1
|
Dong Y, Zhang Y, Xu F, Zou K. Extensive genomic characterization, pre-clinical probiotic evaluation, and safety analysis of Bifidobacterium longum subsp. longum BL21 isolated from infant feces. Microb Pathog 2024; 197:107100. [PMID: 39505088 DOI: 10.1016/j.micpath.2024.107100] [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: 05/12/2024] [Revised: 10/03/2024] [Accepted: 11/01/2024] [Indexed: 11/08/2024]
Abstract
OBJECTIVE This study aimed to evaluate the safety and probiotic properties of Bifidobacterium longum subsp. longum BL21 isolated from infant feces for use as a commercial probiotic strain. METHODS Whole-genome sequencing; physiological and biochemical assessments; enzymatic assays; metabolite, antibiotic sensitivity, cell adhesion and cytotoxicity, and tolerance tests; and a 14-day oral toxicity study were conducted. RESULTS BL21 exhibited genetic integrity, and its genome lacked genes related to antibiotic resistance or virulence. It was found to be non-pathogenic, had efficient carbohydrate metabolism and mucin degradation ability, and was free from biogenic amines. It also showed susceptibility to antibiotics, strong cell adhesion, and resilience to adverse conditions. The aforementioned results confirm that BL21 is a functional probiotic strain with genetic stability, enzymatic capabilities, and non-pathogenic properties that mean it is safe for oral consumption, demonstrating that it is a promising candidate for probiotic applications. CONCLUSION The study demonstrates that BL21 is a genetically stable, non-pathogenic probiotic strain with metabolic potential. The strain lacks virulence and antibiotic resistance genes, and its resilience to gastrointestinal conditions, as well as the results of the 14-day oral toxicity study, suggest that BL21 is safe for oral consumption. However, further long-term studies and clinical trials are needed to confirm its safety and efficacy for therapeutic use.
Collapse
Affiliation(s)
- Yao Dong
- Germline Stem Cells and Microenvironment Lab, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China; Stem Cell Research and Translation Center, Nanjing Agricultural University, Nanjing, 210095, China
| | - Yinan Zhang
- Shanghai Institute of Quality Inspection and Technical Research, Shanghai, 200233, China
| | - Fei Xu
- College of Food Science and Technology, Henan University of Technology, Zhengzhou, China
| | - Kang Zou
- Germline Stem Cells and Microenvironment Lab, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China; Stem Cell Research and Translation Center, Nanjing Agricultural University, Nanjing, 210095, China.
| |
Collapse
|
2
|
Subbarayudu S, Namasivayam SKR, Arockiaraj J. Immunomodulation in Non-traditional Therapies for Methicillin-resistant Staphylococcus aureus (MRSA) Management. Curr Microbiol 2024; 81:346. [PMID: 39240286 DOI: 10.1007/s00284-024-03875-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Accepted: 08/26/2024] [Indexed: 09/07/2024]
Abstract
The rise of methicillin-resistant Staphylococcus aureus (MRSA) poses a significant challenge in clinical settings due to its ability to evade conventional antibiotic treatments. This overview explores the potential of immunomodulatory strategies as alternative therapeutic approaches to combat MRSA infections. Traditional antibiotics are becoming less effective, necessitating innovative solutions that harness the body's immune system to enhance pathogen clearance. Recent advancements in immunotherapy, including the use of antimicrobial peptides, phage therapy, and mechanisms of immune cells, demonstrate promise in enhancing the body's ability to clear MRSA infections. However, the exact interactions between these therapies and immunomodulation are not fully understood, underscoring the need for further research. Hence, this review aims to provide a broad overview of the current understanding of non-traditional therapeutics and their impact on immune responses, which could lead to more effective MRSA treatment strategies. Additionally, combining immunomodulatory agents with existing antibiotics may improve outcomes, particularly for immunocompromised patients or those with chronic infections. As the landscape of antibiotic resistance evolves, the development of effective immunotherapeutic strategies could play a vital role in managing MRSA infections and reducing reliance on traditional antibiotics. Future research must focus on optimizing these approaches and validating their efficacy in diverse clinical populations to address the urgent need for effective MRSA management strategies.
