/10408398.2023.2289646] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2025]
Abstract
Consumer's interest for health promoting foods has reshaped the food industry to come up with novel biological compounds with diverse health benefits. Postbiotic are the cell fractions, or cell lysates which have emerged as potential functional food compounds during the last decade. The health benefits of postbiotic are well established while attempts are underway to understand their interaction, production, processing and safety. The review explore the challenges and opportunities to devise better growth mediums, cell lysis and extraction, characterization, stability and applications of postbiotics in both food and pharma industry along with the market trends, success stories and safety concerns regarding postbiotics. The scientific and commercial interest in postbiotic have resulted in extensive investigations and clinical documentation of various physiological benefits and additional bioactivity. The findings validate food and pharma application of the postbiotics and further emphasize on documentation of bioactivity and safety of these compounds.
Collapse
Affiliation(s)
- Priyanka Suthar
- Department of Food Science and Technology, Dr. Y. S. Parmar University of Horticulture and Forestry, Solan, HP, India
| | - Satish Kumar
- Department of Food Science and Technology, Dr. Y. S. Parmar University of Horticulture and Forestry, Solan, HP, India
| | - Vikas Kumar
- Department of Food Science and Technology, Punjab Agricultural University, Ludhiana, Punjab, India
| | - Vishal Sharma
- Faculty of Applied Sciences and Biotechnology, Shoolini University, Solan, HP, India
| | - Atul Dhiman
- Department of Food Science and Technology, Dr. Y. S. Parmar University of Horticulture and Forestry, Solan, HP, India
| |
Collapse
|
27
|
Zhu Z, Yi B, Tang Z, Chen X, Li M, Xu T, Zhao Z, Tang C. Lactobacillus casei combined with Lactobacillus reuteri alleviate pancreatic cancer by inhibiting TLR4 to promote macrophage M1 polarization and regulate gut microbial homeostasis. BMC Cancer 2023; 23:1044. [PMID: 37904102 PMCID: PMC10614400 DOI: 10.1186/s12885-023-11557-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 10/22/2023] [Indexed: 11/01/2023] Open
Abstract
BACKGROUND Pancreatic cancer is a highly lethal disease with no effective treatments. Lactobacillus casei (L. casei) and Lactobacillus reuteri (L. reuteri) exhibited therapeutic effects on several cancers, but their roles in pancreatic cancer are unknown. This study aims to explore how L. casei & L. reuteri influence pancreatic cancer and the underlying mechanisms. METHODS Pancreatic cancer cells were treated with L. casei & L. reuteri and co-cultured with macrophages in a transwell system in vitro. Pancreatic cancer xenograft model was established and L. casei & L. reuteri was used to treat mice in vivo. MTT, CCK-8 assay or immunohistochemical staining were used to determine the proliferation of pancreatic cancer cells or tumor tissues. Transwell assay was applied to test the migration and invasion of pancreatic cells. RT-qPCR was utilized to assess TLR4 and MyD88 expressions in pancreatic cells or tumor tissues. WB, immunofluorescence staining, or flow cytometry was used to evaluate the M1/M2 polarization of macrophages. Besides, the composition of gut microbiota of tumor-bearing mice was determined by 16 S rRNA sequencing, and ultra-high performance liquid chromatography-mass spectrometry (UPLC-MS) untargeted metabolomics was used to evaluate the metabolic profiles of feces. RESULTS L. casei & L. reuteri inhibited the proliferation, migration, invasion of pancreatic cancer cells and pancreatic cancer cell-induced M2 polarization of macrophages by suppressing TLR4. Meanwhile, L. casei & L. reuteri repressed pancreatic cancer growth and promoted M1 macrophage polarization. Besides, L. casei & L. reuteri reduced fecal Alloprevotella and increased fecal azelate and glutamate in nude mice, while TLR4 inhibitor TAK-242 increased Clostridia UCG-014, azelate, uridine, methionine sulfoxide, oxypurinol, and decreased glyceryl monoester in the feces of pancreatic tumor-bearing mice. Fecal oxypurinol and glyceryl monoester levels were positively or negatively associated with gut Clostridia UCG-014 abundance, respectively. CONCLUSION L. casei & L. reuteri alleviate pancreatic cancer by inhibiting TLR4 to promote macrophage M1 polarization and regulate gut microbial homeostasis.
Collapse
Affiliation(s)
- Zemin Zhu
- Department of Hepatobiliary and Pancreatic Surgery, Zhuzhou Central Hospital, Zhuzhou, China
| | - Bo Yi
- Department of Hepatobiliary and Pancreatic Surgery, Zhuzhou Central Hospital, Zhuzhou, China
| | - Zikai Tang
- Department of Hepatobiliary and Pancreatic Surgery, Zhuzhou Central Hospital, Zhuzhou, China
| | - Xun Chen
- Department of Hepatobiliary and Pancreatic Surgery, Zhuzhou Central Hospital, Zhuzhou, China
- Department of Trauma Center, Zhuzhou Central Hospital, Zhuzhou, China
| | - Ming Li
- Department of Trauma Center, Zhuzhou Central Hospital, Zhuzhou, China
| | - Tao Xu
- Department of Hepatobiliary and Pancreatic Surgery, Zhuzhou Central Hospital, Zhuzhou, China
| | - Zhijian Zhao
- Department of Hepatobiliary and Pancreatic Surgery, Zhuzhou Central Hospital, Zhuzhou, China.
| | - Caixi Tang
- Department of Hepatobiliary and Pancreatic Surgery, Zhuzhou Central Hospital, Zhuzhou, China.
- Department of Trauma Center, Zhuzhou Central Hospital, Zhuzhou, China.
| |
Collapse
|
28
|
Magryś A, Pawlik M. Postbiotic Fractions of Probiotics Lactobacillus plantarum 299v and Lactobacillus rhamnosus GG Show Immune-Modulating Effects. Cells 2023; 12:2538. [PMID: 37947616 PMCID: PMC10648844 DOI: 10.3390/cells12212538] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 10/25/2023] [Accepted: 10/27/2023] [Indexed: 11/12/2023] Open
Abstract
Probiotic bacteria belonging to Lactobacillus spp. are important producers of bioactive molecules, known as postbiotics, that play essential roles in the immunological support of the intestinal mucosa. In this study, the system of co-culture of intestinal epithelial cells with macrophage cells in vitro was used to study the potential effect of postbiotic fractions of L. rhamonosus and L. plantarum on the modulation of the immune response induced by pro-inflammatory stimuli. This study's results revealed that the presence of probiotic bacterial components on the mucosal surface in the early and late stage of inflammatory conditions is based on cellular interactions that control inflammation and consequent damage to the intestinal epithelium. In our studies, heat killed fractions of probiotic bacteria and their extracted proteins showed a beneficial effect on controlling inflammation, regardless of the strain tested, consequently protecting intestinal barrier damage. In conclusion, the presented results emphasize that the fractions of probiotic bacteria of L. plantarum and L. rhamnosus may play a significant role in the regulation of LPS-mediated cytotoxic activity in intestinal epithelial cells. The fractions of probiotic strains of L. rhamnosus and L. plantarum showed the potential to suppress inflammation, effectively activating the anti-inflammatory cytokine IL-10 and modulating the IL-18-related response.