Collapse
Affiliation(s)
- Suthi Subbarayudu
- Toxicology and Pharmacology Laboratory, Department of Biotechnology, Faculty of Science and Humanities, SRM Institute of Science and Technology, Kattankulathur, Chengalpattu District, Tamil Nadu, 603203, India
| | - S Karthick Raja Namasivayam
- Centre for Applied Research, Saveetha School of Engineering, Saveetha Institute of Medical and Technical Sciences (SIMATS), Chennai, Tamil Nadu, 602105, India.
| | - Jesu Arockiaraj
- Toxicology and Pharmacology Laboratory, Department of Biotechnology, Faculty of Science and Humanities, SRM Institute of Science and Technology, Kattankulathur, Chengalpattu District, Tamil Nadu, 603203, India.
| |
Collapse
|
3
|
Kostovova I, Kavanova K, Moravkova M, Gebauer J, Leva L, Vicenova M, Babak V, Faldyna M, Crhanova M. Probiotic bacteria of wild boar origin intended for piglets - An in vitro study. VET MED-CZECH 2024; 69:281-296. [PMID: 39296628 PMCID: PMC11406499 DOI: 10.17221/35/2024-vetmed] [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: 04/25/2024] [Accepted: 06/26/2024] [Indexed: 09/21/2024] Open
Abstract
Using probiotics represents a potential solution to post-weaning diarrheal diseases in piglets on commercial farms. The gastrointestinal tract of wild boars serves as a promising reservoir of novel lactic acid bacteria with suitable probiotic characteristics. In this study, we isolated eight bacterial strains from the intestinal content of wild boars identified as representatives of the species Bifidobacterium apri, Lactobacillus amylovorus, and Ligilactobacillus salivarius. These isolates underwent in vitro analysis and characterisation to assess their biological safety and probiotic properties. Analysis of their full genome sequences revealed the absence of horizontally transferrable genes for antibiotic resistance. However, seven out of eight isolates harboured genes encoding various types of bacteriocins in their genomes, and bacteriocin production was further confirmed by mass spectrometry analysis. Most of the tested strains demonstrated the ability to inhibit the growth of selected pathogenic bacteria, produce exopolysaccharides, and stimulate the expression of interleukin-10 in porcine macrophages. These characteristics deem the isolates characterised in this study as potential candidates for use as probiotics for piglets during the post-weaning period.
Collapse
Affiliation(s)
- Iveta Kostovova
- Department of Microbiology and Antimicrobial Resistance, Veterinary Research Institute, Brno, Czech Republic
| | - Katerina Kavanova
- Department of Microbiology and Antimicrobial Resistance, Veterinary Research Institute, Brno, Czech Republic
| | - Monika Moravkova
- Department of Microbiology and Antimicrobial Resistance, Veterinary Research Institute, Brno, Czech Republic
| | - Jan Gebauer
- Department of Infectious Diseases and Preventive Medicine, Veterinary Research Institute, Brno, Czech Republic
| | - Lenka Leva
- Department of Infectious Diseases and Preventive Medicine, Veterinary Research Institute, Brno, Czech Republic
| | - Monika Vicenova
- Department of Infectious Diseases and Preventive Medicine, Veterinary Research Institute, Brno, Czech Republic
| | - Vladimir Babak
- Department of Microbiology and Antimicrobial Resistance, Veterinary Research Institute, Brno, Czech Republic
| | - Martin Faldyna
- Department of Infectious Diseases and Preventive Medicine, Veterinary Research Institute, Brno, Czech Republic
| | - Magdalena Crhanova
- Department of Microbiology and Antimicrobial Resistance, Veterinary Research Institute, Brno, Czech Republic
| |
Collapse
|
4
|
Chen W, Guo K, Huang X, Zhang X, Li X, Chen Z, Wang Y, Wang Z, Liu R, Qiu H, Wang M, Zeng S. The Association of Neonatal Gut Microbiota Community State Types with Birth Weight. CHILDREN (BASEL, SWITZERLAND) 2024; 11:770. [PMID: 39062221 PMCID: PMC11276374 DOI: 10.3390/children11070770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 06/02/2024] [Accepted: 06/04/2024] [Indexed: 07/28/2024]
Abstract