Collapse
Affiliation(s)
- Agnieszka Magryś
- Chair and Department of Medical Microbiology, Medical University of Lublin, ul. Chodźki 1, 20-093 Lublin, Poland
| | | |
Collapse
|
29
|
SeyedAlinaghi S, Shahidi R, Afzalian A, Paranjkhoo P, Ghorbanzadeh K, Mojdeganlou H, Razi A, Mojdeganlou P, Dashti M, Ghasemzadeh A, Parikhani SN, Pashaei A, Karimi A, Sepide A, Mehraeen E, Hackett D. Probiotics in prevention and treatment of COVID-19: a systematic review of current evidence. RUSSIAN JOURNAL OF INFECTION AND IMMUNITY 2023; 13:709-722. [DOI: 10.15789/2220-7619-pip-2118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
Abstract
Introduction. Clinical evidence suggests that certain probiotics may help treat and prevent viral infections. To date, the effectiveness of probiotics in the alleviation of COVID-19 has not been established. The aim of this systematic review was to assess the role of probiotics in the prevention and treatment of COVID-19.
Materials and methods. An extensive search of four electronic databases was performed which included Embase, Scopus, Web of Science, and PubMed from November 2019 to June 2022. After reviewing the references list of related articles additional studies were identified. A multiple combination of keywords validated by MESH were used to search the databases. Study selection was performed according to an inclusion and exclusion criteria.
Results. Twenty-three articles met the study inclusion criteria. Six articles were conducted in vitro while the remaining studies were conducted in the human population (in vivo). The type of probiotic was defined in eighteen studies. There were two studies that used supplements (vitamins, herbals, minerals, etc.) in addition to probiotics. The largest sample size was 445 850 participants which were from a study that used an application-based survey. The majority of studies found that probiotics had a positive effect on the COVID-19 disease. The benefits included early remission of COVID-19 symptoms and a shorter duration of sickness (10 studies), lower mortality rates (3 studies), and decreased hospitalization and length of stay (3 studies). Six in vitro studies found that probiotics were beneficial against SARS-CoV-2 through antiviral effects. There were only two studies that found probiotics to be ineffective or caused negative effects when consumed in COVID-19 patients.
Conclusion. Available evidence supports the antiviral role of probiotics on prevention and treatment of COVID-19. The antiviral potential of Lactobacillus paracasei metabolite PlnE and PlnF against SARS-CoV-2 may explain the effectiveness of probiotics on COVID-19.
Collapse
|
30
|
Antony MA, Patel S, Verma V, Kant R. The Role of Gut Microbiome Supplementation in COVID-19 Management. Cureus 2023. [DOI: https:/doi.org/10.7759/cureus.46960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/11/2025] Open
|
31
|
Antony MA, Patel S, Verma V, Kant R. The Role of Gut Microbiome Supplementation in COVID-19 Management. Cureus 2023; 15:e46960. [PMID: 38021562 PMCID: PMC10640765 DOI: 10.7759/cureus.46960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/13/2023] [Indexed: 12/01/2023] Open
Abstract
COVID-19, which is caused by the RNA virus, SARS-CoV-2, mainly affects the respiratory system and has a varied clinical presentation. However, several studies have shown that COVID-19 can also affect the gastrointestinal (GI) system. Patients can experience various GI symptoms, such as vomiting and diarrhea, and the virus has been detected in the stool samples of patients hospitalized with COVID-19. There have also been rare reports of COVID-19 presenting with isolated GI symptoms and lack of respiratory symptoms, and the virus has also been detected for prolonged periods in the fecal samples of COVID-19 patients. Major alterations in the gut microbiome in the form of depletion of beneficial organisms and an abundance of pathogenic organisms have been reported in the fecal samples of hospitalized COVID-19 patients. Although the US FDA has approved several drugs to manage COVID-19, their efficacy remains modest. So, there is a constant ongoing effort to investigate novel treatment options for COVID-19. Health supplements like probiotics, prebiotics, postbiotics, and synbiotics have been popularly known for their various health benefits. In this review, we have summarized the current literature, which shows the potential benefit of these health supplements to mitigate and/or prevent the clinical presentation of COVID-19.
Collapse
Affiliation(s)
- Mc Anto Antony
- Department of Endocrinology, Diabetes and Metabolism, Medical University of South Carolina, Anderson, USA
| | - Siddharth Patel
- Department of Internal Medicine, Decatur Morgan Hospital, Decatur, USA
| | - Vipin Verma
- Department of Internal Medicine, Medical University of South Carolina, Anderson, USA
| | - Ravi Kant
- Department of Endocrinology, Diabetes and Metabolism, Medical University of South Carolina, Anderson, USA
| |
Collapse
|
32
|
Jain M, Stitt G, Son L, Enioutina EY. Probiotics and Their Bioproducts: A Promising Approach for Targeting Methicillin-Resistant Staphylococcus aureus and Vancomycin-Resistant Enterococcus. Microorganisms 2023; 11:2393. [PMID: 37894051 PMCID: PMC10608974 DOI: 10.3390/microorganisms11102393] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 09/16/2023] [Accepted: 09/22/2023] [Indexed: 10/29/2023] Open
Abstract
Antibiotic resistance is a serious global health problem that poses a threat to the successful treatment of various bacterial infections, especially those caused by methicillin-resistant Staphylococcus aureus (MRSA) and vancomycin-resistant Enterococcus (VRE). Conventional treatment of MRSA and VRE infections is challenging and often requires alternative or combination therapies that may have limited efficacy, higher costs, and/or more adverse effects. Therefore, there is an urgent need to find new strategies to combat antibiotic-resistant bacteria. Probiotics and antimicrobial peptides (AMPs) are two promising approaches that have shown potential benefits in various diseases. Probiotics are live microorganisms that confer health benefits to the host when administered in adequate amounts. AMPs, usually produced with probiotic bacteria, are short amino acid sequences that have broad-spectrum activity against bacteria, fungi, viruses, and parasites. Both probiotics and AMPs can modulate the host immune system, inhibit the growth and adhesion of pathogens, disrupt biofilms, and enhance intestinal barrier function. In this paper, we review the current knowledge on the role of probiotics and AMPs in targeting multi-drug-resistant bacteria, with a focus on MRSA and VRE. In addition, we discuss future directions for the clinical use of probiotics.
Collapse
Affiliation(s)
| | | | | | - Elena Y. Enioutina
- Division of Clinical Pharmacology, Department of Pediatrics, Spencer Fox Eccles School of Medicine, University of Utah, Salt Lake City, UT 84108, USA; (M.J.); (G.S.); (L.S.)
| |
Collapse
|
33
|
Das S, Sharma T, Bhardwaj A, Srivastava RK. COVID-19 induced ARDS: immunopathology and therapeutics. EXPLORATION OF IMMUNOLOGY 2023:255-275. [DOI: 10.37349/ei.2023.00101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 06/14/2023] [Indexed: 01/03/2025]
Abstract
The coronavirus disease-2019 (COVID-19) pandemic is a significant threat in the modern era. Clinical studies show that the most common symptom of severe COVID-19 is viral pneumonia-induced acute respiratory distress syndrome (ARDS). The underlying mechanisms by which severe respiratory disease syndrome-coronavirus-2 (SARS-CoV-2) results in ARDS and how certain host factors confer an increased risk of developing severe disease remain unknown. Therefore, identifying the distinctive features of this severe and fatal disease and the therapeutic approaches to COVID-19-induced ARDS remains an immediate need to serve as a basis for best practice models of standardized ARDS treatment. This review article aims to comprehensively discuss the immunopathology of ARDS and provides an overview of the precise role of both the innate and adaptive immune system, with emphasis on the current treatment strategies being tested in the COVID-19-induced ARDS patients. This knowledge will supposedly help in revealing further mechanistic insights into understanding COVID-19-induced ARDS.