BACKGROUND while most gut microbiota research has focused on term infants, the health outcomes of preterm infants are equally important. Very-low-birth-weight (VLBW) or extremely-low-birth-weight (ELBW) preterm infants have a unique gut microbiota structure, and probiotics have been reported to somewhat accelerate the maturation of the gut microbiota and reduce intestinal inflammation in very-low preterm infants, thereby improving their long-term outcomes. The aim of this study was to investigate the structure of gut microbiota in ELBW neonates to facilitate the early identification of different types of low-birth-weight (LBW) preterm infants. METHODS a total of 98 fecal samples from 39 low-birth-weight preterm infants were included in this study. Three groups were categorized according to different birth weights: ELBW (n = 39), VLBW (n = 39), and LBW (n = 20). The gut microbiota structure of neonates was obtained by 16S rRNA gene sequencing, and microbiome analysis was conducted. The community state type (CST) of the microbiota was predicted, and correlation analysis was conducted with clinical indicators. Differences in the gut microbiota composition among ELBW, VLBW, and LBW were compared. The value of gut microbiota composition in the diagnosis of extremely low birth weight was assessed via a random forest-machine learning approach. RESULTS we briefly analyzed the structure of the gut microbiota of preterm infants with low birth weight and found that the ELBW, VLBW, and LBW groups exhibited gut microbiota with heterogeneous compositions. Low-birth-weight preterm infants showed five CSTs dominated by Enterococcus, Staphylococcus, Klebsiella, Streptococcus, Pseudescherichia, and Acinetobacter. The birth weight and clinical indicators related to prematurity were associated with the CST. We found the composition of the gut microbiota was specific to the different types of low-birth-weight premature infants, namely, ELBW, VLBW, and LBW. The ELBW group exhibited significantly more of the potentially harmful intestinal bacteria Acinetobacter relative to the VLBW and LBW groups, as well as a significantly lower abundance of the intestinal probiotic Bifidobacterium. Based on the gut microbiota's composition and its correlation with low weight, we constructed random forest model classifiers to distinguish ELBW and VLBW/LBW infants. The area under the curve of the classifiers constructed with Enterococcus, Klebsiella, and Acinetobacter was found to reach 0.836 by machine learning evaluation, suggesting that gut microbiota composition may be a potential biomarker for ELBW preterm infants. CONCLUSIONS the gut bacteria of preterm infants showed a CST with Enterococcus, Klebsiella, and Acinetobacter as the dominant genera. ELBW preterm infants exhibit an increase in the abundance of potentially harmful bacteria in the gut and a decrease in beneficial bacteria. These potentially harmful bacteria may be potential biomarkers for ELBW preterm infants.
Collapse
Affiliation(s)
- Wanling Chen
- Shenzhen Clinical Medical College, Guangzhou University of Chinese Medicine, Shenzhen 518116, China
- Microbiome Therapy Center, South China Hospital, Medical School, Shenzhen University, Shenzhen 518111, China
| | - Kaiping Guo
- Division of Pediatrics, Longgang District Central Hospital of Shenzhen, Shenzhen 518116, China
| | - Xunbin Huang
- Division of Neonatology, Longgang District Central Hospital of Shenzhen, Shenzhen 518116, China
| | - Xueli Zhang
- Division of Neonatology, Shenzhen Longhua People’s Hospital, Shenzhen 518109, China
| | - Xiaoxia Li
- Division of Neonatology, Shenzhen Longhua People’s Hospital, Shenzhen 518109, China
| | - Zimiao Chen
- Department of Burn Plastic Surgery, South China Hospital, Shenzhen University, Shenzhen 518111, China
| | - Yanli Wang
- Department of Pediatrics, South China Hospital, Shenzhen University, Shenzhen 518111, China
| | - Zhangxing Wang
- Division of Neonatology, Shenzhen Longhua People’s Hospital, Shenzhen 518109, China
| | - Rongtian Liu
- Department of Pediatrics, Shenzhen Second People’s Hospital, Shenzhen 518035, China
| | - Huixian Qiu
- Division of Neonatology, Longgang District Central Hospital of Shenzhen, Shenzhen 518116, China
| | - Mingbang Wang
- Microbiome Therapy Center, South China Hospital, Medical School, Shenzhen University, Shenzhen 518111, China
- Department of Neonatology, Longgang District Maternity & Child Healthcare Hospital of Shenzhen City (Longgang Maternity and Child Institute of Shantou University Medical College), Shenzhen 518172, China
| | - Shujuan Zeng
- Division of Neonatology, Longgang District Central Hospital of Shenzhen, Shenzhen 518116, China
| |
Collapse
|
5
|
Green GBH, Cox-Holmes AN, Backan O, Valbak O, Potier ACE, Chen D, Morrow CD, Willey CD, McFarland BC. Exploring Gut Microbiota Alterations with Trimethoprim-Sulfamethoxazole and Dexamethasone in a Humanized Microbiome Mouse Model. Microorganisms 2024; 12:1015. [PMID: 38792844 PMCID: PMC11124107 DOI: 10.3390/microorganisms12051015] [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: 04/16/2024] [Revised: 05/06/2024] [Accepted: 05/10/2024] [Indexed: 05/26/2024] Open
Abstract
Along with the standard therapies for glioblastoma, patients are commonly prescribed trimethoprim-sulfamethoxazole (TMP-SMX) and dexamethasone for preventing infections and reducing cerebral edema, respectively. Because the gut microbiota impacts the efficacy of cancer therapies, it is important to understand how these medications impact the gut microbiota of patients. Using mice that have been colonized with human microbiota, this study sought to examine how TMP-SMX and dexamethasone affect the gut microbiome. Two lines of humanized microbiota (HuM) Rag1-/- mice, HuM1Rag and HuM2Rag, were treated with either TMP-SMX or dexamethasone via oral gavage once a day for a week. Fecal samples were collected pre-treatment (pre-txt), one week after treatment initiation (1 wk post txt), and three weeks post-treatment (3 wk post txt), and bacterial DNA was analyzed using 16S rRNA-sequencing. The HuM1Rag mice treated with TMP-SMX had significant shifts in alpha diversity, beta diversity, and functional pathways at all time points, whereas in the HuM2Rag mice, it resulted in minimal changes in the microbiome. Likewise, dexamethasone treatment resulted in significant changes in the microbiome of the HuM1Rag mice, whereas the microbiome of the HuM2Rag mice was mostly unaffected. The results of our study show that routine medications used during glioblastoma treatment can perturb gut microbiota, with some microbiome compositions being more sensitive than others, and these treatments could potentially affect the overall efficacy of standard-of-care therapy.
Collapse
Affiliation(s)
- George B. H. Green
- Department of Cell, Developmental and Integrative Biology, Birmingham, AL 35294, USA
| | - Alexis N. Cox-Holmes
- Department of Cell, Developmental and Integrative Biology, Birmingham, AL 35294, USA
| | - Olivia Backan
- Department of Cell, Developmental and Integrative Biology, Birmingham, AL 35294, USA
- Undergraduate Cancer Biology Program, Birmingham, AL 35294, USA
| | - Olivia Valbak
- Department of Cell, Developmental and Integrative Biology, Birmingham, AL 35294, USA
- Undergraduate Cancer Biology Program, Birmingham, AL 35294, USA
| | - Anna Claire E. Potier
- Department of Cell, Developmental and Integrative Biology, Birmingham, AL 35294, USA
- Undergraduate Cancer Biology Program, Birmingham, AL 35294, USA
| | | | - Casey D. Morrow
- Department of Cell, Developmental and Integrative Biology, Birmingham, AL 35294, USA
| | - Christopher D. Willey
- Department of Radiation Oncology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Braden C. McFarland
- Department of Cell, Developmental and Integrative Biology, Birmingham, AL 35294, USA
| |
Collapse
|
6
|
Liu D, Li C, Cao T, Lv X, Yue Y, Li S, Cheng Y, Liu F, Huo G, Li B. Bifidobacterium longum K5 Prevents Enterohaemorrhagic Escherichia coli O157:H7 Infection in Mice through the Modulation of the Gut Microbiota. Nutrients 2024; 16:1164. [PMID: 38674854 PMCID: PMC11053520 DOI: 10.3390/nu16081164] [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: 03/21/2024] [Revised: 04/11/2024] [Accepted: 04/11/2024] [Indexed: 04/28/2024] Open
Abstract
Enterohemorrhagic Escherichia coli (EHEC) serotype O157:H7 is a commonly encountered foodborne pathogen that can cause hemorrhagic enteritis and lead to hemolytic uremic syndrome (HUS) in severe cases. Bifidobacterium is a beneficial bacterium that naturally exists in the human gut and plays a vital role in maintaining a healthy balance in the gut microbiota. This study investigated the protective effects of B. longum K5 in a mouse model of EHEC O157:H7 infection. The results indicated that pretreatment with B. longum K5 mitigated the clinical symptoms of EHEC O157:H7 infection and attenuated the increase in myeloperoxidase (MPO) activity in the colon of the mice. In comparison to the model group, elevated serum D-lactic acid concentrations and diamine oxidase (DAO) levels were prevented in the K5-EHEC group of mice. The reduced mRNA expression of tight junction proteins (ZO-1, Occludin, and Claudin-1) and mucin MUC2, as well as the elevated expression of virulence factors Stx1A and Stx2A, was alleviated in the colon of both the K5-PBS and K5-EHEC groups. Additionally, the increase in the inflammatory cytokine levels of TNF-α and IL-1β was inhibited and the production of IL-4 and IL-10 was promoted in the K5-EHEC group compared with the model group. B. longum K5 significantly prevented the reduction in the abundance and diversity of mouse gut microorganisms induced by EHEC O157:H7 infection, including blocking the decrease in the relative abundance of Roseburia, Lactobacillus, and Oscillibacter. Meanwhile, the intervention with B. longum K5 promoted the production of acetic acid and butyric acid in the gut. This study provides insights into the use of B. longum K5 for developing probiotic formulations to prevent intestinal diseases caused by pathogenic bacterial infections.