Collapse
Affiliation(s)
- Sneha Das
- Translational Immunology, Osteoimmunology & Immunoporosis Lab (TIOIL), Department of Biotechnology, All India Institute of Medical Sciences (AIIMS), New Delhi 110029, India
| | - Tamanna Sharma
- Translational Immunology, Osteoimmunology & Immunoporosis Lab (TIOIL), Department of Biotechnology, All India Institute of Medical Sciences (AIIMS), New Delhi 110029, India
| | - Asha Bhardwaj
- Translational Immunology, Osteoimmunology & Immunoporosis Lab (TIOIL), Department of Biotechnology, All India Institute of Medical Sciences (AIIMS), New Delhi 110029, India
| | - Rupesh K. Srivastava
- Translational Immunology, Osteoimmunology & Immunoporosis Lab (TIOIL), Department of Biotechnology, All India Institute of Medical Sciences (AIIMS), New Delhi 110029, India
| |
Collapse
|
34
|
Zhang X, Zhang N, Wang Z. Eosinophilic esophagitis and esophageal microbiota. Front Cell Infect Microbiol 2023; 13:1206343. [PMID: 37600943 PMCID: PMC10434796 DOI: 10.3389/fcimb.2023.1206343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Accepted: 07/20/2023] [Indexed: 08/22/2023] Open
Abstract
Eosinophilic esophagitis (EoE) is an antigen-mediated chronic inflammatory disease of the esophagus, the prevalence of which has steadily increased in recent years. The pathogenesis of EoE is not yet well-defined; however, recent studies have demonstrated that the esophageal microbiota is an essential regulator of physiological and pathological processes of EoE. Currently, research on EoE and microbiota is an emerging field of study that is receiving increasing attention. Here, we review existing EoE-related esophageal microbiota studies to explore the potential mechanisms underlying esophageal microbiota-mediated EoE. The esophageal microbiome is altered in patients with EoE. Although α diversity is usually not significantly different, an increase in Haemophilus and a decrease in Firmicutes were observed in EoE patients. The role of microbiota in initiating and perpetuating inflammation is not fully understood. Current evidence suggests that the penetration of microbiota leads to the activation of epithelial cells as well as innate and adaptive immune cells, with the subsequent release of cytokines, leading to immune responses and inflammation. The involvement of toll-like receptors in EoE also supports the potential role of the microbiota in the progression of this disease. While EoE-induced inflammation can also lead to alterations in the local microbiome. Moreover, dietary modifications, proton pump inhibitors, and corticosteroids can modulate the esophageal microbiota; however, definitive conclusions about the alterations of microbes after treatment cannot be drawn. These findings provide promising avenues for future studies.
Collapse
Affiliation(s)
- Xiaohan Zhang
- Microbiota Division, Department of Gastroenterology and Hepatology, The First Medical Center, Chinese PLA General Hospital, Beijing, China
- Medical School, Nankai University, Tianjin, China
| | - Nana Zhang
- Microbiota Division, Department of Gastroenterology and Hepatology, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Zikai Wang
- Microbiota Division, Department of Gastroenterology and Hepatology, The First Medical Center, Chinese PLA General Hospital, Beijing, China
- National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
35
|
González-Orozco BD, Kosmerl E, Jiménez-Flores R, Alvarez VB. Enhanced probiotic potential of Lactobacillus kefiranofaciens OSU-BDGOA1 through co-culture with Kluyveromyces marxianus bdgo-ym6. Front Microbiol 2023; 14:1236634. [PMID: 37601389 PMCID: PMC10434783 DOI: 10.3389/fmicb.2023.1236634] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Accepted: 07/21/2023] [Indexed: 08/22/2023] Open
Abstract
Introduction Due to the increasing consumer demand for the development and improvement of functional foods containing probiotics, new probiotic candidates need to be explored as well as novel means to enhance their beneficial effects. Lactobacillus kefiranofaciens OSU-BDGOA1 is a strain isolated from kefir grains that has demonstrated probiotic traits. This species is the main inhabitant of kefir grains and is responsible for the production of an exopolysaccharide (EPS) whit vast technological applications and potential bioactivities. Research has shown that interkingdom interactions of yeast and lactic acid bacteria can enhance metabolic activities and promote resistance to environmental stressors. Methods Comparative genomic analyses were performed to distinguish OSU-BDGOA1 from other strains of the same species, and the genome was mined to provide molecular evidence for relevant probiotic properties. We further assessed the cumulative effect on the probiotic properties of OSU-BDGOA1 and Kluyveromyces marxianus bdgo-ym6 yeast co-culture compared to monocultures. Results Survival during simulated digestion assessed by the INFOGEST digestion model showed higher survival of OSU-BDGOA1 and bdgo-ym6 in co-culture. The adhesion to intestinal cells assessed with the Caco-2 intestinal cell model revealed enhanced adhesion of OSU-BDGOA1 in co-culture. The observed increase in survival during digestion could be associated with the increased production of EPS during the late exponential and early stationary phases of co-culture that, by enhancing co-aggregation between the yeast and the bacterium, protects the microorganisms from severe gastrointestinal conditions as observed by SEM images. Immune modulation and barrier function for recovery and prevention of flagellin-mediated inflammation by Salmonella Typhimurium heat-killed cells (HKSC) in Caco-2 cells were also measured. OSU-BDGOA1 in mono- and co-culture regulated inflammation through downregulation of pro-inflammatory cytokine expression and increased membrane barrier integrity assessed by TEER, FD4 permeability, and expression of tight junctions. Discussion The results of the study warrant further research into the application of co-cultures of yeast and LAB in functional probiotic products and the potential to increase EPS production by co-culture strategies.
Collapse
Affiliation(s)
| | | | | | - Valente B. Alvarez
- Department of Food Science and Technology, The Ohio State University, Columbus, OH, United States
| |
Collapse
|
36
|
Liu Y, Armbrister SA, Okeugo B, Mills TW, Daniel RC, Oh JH, van Pijkeren JP, Park ES, Saleh ZM, Lahiri S, Roos S, Rhoads JM. Probiotic-Derived Ecto-5'-Nucleotidase Produces Anti-Inflammatory Adenosine Metabolites in Treg-Deficient Scurfy Mice. Probiotics Antimicrob Proteins 2023; 15:1001-1013. [PMID: 37178405 PMCID: PMC10926147 DOI: 10.1007/s12602-023-10089-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/04/2023] [Indexed: 05/15/2023]
Abstract
Probiotic Limosilactobacillus reuteri DSM 17938 (DSM 17938) prolongs the survival of Treg-deficient scurfy (SF) mice and reduces multiorgan inflammation by a process requiring adenosine receptor 2A (A2A) on T cells. We hypothesized that L. reuteri-derived ecto-5'-nucleotidase (ecto-5'NT) activity acts to generate adenosine, which may be a central mediator for L. reuteri protection in SF mice. We evaluated DSM 17938-5'NT activity and the associated adenosine and inosine levels in plasma, gut, and liver of SF mice. We examined orally fed DSM 17938, DSM 17938Δ5NT (with a deleted 5'NT gene), and DSM 32846 (BG-R46) (a naturally selected strain derived from DSM 17938). Results showed that DSM 17938 and BG-R46 produced adenosine while "exhausting" AMP, whereas DSM 17938∆5NT did not generate adenosine in culture. Plasma 5'NT activity was increased by DSM 17938 or BG-R46, but not by DSM 17938Δ5NT in SF mice. BG-R46 increased both adenosine and inosine levels in the cecum of SF mice. DSM 17938 increased adenosine levels, whereas BG-R46 increased inosine levels in the liver. DSM 17938Δ5NT did not significantly change the levels of adenosine or inosine in the GI tract or the liver of SF mice. Although regulatory CD73+CD8+ T cells were decreased in spleen and blood of SF mice, these regulatory T cells could be increased by orally feeding DSM 17938 or BG-R46, but not DSM 17938Δ5NT. In conclusion, probiotic-5'NT may be a central mediator of DSM 17938 protection against autoimmunity. Optimal 5'NT activity from various probiotic strains could be beneficial in treating Treg-associated immune disorders in humans.