Collapse
Affiliation(s)
- Deyu Liu
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin 150030, China; (D.L.); (C.L.); (T.C.); (X.L.); (Y.Y.); (S.L.); (Y.C.); (F.L.); (B.L.)
- Food College, Northeast Agricultural University, Harbin 150030, China
| | - Chunyan Li
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin 150030, China; (D.L.); (C.L.); (T.C.); (X.L.); (Y.Y.); (S.L.); (Y.C.); (F.L.); (B.L.)
- Food College, Northeast Agricultural University, Harbin 150030, China
| | - Ting Cao
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin 150030, China; (D.L.); (C.L.); (T.C.); (X.L.); (Y.Y.); (S.L.); (Y.C.); (F.L.); (B.L.)
- Food College, Northeast Agricultural University, Harbin 150030, China
| | - Xiuli Lv
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin 150030, China; (D.L.); (C.L.); (T.C.); (X.L.); (Y.Y.); (S.L.); (Y.C.); (F.L.); (B.L.)
- Food College, Northeast Agricultural University, Harbin 150030, China
| | - Yingxue Yue
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin 150030, China; (D.L.); (C.L.); (T.C.); (X.L.); (Y.Y.); (S.L.); (Y.C.); (F.L.); (B.L.)
- Food College, Northeast Agricultural University, Harbin 150030, China
| | - Shuang Li
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin 150030, China; (D.L.); (C.L.); (T.C.); (X.L.); (Y.Y.); (S.L.); (Y.C.); (F.L.); (B.L.)
- Food College, Northeast Agricultural University, Harbin 150030, China
| | - Yang Cheng
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin 150030, China; (D.L.); (C.L.); (T.C.); (X.L.); (Y.Y.); (S.L.); (Y.C.); (F.L.); (B.L.)
- Food College, Northeast Agricultural University, Harbin 150030, China
| | - Fei Liu
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin 150030, China; (D.L.); (C.L.); (T.C.); (X.L.); (Y.Y.); (S.L.); (Y.C.); (F.L.); (B.L.)
- Food College, Northeast Agricultural University, Harbin 150030, China
| | - Guicheng Huo
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin 150030, China; (D.L.); (C.L.); (T.C.); (X.L.); (Y.Y.); (S.L.); (Y.C.); (F.L.); (B.L.)
- Food College, Northeast Agricultural University, Harbin 150030, China
| | - Bailiang Li
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin 150030, China; (D.L.); (C.L.); (T.C.); (X.L.); (Y.Y.); (S.L.); (Y.C.); (F.L.); (B.L.)
- Food College, Northeast Agricultural University, Harbin 150030, China
| |
Collapse
|
7
|
Zhong L, Hu Q, Zhan Q, Zhao M, Zhao L. Oat protein isolate- Pleurotus ostreatus β-glucan conjugate nanoparticles bound to β-carotene effectively alleviate immunosuppression by regulating gut microbiota. Food Funct 2024; 15:1867-1883. [PMID: 38236028 DOI: 10.1039/d3fo05158g] [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: 01/19/2024]
Abstract
Individuals with immune disorders cannot establish an adequate defense to pathogens, leading to gut microbiota dysbiosis. β-Carotene can regulate immune response, but its bioavailability in vivo is very low. Herein, we developed a glycosylated oat protein-based nanoparticle to improve the application of β-carotene for mitigating cyclophosphamide-induced immunosuppression and gut microbiota imbalance in mice. The results showed that the nanoparticles facilitated a conversion of β-carotene to retinol or retinyl palmitate into the systemic circulation, leading to an increased bioavailability of β-carotene. The encapsulated β-carotene bolstered humoral immunity by elevating immunoglobulin levels, augmenting splenic T lymphocyte subpopulations, and increasing splenic cytokine concentrations in immunosuppressed mice. This effect was accompanied by the alleviation of pathological features observed in the spleen. In addition, the encapsulated β-carotene restored the abnormal gut microbiota associated with immunosuppression, including Erysipelotrichaceae, Akkermansia, Bifidobacterium and Roseburia. This study suggested that nanoparticles loaded with β-carotene have great potential for therapeutic intervention in human immune disorders by specifically targeting the gut microbiota.
Collapse
Affiliation(s)
- Lei Zhong
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing, 210095, P.R. China
| | - Qiuhui Hu
- College of Food Science and Engineering, Nanjing University of Finance and Economics/Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing, 210023, P.R. China.
| | - Qiping Zhan
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing, 210095, P.R. China
| | - Mingwen Zhao
- Key Laboratory of Agricultural Environmental Microbiology, Ministry of Agriculture; Microbiology Department, College of Life Sciences, Nanjing Agricultural University, Nanjing, 210095, P.R. China
| | - Liyan Zhao
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing, 210095, P.R. China
| |
Collapse
|
8
|
Petruzziello C, Saviano A, Manetti LL, Macerola N, Ojetti V. The Role of Gut Microbiota and the Potential Effects of Probiotics in Heart Failure. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:271. [PMID: 38399558 PMCID: PMC10890346 DOI: 10.3390/medicina60020271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 01/25/2024] [Accepted: 01/30/2024] [Indexed: 02/25/2024]
Abstract
Heart failure (HF) remains a significant global health challenge, affecting millions of individuals worldwide and posing a substantial burden on healthcare systems. HF is a syndrome of intricate pathophysiology, involving systemic inflammation, oxidative stress, metabolic perturbations, and maladaptive structural changes in the heart. It is influenced by complex interactions between cardiac function, systemic physiology, and environmental factors. Among these factors, the gut microbiota has emerged as a novel and intriguing player in the landscape of HF pathophysiology. The gut microbiota, beyond its role in digestion and nutrient absorption, impacts immune responses, metabolic processes, and, as suggested by evidence in the literature, the development and progression of HF. There is a bidirectional communication between the gut and the heart, often known as the gut-heart axis, through which gut microbiota-derived metabolites, immune signals, and microbial products exert profound effects on cardiovascular health. This review aims to provide a comprehensive overview of the intricate relationship between the gut microbiota and HF. Additionally, we explore the potential of using probiotics as a therapeutic strategy to modulate the gut microbiota's composition and attenuate the adverse effects observed in HF. Conventional therapeutic approaches targeting hemodynamic and neurohormonal dysregulation have substantially improved the management of HF, but emerging research is exploring the potential implications of harnessing the gut microbiota for innovative approaches in HF treatment.
Collapse
Affiliation(s)
- Carmine Petruzziello
- Emergency Department, Ospedale San Carlo di Nancy—GVM Care & Research, 00165 Rome, Italy; (C.P.); (L.L.M.)
| | - Angela Saviano
- Emergency Department, Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy;
| | - Luca Luigi Manetti
- Emergency Department, Ospedale San Carlo di Nancy—GVM Care & Research, 00165 Rome, Italy; (C.P.); (L.L.M.)
| | - Noemi Macerola
- Internal Medicine, Ospedale San Carlo di Nancy—GVM Care & Research, 00165 Rome, Italy;
| | - Veronica Ojetti
- Internal Medicine, Ospedale San Carlo di Nancy—GVM Care & Research, 00165 Rome, Italy;
- Deaprtment of Internal Medicine, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| |
Collapse
|
9
|
Taufer CR, Rampelotto PH. Lactobacilli in COVID-19: A Systematic Review Based on Next-Generation Sequencing Studies. Microorganisms 2024; 12:284. [PMID: 38399688 PMCID: PMC10891515 DOI: 10.3390/microorganisms12020284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 01/25/2024] [Accepted: 01/26/2024] [Indexed: 02/25/2024] Open
Abstract
The global pandemic was caused by the SARS-CoV-2 virus, known as COVID-19, which primarily affects the respiratory and intestinal systems and impacts the microbial communities of patients. This systematic review involved a comprehensive search across the major literature databases to explore the relationship between lactobacilli and COVID-19. Our emphasis was on investigations employing NGS technologies to explore this connection. Our analysis of nine selected studies revealed that lactobacilli have a reduced abundance in the disease and an association with disease severity. The protective mechanisms of lactobacilli in COVID-19 and other viral infections are likely to be multifaceted, involving complex interactions between the microbiota, the host immune system, and the virus itself. Moreover, upon closely examining the NGS methodologies and associated statistical analyses in each research study, we have noted concerns regarding the approach used to delineate the varying abundance of lactobacilli, which involves potential biases and the exclusion of pertinent data elements. These findings provide new insight into the relationship between COVID-19 and lactobacilli, highlighting the potential for microbiota modulation in COVID-19 treatment.