Collapse
Affiliation(s)
- Yuying Liu
- Department of Pediatrics, Division of Gastroenterology, McGovern Medical School, the University of Texas Health Science Center at Houston, Houston, TX, 77030, USA.
| | - Shabba A Armbrister
- Department of Pediatrics, Division of Gastroenterology, McGovern Medical School, the University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Beanna Okeugo
- Department of Pediatrics, Division of Gastroenterology, McGovern Medical School, the University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Tingting W Mills
- Department of Biochemistry & Molecular Biology, McGovern Medical School, the University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Rhea C Daniel
- Department of Pediatrics, Division of Gastroenterology, McGovern Medical School, the University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Jee-Hwan Oh
- Department of Food Science, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | | | - Evelyn S Park
- Department of Pediatrics, Division of Gastroenterology, McGovern Medical School, the University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Zeina M Saleh
- Department of Pediatrics, Division of Gastroenterology, McGovern Medical School, the University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Sharmistha Lahiri
- Department of Pediatrics, Division of Gastroenterology, McGovern Medical School, the University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Stefan Roos
- Department of Molecular Sciences, Uppsala BioCenter, Swedish University of Agricultural Sciences, Uppsala, Sweden
- BioGaia AB, Stockholm, Sweden
| | - JMarc Rhoads
- Department of Pediatrics, Division of Gastroenterology, McGovern Medical School, the University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| |
Collapse
|
37
|
Darbandi A, Banar M, Koupaei M, Afifirad R, Asadollahi P, Bafandeh E, Rasooli I, Emamie A, Navidifar T, Owlia P. Clinical efficacy of probiotics in prevention of infectious diseases among hospitalized patients in ICU and non-ICU wards in clinical randomized trials: A systematic review. Health Sci Rep 2023; 6:e1469. [PMID: 37547361 PMCID: PMC10400784 DOI: 10.1002/hsr2.1469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 07/13/2023] [Accepted: 07/18/2023] [Indexed: 08/08/2023] Open
Abstract
Background and Aims The present study aimed to review probiotics' clinical efficacy in preventing infectious diseases among hospitalized patients in ICU and non-ICU wards. Methods A search of Medline, EMBASE, The Cochrane Library, Science Direct, Open Grey, and Google Scholar was conducted for eligible publications from 2002 to 2020 following the requirements outlined in the PRISMA guideline. The search strategy was based on the combination of the following terms: "probiotics," "prebiotics," "synbiotics," and "cross-infection." The logical operators "AND" (or the equivalent operator for the databases) and "OR" (e.g., probiotics OR prebiotics OR synbiotics) were used. Results The results indicated that the probiotic consumption caused a significant reduction in antibiotic-associated diarrhea (AAD) and Clostridioides difficile infection (CDI) in 2/8 randomized clinical trials (RCTs) investigating AAD/CDI. Also, 5/12 clinical trials highlighted the considerable effects of probiotics on the reduction or prevention of ventilator associated pneumoniae (VAP), so the mean prevalence of VAP was lower in the probiotic group than in the placebo group. The total rate of nosocomial infections among preterm infants was nonsignificantly higher in the probiotic group compared to the control group. Conclusion This systematic review shows that the administration of probiotics has moderate preventive or mitigating effects on the occurrence of VAP in ICU patients, CDI, AAD, and nosocomial infections among children. Consequently, applying antibiotics along with the proper probiotic species can be advantageous.
Collapse
Affiliation(s)
- Atieh Darbandi
- Molecular Microbiology Research CenterShahed UniversityTehranIran
| | - Maryam Banar
- Department of PathobiologySchool of Public Health, Tehran University of Medical SciencesTehranIran
| | - Maryam Koupaei
- Department of Microbiology and ImmunologySchool of Medicine, Kashan University of Medical SciencesKashanIran
| | - Roghayeh Afifirad
- Department of MicrobiologySchool of Medicine, Tehran University of Medical SciencesTehranIran
| | - Parisa Asadollahi
- Department of MicrobiologyFaculty of Medicine, Ilam University of Medical SciencesIlamIran
| | - Elnaz Bafandeh
- Department of Medical BiotechnologyFaculty of Medicine, Lorestan University of Medical SciencesKhorramabadIran
| | - Iraj Rasooli
- Molecular Microbiology Research Center, Faculty of SciencesShahed UniversityTehranIran
| | - Amir Emamie
- Department of PathobiologySchool of Public Health, Tehran University of Medical SciencesTehranIran
| | | | - Parviz Owlia
- Molecular Microbiology Research CenterShahed UniversityTehranIran
- Molecular Microbiology Research Center, Faculty of SciencesShahed UniversityTehranIran
| |
Collapse
|
38
|
Hao Y, Jiang L, Han D, Si D, Sun Z, Wu Z, Dai Z. Limosilactobacillus mucosae and Lactobacillus amylovorus Protect Against Experimental Colitis via Upregulation of Colonic 5-Hydroxytryptamine Receptor 4 and Transforming Growth Factor-β2. J Nutr 2023; 153:2512-2522. [PMID: 37356501 DOI: 10.1016/j.tjnut.2023.06.031] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 06/16/2023] [Accepted: 06/22/2023] [Indexed: 06/27/2023] Open
Abstract
BACKGROUND Limosilactobacillusmucosae (LM) exerts anti-inflammatory and health-promoting effects. However, its role in the modulation of gut serotonin or 5-hydroxytryptamine (5-HT) metabolism and 5-HT receptors (HTRs) in inflammation requires further investigation. OBJECTIVES We compared LM with Lactobacillus amylovorus (LA) for the regulation of 5-HT, HTRs, inflammatory mediators, and their correlations in the colon of mice with experimental colitis. METHODS Male C57BL/6 mice were randomly assigned to 6 groups: control (Con), LM, LA, dextran sodium sulfate (DSS), and DSS with pre-administration of LM (+LM) or LA (+LA). After 7 d of DSS treatment, mice were killed to analyze the expression of inflammatory mediators, HTRs, and concentrations of 5-HT and microbial metabolites in the colon. RESULTS LM was more effective than LA in alleviating DSS-induced colonic inflammation. Compared with mice in the DSS group, mice receiving DSS + LM or DSS + LA treatment had lower (P < 0.05) colonic mRNA expression of proinflammatory cytokines. DSS + LM treatment had lower mRNA expression of Il1b, Tnfa, and Ccl3, an abundance of p-STAT3, and greater expression of Tgfb2 and Htr4 in the colon (P < 0.05). The expression of inflammatory mediators (including Tgfb-1) was positively correlated (P < 0.05) with 5-HT and Htr2a and negatively correlated (P < 0.05) with Htr4. However, the expression of Tgfb-2 showed reversed correlations with the 5-HT and HTRs described above. Patterns for these correlations were different for LM and LA. Mice receiving the DSS + LM treatment had greater (P < 0.05) concentrations of acetate and valerate and lower (P < 0.05) concentrations of indole-3-acetic acid in the cecal and colonic contents. CONCLUSIONS LM showed greater efficacy than LA in alleviating DSS-induced colonic inflammation. The coordinated regulation of transforming growth factor-β subtypes and serotonin receptors in the colon may be one of the most important mechanisms underlying the probiotic effects of lactobacilli in gut inflammation.