Collapse
Affiliation(s)
- Clarissa Reginato Taufer
- Graduate Program in Genetics and Molecular Biology, Universidade Federal do Rio Grande do Sul, Porto Alegre 91501-970, Brazil
| | - Pabulo Henrique Rampelotto
- Bioinformatics and Biostatistics Core Facility, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre 91501-970, Brazil
| |
Collapse
|
10
|
Samarra A, Cabrera-Rubio R, Martínez-Costa C, Collado MC. The role of Bifidobacterium genus in modulating the neonate microbiota: implications for antibiotic resistance acquisition in early life. Gut Microbes 2024; 16:2357176. [PMID: 38798019 PMCID: PMC11135851 DOI: 10.1080/19490976.2024.2357176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Accepted: 05/15/2024] [Indexed: 05/29/2024] Open
Abstract
Resistance to antibiotics in newborns is a huge concern as their immune system is still developing, and infections and resistance acquisition in early life have short- and long-term consequences for their health. Bifidobacterium species are important commensals capable of dominating the infant gut microbiome and are known to be less prone to possess antimicrobial resistance genes than other taxa that may colonize infants. We aimed to study the association between Bifidobacterium-dominated infant gut microbiota and the antibiotic resistant gene load in neonates, and to ascertain the perinatal factors that may contribute to the antibiotic resistance acquisition. Two hundred infant fecal samples at 7 days and 1 month of age from the MAMI birth cohort were included in the study and for whom maternal-neonatal clinical records were available. Microbiota profiling was carried out by 16S rRNA amplicon sequencing, and targeted antibiotic resistance genes (ARGs) including tetM, tetW, tetO, blaTEM, blaSHV and ermB were quantified by qPCR. Infant microbiota clustered into two distinct groups according to their Bifidobacterium genus abundance: high and low. The main separation of groups or clusters at each time point was performed with an unsupervised non-linear algorithm of k-means partitioning to cluster data by time points based on Bifidobacterium genus relative abundance. Microbiota composition differed significantly between both groups, and specific bifidobacterial species were enriched in each cluster. Lower abundance of Bifidobacterium in the infant gut was associated with a higher load of antibiotic resistance genes. Our results highlight the relevance of Bifidobacterium genus in the early acquisition and establishment of antibiotic resistance in the gut. Further studies are needed to develop strategies to promote a healthy early colonization and fight against the spread of antibiotic resistances.