Collapse
Affiliation(s)
- Youling Hao
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Lili Jiang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Dandan Han
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Dayong Si
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Zhiyuan Sun
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Zhenlong Wu
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Zhaolai Dai
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China.
| |
Collapse
|
39
|
Vittoria M, Saggese A, Isticato R, Baccigalupi L, Ricca E. Probiotics as an Alternative to Antibiotics: Genomic and Physiological Characterization of Aerobic Spore Formers from the Human Intestine. Microorganisms 2023; 11:1978. [PMID: 37630538 PMCID: PMC10458579 DOI: 10.3390/microorganisms11081978] [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: 07/07/2023] [Revised: 07/28/2023] [Accepted: 07/30/2023] [Indexed: 08/27/2023] Open
Abstract
A total of thirty-two aerobic spore former strains were isolated from intestinal samples of healthy children and analyzed for their hemolytic and antibiotic-resistant activities. Four strains selected as non-hemolytic and sensitive to all antibiotics recommended as relevant by regulatory agencies were short-listed and evaluated for their in silico and in vitro probiotic potentials. The four selected strains were assigned to the Bacillus velezensis (MV4 and MV11), B. subtilis (MV24), and Priestia megaterium (formerly Bacillus megaterium) (MV30) species. A genomic analysis indicated that MV4, MV11, and MV24 contained a homolog of the gene coding for the fibrinolytic enzyme nattokinase while only MV30 encoded a glutamic acid decarboxylase essential to synthesize the neurotransmitter GABA. All four strains contained gene clusters potentially coding for new antimicrobials, showed strong antioxidant activity, formed biofilm, and produced/secreted quorum-sensing peptides able to induce a cytoprotective stress response in a model of human intestinal (HT-29) cells. Altogether, genomic and physiological data indicate that the analyzed strains do not pose safety concerns and have in vitro probiotic potentials allowing us to propose their use as an alternative to antibiotics.
Collapse
Affiliation(s)
- Maria Vittoria
- Department of Biology, Federico II University of Naples, 80125 Naples, Italy; (M.V.); (A.S.); (R.I.)
| | - Anella Saggese
- Department of Biology, Federico II University of Naples, 80125 Naples, Italy; (M.V.); (A.S.); (R.I.)
| | - Rachele Isticato
- Department of Biology, Federico II University of Naples, 80125 Naples, Italy; (M.V.); (A.S.); (R.I.)
- National Biodiversity Future Center (NBFC), 90133 Palermo, Italy;
| | - Loredana Baccigalupi
- National Biodiversity Future Center (NBFC), 90133 Palermo, Italy;
- Department of Molecular Medicine and Medical Biotechnology, Federico II University of Naples, 80131 Naples, Italy
| | - Ezio Ricca
- Department of Biology, Federico II University of Naples, 80125 Naples, Italy; (M.V.); (A.S.); (R.I.)
| |
Collapse
|
40
|
Batoni G, Kaya E, Catelli E, Quinti S, Botti M, De Carli A, Bianchi M, Maisetta G, Esin S. Lactobacillus Probiotic Strains Differ in Their Ability to Adhere to Human Lung Epithelial Cells and to Prevent Adhesion of Clinical Isolates of Pseudomonas aeruginosa from Cystic Fibrosis Lung. Microorganisms 2023; 11:1707. [PMID: 37512880 PMCID: PMC10385620 DOI: 10.3390/microorganisms11071707] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 06/21/2023] [Accepted: 06/26/2023] [Indexed: 07/30/2023] Open
Abstract
The field of probiotic applications is rapidly expanding, including their use for the control of respiratory tract infections. Nevertheless, probiotics ability to colonize the lung environment and to compete with pulmonary pathogens is still a poorly investigated research area. In this study, we aimed to evaluate the adhesion ability of a number of commercial probiotic strains to the human lung epithelial cell line A549. Furthermore, we assessed probiotic ability to prevent host cell adhesion of one of the major lung pathogens in cystic fibrosis, Pseudomonas aeruginosa, and to reduce the pathogen-induced inflammatory response of human peripheral blood mononuclear cells (PBMCs) in terms of cytokine release. Lactobacillus acidophilus displayed the highest adhesion ability to A549 cells evaluated as percent of adhered bacteria compared to the inoculum. In agreement with such an observation, L. acidophilus was the most efficient in preventing adhesion to A549 cells of a P. aeruginosa isolate from CF sputum. Three-color fluorescence labeling of A549 cells, P. aeruginosa, and L. acidophilus, and confocal microcopy image analyses revealed a likely exclusion effect played by both live and UV-killed L. acidophilus towards P. aeruginosa. Such results were confirmed by CFU count. When co-cultured with PBMCs, both live and UV-killed L. acidophilus reduced the amount of IL-1β and IL-6 in culture supernatants in a statistically significant manner. Overall, the results obtained point to L. acidophilus as an interesting candidate for further studies for a potential aerogenous administration to control P. aeruginosa infections.
Collapse
Affiliation(s)
- Giovanna Batoni
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56123 Pisa, Italy
| | - Esingül Kaya
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56123 Pisa, Italy
| | - Elisa Catelli
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56123 Pisa, Italy
| | - Sabrina Quinti
- Cystic Fibrosis Supporting Service, Azienda USL Toscana Nord-Ovest, 57128 Livorno, Italy
| | - Matteo Botti
- Cystic Fibrosis Supporting Service, Azienda USL Toscana Nord-Ovest, 57128 Livorno, Italy
| | - Alessandro De Carli
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56123 Pisa, Italy
- Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy
| | - Marta Bianchi
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56123 Pisa, Italy
| | - Giuseppantonio Maisetta
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56123 Pisa, Italy
| | - Semih Esin
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56123 Pisa, Italy
| |
Collapse
|
41
|
Halemani K, Shetty AP, Thimmappa L, Issac A, Dhiraaj S, Radha K, Mishra P, Mathias EG. Impact of probiotic on anxiety and depression symptoms in pregnant and lactating women and microbiota of infants: A systematic review and meta-analysis. J Glob Health 2023; 13:04038. [PMID: 37218177 PMCID: PMC10173681 DOI: 10.7189/jogh.13.04038] [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: 05/24/2023] Open
Abstract
Background Probiotics are non-invasive therapies composed of live bacteria and yeast. Administration of prebiotics improved the health status of pregnant and lactating women, as well as newborns. This review aimed to appraise the evidence concerning the effectiveness of probiotics on the mental health of pregnant women, lactating mother and the microbiota of the newborn. Methods This systematic review and meta-analysis ascertained quantitative studies published in Medline (PubMed), Clinical Key, EMBASE, Cumulative Index to Nursing and Allied Health Literature (CINAHL), the Cochrane Library, and Google scholar. Two authors independently screened and extracted the data from the primary studies that analysed the efficacy of probiotics on the mental health of pregnant and lactating women and the microbiota of the newborn. We adopted Cochrane Collaboration guidelines and reported using the Preferred Reporting Items for Systematic review and Meta-Analysis (PRISMA) statement. The qualities of included trials were assessed by Cochrane collaboration's risk of bias tool (ROB-2). Results Sixteen trials comprised 946 pregnant women, 524 were lactating mothers, and 1678 were infants. The sample size of primary studies ranged from 36 to 433. Probiotics were administered as interventions, using either a single strain of Bifidobacterium or Lactobacillus or a double-strain combination of Lactobacillus and Bifidobacterium. Probiotics supplementation reduced anxiety in pregnant (n = 676, standardised mean difference (SMD) = 0.01; 95% confidence interval (CI) = -0.28,0.30, P = 0.04, I2 = 70) and lactating women (n = 514, SMD = -0.17; 95% CI = -1.62,1.27, P = 0.98, I2 = 0). Similarly, probiotics decreased depression in pregnant (n = 298, SMD = 0.05; 95% CI = -0.24,0.35, P = 0.20, I2 = 40) and lactating women (n = 518, SMD = -0.10; 95% CI = -1.29,-1.05, P = 0.11, I2 = 60%). Similarly, probiotics supplementation improved the gut microbiota and reduced the duration of crying, abdominal distension, abdominal colic and diarrhoea. Conclusion Non-invasive probiotic therapies are more useful to pregnant and lactating women and newborns. Registration The review protocol was registered with PROSPERO (CRD42022372126).
Collapse
Affiliation(s)
- Kurvatteppa Halemani
- Sanjay Gandhi Post Graduate Institute of Medical Sciences, Lucknow, Uttar Pradesh, India
| | - Asha P Shetty
- All India Institute of Medical Sciences, Bhubaneswar, Odisha, India
| | - Latha Thimmappa
- All India Institute of Medical Sciences, Kalyani, West- Bengal, India
| | - Alwin Issac
- All India Institute of Medical Sciences, Bhubaneswar, Odisha, India
| | - Sanjay Dhiraaj
- Sanjay Gandhi Post Graduate Institute of Medical Sciences, Lucknow, Uttar Pradesh, India
| | - K Radha
- Sanjay Gandhi Post Graduate Institute of Medical Sciences, Lucknow, Uttar Pradesh, India
| | - Prabhaker Mishra
- Sanjay Gandhi Post Graduate Institute of Medical Sciences, Lucknow, Uttar Pradesh, India
| | | |
Collapse
|
42
|
Freguia CF, Pascual DW, Fanger GR. Sjögren's Syndrome Treatments in the Microbiome Era. ADVANCES IN GERIATRIC MEDICINE AND RESEARCH 2023; 5:e230004. [PMID: 37323129 PMCID: PMC10270702 DOI: 10.20900/agmr20230004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Sjögren's syndrome (SS) is a chronic autoimmune disease characterized by inflammatory cell infiltration of the salivary and lacrimal glands, resulting in acinar epithelial cell atrophy, cell death, and loss of exocrine function. At least half of SS patients develop extraglandular inflammatory disease and have a wide range of systemic clinical manifestations that can affect any organ system, including connective tissues. As many as 3.1 million people in the U.S. suffer from SS, a disease that causes severe impairment. Women are nine times more likely than men to be affected by this condition. Unfortunately, there is currently no effective treatment for SS, and the available options only provide partial relief. Treatment involves using replacement therapies such as artificial saliva and eye lubricants, or immunosuppressive agents that have limited efficacy. The medical community recognizes that there is a significant need for more effective treatments for SS. Increasing evidence demonstrates the links between the dysfunction of the human microbial community and the onset and development of many human diseases, signifying the potential use of microorganisms as an alternative strategy to conquer these issues. The role of the microbiome in controlling immune function of the human host in the context of autoimmune diseases like SS is now becoming better understood and may help to enable new drug development strategies. Natural probiotics and synthetic biology applications hold promise for novel treatment approaches to solve the encryption of many complex and multifactorial immune disorders, like SS.
Collapse
Affiliation(s)
| | - David W. Pascual
- Department of Infectious Diseases & Immunology, College of Veterinary Medicine, University of Florida, P.O. Box 110880, Gainesville, FL 32611, USA
| | - Gary R. Fanger
- Rise Therapeutics, 1405 Research Blvd., Rockville, MD 20850, USA
| |
Collapse
|
43
|
Liu Y, Armbrister SA, Okeugo B, Mills TW, Daniel RC, Oh JH, Pijkeren JP, Park ES, Saleh ZM, Lahiri S, Roos S, Rhoads JM. Probiotic-derived ecto-5'-nucleotidase produces anti-inflammatory adenosine metabolites in Treg-deficient scurfy mice. RESEARCH SQUARE 2023:rs.3.rs-2781715. [PMID: 37066419 PMCID: PMC10104250 DOI: 10.21203/rs.3.rs-2781715/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/18/2023]
Abstract
Probiotic Limosilactobacillus reuteri DSM 17938 (DSM 17938) prolonges the survival of Treg-deficient scurfy (SF) mice and reduces multiorgan inflammation by a process requiring adenosine receptor 2A (A 2A ) on T cells. We hypothesized that L. reuteri -derived ecto-5'-nucleotidase (ecto-5'NT) activity acts to generate adenosine, which may be a central mediator for L. reuteri protection in SF mice. We evaluated DSM 17938-5'NT activity and the associated adenosine and inosine levels in plasma, gut and liver of SF mice. We examined orally fed DSM 17938, DSM 17938Δ5NT (with a deleted 5'NT gene), and DSM 32846 (BG-R46) (a naturally selected strain derived from DSM 17938). Results showed that DSM 17938 and BG-R46 produced adenosine while "exhausting" AMP, whereas DSM 17938∆5NT did not generate adenosine in culture. Plasma 5'NT activity was increased by DSM 17938 or BG-R46, but not by DSM 17938Δ5NT in SF mice. BG-R46 increased both adenosine and inosine levels in the cecum of SF mice. DSM 17938 increased adenosine levels, whereas BG-R46 increased inosine levels in the liver. DSM 17938Δ5NT did not significantly change the levels of adenosine or inosine in the GI tract or the liver of SF mice. Although regulatory CD73 + CD8 + T cells were decreased in spleen and blood of SF mice, these regulatory T cells could be increased by orally feeding DSM 17938 or BG-R46, but not DSM 17938Δ5NT. In conclusion, probiotic-5'NT may be a central mediator of DSM 17938 protection against autoimmunity. Optimal 5'NT activity from various probiotic strains could be beneficial in treating Treg-associated immune disorders in humans.
Collapse
|
44
|
Sharma M, Dhaliwal M, Tyagi R, Goyal T, Sharma S, Rawat A. Microbiome and Its Dysbiosis in Inborn Errors of Immunity. Pathogens 2023; 12:pathogens12040518. [PMID: 37111404 PMCID: PMC10145396 DOI: 10.3390/pathogens12040518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 03/20/2023] [Accepted: 03/24/2023] [Indexed: 03/29/2023] Open
Abstract
Inborn errors of immunity (IEI) can present with infections, autoimmunity, lymphoproliferation, granulomas, and malignancy. IEIs are due to genetic abnormalities that disrupt normal host-immune response or immune regulation. The microbiome appears essential for maintaining host immunity, especially in patients with a defective immune system. Altered gut microbiota in patients with IEI can lead to clinical symptoms. Microbial dysbiosis is the consequence of an increase in pro-inflammatory bacteria or a reduction in anti-inflammatory bacteria. However, functional and compositional differences in microbiota are also involved. Dysbiosis and a reduced alpha-diversity are well documented, particularly in conditions like common variable immunodeficiency. Deranged microbiota is also seen in Wiskott–Aldrich syndrome, severe combined immunodeficiency, chronic granulomatous disease, selective immunoglobulin-A deficiency, Hyper IgE syndrome (HIGES), X-linked lymphoproliferative disease-2, immunodysregulation, polyendocrinopathy, enteropathy, x-linked syndrome, and defects of IL10 signalling. Distinct gastrointestinal, respiratory, and cutaneous symptoms linked to dysbiosis are seen in several IEIs, emphasizing the importance of microbiome identification. In this study, we discuss the processes that maintain immunological homeostasis between commensals and the host and the disruptions thereof in patients with IEIs. As the connection between microbiota, host immunity, and infectious illnesses is better understood, microbiota manipulation as a treatment strategy or infection prevention method would be more readily employed. Therefore, optimal prebiotics, probiotics, postbiotics, and fecal microbial transplantation can be promising strategies to restore the microbiota and decrease disease pathology in patients with IEIs.
Collapse
|
45
|
Liu Y, Hoang TK, Park ES, Freeborn J, Okeugo B, Tran DQ, Rhoads JM. Probiotic-educated Tregs are more potent than naïve Tregs for immune tolerance in stressed new-born mice. Benef Microbes 2023; 14:73-84. [PMID: 36815493 PMCID: PMC10124588 DOI: 10.3920/bm2022.0095] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/24/2023]
Abstract
When new-born mice are subjected to acute maternal separation stress, cow-milk based formula feeding, and brief recurrent hypoxia with cold stress, they develop gut inflammation similar to the phenotype of neonatal necrotizing enterocolitis, characterised by an increase in gut mucosal effector T (Teffs) and reduced Foxp3+ regulatory T (Tregs) cells. The imbalance can be prevented by probiotic Limosilactobacillus reuteri DSM 17938 (LR 17938). We hypothesised that LR 17938 could potentiate a tolerogenic function of Tregs. To analyse whether LR 17938 can educate Tregs to improve their tolerogenic potency during neonatal stress, we isolated T cells (Tregs and Teffs) from 'donor' mice fed with either LR 17938 (107 cfu) or control media. The cells were adoptively transferred (AT) by intraperitoneal injection (5 × 105 cells/mouse) to new-born (d5) recipient mice. Mice were then separated from their dams, fed formula by gavage, and exposed to hypoxia and cold stress (NeoStress) for 4 days. We analysed the percentage of Tregs in CD4+T helper cells in the intestine (INT) and mesenteric lymph nodes (MLN) of recipient mice. We found that: (1) the percentage of Tregs in the INT and MLN following NeoStress were significantly reduced compared to dam-fed unstressed mice; (2) AT of either naïve Tregs or LR-educated Tregs to mice with Neostress increased the percentage of Tregs in the INT and MLN compared to the percentage in NeoStress mice without Treg treatment; however, LR-educated Tregs increased the Tregs significantly more than naïve Tregs; and (3) AT of LR-educated Tregs reduced pro-inflammatory CD44+Foxp3-NonTregs and inflammatory CX3CR1+ dendritic cells in the intestinal mucosa of NeoStress mice. In conclusion, adoptive transfer of Tregs promotes the generation of and/or migration of endogenous Tregs in the intestinal mucosa of recipient mice. Importantly, probiotic-educated Tregs are more potent than naïve Tregs to enhance immune tolerance following neonatal stress.
Collapse
Affiliation(s)
- Y Liu
- Department of Pediatrics, Division of Gastroenterology, McGovern Medical School, University of Texas Health Science Center at Houston, 6431 Fannin Street, MSB 3.137, Houston, TX 77030, USA
| | - T K Hoang
- Department of Pediatrics, Division of Gastroenterology, McGovern Medical School, University of Texas Health Science Center at Houston, 6431 Fannin Street, MSB 3.137, Houston, TX 77030, USA
| | - E S Park
- Department of Pediatrics, Division of Gastroenterology, McGovern Medical School, University of Texas Health Science Center at Houston, 6431 Fannin Street, MSB 3.137, Houston, TX 77030, USA
| | - J Freeborn
- Department of Pediatrics, Division of Gastroenterology, McGovern Medical School, University of Texas Health Science Center at Houston, 6431 Fannin Street, MSB 3.137, Houston, TX 77030, USA
| | - B Okeugo
- Department of Pediatrics, Division of Gastroenterology, McGovern Medical School, University of Texas Health Science Center at Houston, 6431 Fannin Street, MSB 3.137, Houston, TX 77030, USA
| | - D Q Tran
- Department of Pediatrics, Division of Gastroenterology, McGovern Medical School, University of Texas Health Science Center at Houston, 6431 Fannin Street, MSB 3.137, Houston, TX 77030, USA
| | - J M Rhoads
- Department of Pediatrics, Division of Gastroenterology, McGovern Medical School, University of Texas Health Science Center at Houston, 6431 Fannin Street, MSB 3.137, Houston, TX 77030, USA
| |
Collapse
|
46
|
Liu X, Qin Y, Dong L, Han Z, Liu T, Tang Y, Yu Y, Ye J, Tao J, Zeng X, Feng J, Zhang XZ. Living symbiotic bacteria-involved skin dressing to combat indigenous pathogens for microbiome-based biotherapy toward atopic dermatitis. Bioact Mater 2023; 21:253-266. [PMID: 36157249 PMCID: PMC9477860 DOI: 10.1016/j.bioactmat.2022.08.019] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 08/08/2022] [Accepted: 08/23/2022] [Indexed: 11/12/2022] Open
Abstract
Many skin diseases, such as atopic dermatitis (AD), are featured with the dysbiosis of skin microbiota. The clinically recommended options for AD treatments suffer from poor outcomes and high side-effects, leading to severe quality-of-life impairment. To deal with this long-term challenge, we develop a living bacterial formulation (Hy@Rm) that integrates skin symbiotic bacteria of Roseomonas mucosa with poly(vinyl pyrrolidone), poly(vinyl alcohol) and sodium alginate into a skin dressing by virtue of the Ca2+-mediated cross-linking and the freezing-thawing (F-T) cycle method. Hy@Rm dressing creates a favorable condition to not only serve as extrinsic culture harbors but also as nutrient suppliers to support R. mucosa survival in the harsh microenvironment of AD sites to defeat S. aureus, which predominantly colonizes AD skins as an indigenous pathogen, mainly through the secretion of sphingolipids metabolites by R. mucosa like a therapeutics bio-factory. Meanwhile, this elaborately designed skin dressing could accelerate wound healing, normalize aberrant skin characters, recover skin barrier functions, alleviate AD-associated immune/inflammation responses, functioning like a combinational therapy. This study offers a promising means for the topical bacteria transplant to realize effective microbe biotherapy toward the skin diseases feature with microbe milieu disorders, including but not limited to AD disease. Symbiotic bacteria were employed to defeat pathogenic bacteria in the diseased skins to treat atopic dermatitis (AD). A living symbiotic bacteria-involved skin dressing was designed to serve as extrinsic culture harbors and nutrient suppliers to support R. mucosa survival in the harsh microenvironment to defeat S. aureus, which predominantly colonizes AD skins. This study offered a promising means for the topical bacteria transplant to realize effective microbiome-based biotherapy toward the skin diseases feature with microbe milieu disorders.
Collapse
|
47
|
Akram N, Saeed F, Afzaal M, Shah YA, Qamar A, Faisal Z, Ghani S, Ateeq H, Akhtar MN, Tufail T, Hussain M, Asghar A, Rasheed A, Jbawi EA. Gut microbiota and synbiotic foods: Unveiling the relationship in COVID-19 perspective. Food Sci Nutr 2023; 11:1166-1177. [PMID: 36911846 PMCID: PMC10002946 DOI: 10.1002/fsn3.3162] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 11/14/2022] [Accepted: 11/18/2022] [Indexed: 01/13/2023] Open
Abstract
The Coronavirus disease 2019 (COVID-19) has spread across the globe and is causing widespread disaster. The impact of gut microbiota on lung disease has been widely documented. Diet, environment, and genetics all play a role in shaping the gut microbiota, which can influence the immune system. Improving the gut microbiota profile through customized diet, nutrition, and supplementation has been shown to boost immunity, which could be one of the preventative methods for reducing the impact of various diseases. Poor nutritional status is frequently linked to inflammation and oxidative stress, both of which can affect the immune system. This review emphasizes the necessity of maintaining an adequate level of important nutrients to effectively minimize inflammation and oxidative stress, moreover to strengthen the immune system during the COVID-19 severity. Furthermore, the purpose of this review is to present information and viewpoints on the use of probiotics, prebiotics, and synbiotics as adjuvants for microbiota modification and its effects on COVID-19 prevention and treatment.
Collapse
Affiliation(s)
- Noor Akram
- Department of Food and NutritionGovernment College UniversityFaisalabadPakistan
| | - Farhan Saeed
- Department of Food ScienceGovernment College UniversityFaisalabadPakistan
| | - Muhammad Afzaal
- Department of Food ScienceGovernment College UniversityFaisalabadPakistan
| | - Yasir Abbas Shah
- Department of Food ScienceGovernment College UniversityFaisalabadPakistan
| | - Aiza Qamar
- Department of Nutrition and Health PromotionUniversity of Home Economics LahoreLahorePakistan
| | - Zargham Faisal
- Institute of Food Science and NutritionBahauddin Zakariya University MultanMultanPakistan
| | - Samia Ghani
- Faculty of Pharmaceutical SciencesGovernment College University FaisalabadPunjabPakistan
| | - Huda Ateeq
- Department of Food ScienceGovernment College UniversityFaisalabadPakistan
| | - Muhammad Nadeem Akhtar
- University Institute of Diet and Nutritional SciencesThe University of LahoreLahorePakistan
| | - Tabassum Tufail
- University Institute of Diet and Nutritional SciencesThe University of LahoreLahorePakistan
| | - Muzzamal Hussain
- Department of Food ScienceGovernment College UniversityFaisalabadPakistan
| | - Aasma Asghar
- Department of Food ScienceGovernment College UniversityFaisalabadPakistan
| | - Ammara Rasheed
- Department of Food and NutritionGovernment College UniversityFaisalabadPakistan
| | | |
Collapse
|
48
|
Multi-dimensional experimental and computational exploration of metabolism pinpoints complex probiotic interactions. Metab Eng 2023; 76:120-132. [PMID: 36720400 DOI: 10.1016/j.ymben.2023.01.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Revised: 12/13/2022] [Accepted: 01/21/2023] [Indexed: 01/29/2023]
Abstract
Multi-strain probiotics are widely regarded as effective products for improving gut microbiota stability and host health, providing advantages over single-strain probiotics. However, in general, it is unclear to what extent different strains would cooperate or compete for resources, and how the establishment of a common biofilm microenvironment could influence their interactions. In this work, we develop an integrative experimental and computational approach to comprehensively assess the metabolic functionality and interactions of probiotics across growth conditions. Our approach combines co-culture assays with genome-scale modelling of metabolism and multivariate data analysis, thus exploiting complementary data- and knowledge-driven systems biology techniques. To show the advantages of the proposed approach, we apply it to the study of the interactions between two widely used probiotic strains of Lactobacillus reuteri and Saccharomyces boulardii, characterising their production potential for compounds that can be beneficial to human health. Our results show that these strains can establish a mixed cooperative-antagonistic interaction best explained by competition for shared resources, with an increased individual exchange but an often decreased net production of amino acids and short-chain fatty acids. Overall, our work provides a strategy that can be used to explore microbial metabolic fingerprints of biotechnological interest, capable of capturing multifaceted equilibria even in simple microbial consortia.
Collapse
|
49
|
Pramanik S, Venkatraman S, Karthik P, Vaidyanathan VK. A systematic review on selection characterization and implementation of probiotics in human health. Food Sci Biotechnol 2023; 32:423-440. [PMID: 36911328 PMCID: PMC9992678 DOI: 10.1007/s10068-022-01210-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Revised: 10/29/2022] [Accepted: 11/14/2022] [Indexed: 01/12/2023] Open
Abstract
Probiotics are live bacteria found in food that assist the body's defence mechanisms against pathogens by reconciling the gut microbiota. Probiotics are believed to aid with gut health, the immune system, and brain function, among other factors. They've furthermore been shown to help with constipation, high blood pressure, and skin issues. The global probiotics market has been incrementally growing in recent years, as consumers' demand for healthy diets and wellness has continued to increase. This has prompted the food industry to develop new probiotic-containing food products, as well as researchers to explore their specific characteristics and impacts on human health. Although most probiotics are fastidious microorganisms that are nutritionally demanding and sensitive to environmental conditions, they become less viable as they are processed and stored. In this review we studied the current literature on the fundamental idea of probiotic bacteria, their medical benefits, and their selection, characterization, and implementations. Graphical Abstract
Collapse
Affiliation(s)
- Shreyasi Pramanik
- Integrated Bioprocessing Laboratory, Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology (SRM IST), 603 203, Kattankulathur, India
| | - Swethaa Venkatraman
- Integrated Bioprocessing Laboratory, Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology (SRM IST), 603 203, Kattankulathur, India
| | - Pothiyappan Karthik
- Department of Food Biotechnology, Karpagam Academic of Higher Education, Coimbatore, India
| | - Vinoth Kumar Vaidyanathan
- Integrated Bioprocessing Laboratory, Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology (SRM IST), 603 203, Kattankulathur, India
| |
Collapse
|
50
|
Tiwari P, Dwivedi R, Bansal M, Tripathi M, Dada R. Role of Gut Microbiota in Neurological Disorders and Its Therapeutic Significance. J Clin Med 2023; 12:1650. [PMID: 36836185 PMCID: PMC9965848 DOI: 10.3390/jcm12041650] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 02/14/2023] [Accepted: 02/17/2023] [Indexed: 02/22/2023] Open
Abstract
In humans, the gut microbiota (GM) are known to play a significant role in the metabolism of nutrients and drugs, immunomodulation, and pathogen defense by inhabiting the gastrointestinal tract (GIT). The role of the GM in the gut-brain axis (GBA) has been documented for different regulatory mechanisms and associated pathways and it shows different behaviors with individualized bacteria. In addition, the GM are known as susceptibility factor for neurological disorders in the central nervous system (CNS), regulating disease progression and being amenable to intervention. Bidirectional transmission between the brain and the GM occurs in the GBA, implying that it performs a significant role in neurocrine, endocrine, and immune-mediated signaling pathways. The GM regulates multiple neurological disorders by supplementing them with prebiotics, probiotics, postbiotics, synbiotics, fecal transplantations, and/or antibiotics. A well-balanced diet is critically important for establishing healthy GM, which can alter the enteric nervous system (ENS) and regulate multiple neurological disorders. Here, we have discussed the function of the GM in the GBA from the gut to the brain and the brain to the gut, the pathways associated with neurology that interacts with the GM, and the various neurological disorders associated with the GM. Furthermore, we have highlighted the recent advances and future prospects of the GBA, which may require addressing research concerns about GM and associated neurological disorders.
Collapse
Affiliation(s)
- Prabhakar Tiwari
- Molecular Reproduction and Genetics Facility, Department of Anatomy, All India Institute of Medical Sciences (AIIMS), New Delhi 110029, India
| | - Rekha Dwivedi
- Department of Neurology, All India Institute of Medical Sciences (AIIMS), New Delhi 110029, India
| | - Manisha Bansal
- Molecular Reproduction and Genetics Facility, Department of Anatomy, All India Institute of Medical Sciences (AIIMS), New Delhi 110029, India
| | - Manjari Tripathi
- Department of Neurology, All India Institute of Medical Sciences (AIIMS), New Delhi 110029, India
| | - Rima Dada
- Molecular Reproduction and Genetics Facility, Department of Anatomy, All India Institute of Medical Sciences (AIIMS), New Delhi 110029, India
| |
Collapse
|