Collapse
Affiliation(s)
- Anna Samarra
- Department of Biotechnology, Institute of Agrochemistry and Food Technology- National Research Council (IATA-CSIC), Paterna, Valencia, Spain
| | - Raúl Cabrera-Rubio
- Department of Biotechnology, Institute of Agrochemistry and Food Technology- National Research Council (IATA-CSIC), Paterna, Valencia, Spain
| | - Cecilia Martínez-Costa
- Department of Pediatrics, School of Medicine, University of Valencia, Valencia, Spain
- Pediatric Gastroenterology and Nutrition Section, Hospital Clínico Universitario Valencia, Valencia, Spain
| | - Maria Carmen Collado
- Department of Biotechnology, Institute of Agrochemistry and Food Technology- National Research Council (IATA-CSIC), Paterna, Valencia, Spain
| |
Collapse
|
11
|
Zhang G, He M, Xiao L, Jiao Y, Han J, Li C, Miller MJ, Zhang L. Milk fat globule membrane protects Bifidobacterium longum ssp. infantis ATCC 15697 against bile stress by modifying global transcriptional responses. J Dairy Sci 2024; 107:91-104. [PMID: 37678788 DOI: 10.3168/jds.2023-23591] [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: 04/07/2023] [Accepted: 07/17/2023] [Indexed: 09/09/2023]
Abstract
The milk fat globule membrane (MFGM) can protect probiotic bacteria from bile stress. However, its potential mechanism has not been reported. In this study, the viability, morphology and gene transcriptional response of Bifidobacterium longum ssp. infantis ATCC 15697 (BI_15697) stressed by bile salts with or without MFGM were investigated. It was shown that MFGM alleviated the reduction in BI_15697 population induced by 0.2% porcine bile stress and restored the population to the control levels. MFGM ameliorated the shrunken, fragmented appearance and irregular morphology of BI_15697 and maintained cell integrity disrupted by bile stress. RNA-sequencing results showed that MFGM increased transport of glucose and raffinose and decreased that of branched-chain amino acids (BCAA) in the presence of bile salts. MFGM stimulated the expression of genes involved in the synthesis of raffinose in galactose metabolism and the metabolism of BCAA, suggesting that MFGM stimulated the accumulation of raffinose and BCAA in the presence of bile. In addition, MFGM stimulated the expression of 2 bile efflux transporters under bile stress. Together, the multifactorial response helps BI_15697 excrete bile salts and maintain cellular integrity in response to bile stress. This study proposes a mechanism for the protection of BI_15697 against bile salt stress by MFGM, thereby providing a molecular basis for its application in incorporation of probiotics.
Collapse
Affiliation(s)
- Gongsheng Zhang
- Key Laboratory of Dairy Science, Ministry of Education, College of Food Science, Northeast Agricultural University, Harbin 150030, China
| | - Mingxue He
- Key Laboratory of Dairy Science, Ministry of Education, College of Food Science, Northeast Agricultural University, Harbin 150030, China
| | - Lihong Xiao
- Key Laboratory of Dairy Science, Ministry of Education, College of Food Science, Northeast Agricultural University, Harbin 150030, China
| | - Yuehua Jiao
- Center of Drug Safety Evaluation, Heilongjiang University of Chinese Medicine, Harbin 150040, China
| | - Jianchun Han
- Key Laboratory of Dairy Science, Ministry of Education, College of Food Science, Northeast Agricultural University, Harbin 150030, China; Heilongjiang Green Food Science Research Institute, Harbin 150030, China.
| | - Chun Li
- Key Laboratory of Dairy Science, Ministry of Education, College of Food Science, Northeast Agricultural University, Harbin 150030, China; Center of Drug Safety Evaluation, Heilongjiang University of Chinese Medicine, Harbin 150040, China
| | - Michael J Miller
- Department of Food Science and Human Nutrition, University of Illinois Urbana-Champaign, Urbana, IL 61801
| | - Lili Zhang
- Key Laboratory of Dairy Science, Ministry of Education, College of Food Science, Northeast Agricultural University, Harbin 150030, China.
| |
Collapse
|
12
|
Qiao Y, Chen J, Jiang Y, Zhang Z, Wang H, Liu T, Yang Z, Fu G, Chen Y. Gut microbiota composition may be an indicator of erectile dysfunction. Microb Biotechnol 2024; 17:e14403. [PMID: 38226944 PMCID: PMC10832513 DOI: 10.1111/1751-7915.14403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 11/29/2023] [Accepted: 12/21/2023] [Indexed: 01/17/2024] Open
Abstract
Erectile Dysfunction (ED) is considered a physical and mental illness. A variety of potential associations between gut microbiota and health or disease have been found. By comparing the gut microbiota of healthy controls and ED patients, our study investigated the relationship between ED and gut microbiota. The results revealed that the ED group exhibited a significantly higher relative abundance of Bacteroides, Fusobacterium, Lachnoclostridium, Escherichia-Shigella and Megamonas, while showing a significantly lower relative abundance of Bifidobacterium compared to the control group. The dysbiosis of gut microbiota played a role in the onset and progression of ED by influencing the gut barrier, cardiovascular system and mental health, which provided a novel perspective on understanding the pathophysiology of ED. What is more, we had identified several key gut microbiota. By combining 16S rRNA sequencing with machine learning techniques, we were able to uncover the significant value and impact of gut microbiota in the early detection of ED.
Collapse
Affiliation(s)
- Yu Qiao
- Department of AndrologyJiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese MedicineNanjingJiangsuChina
- Center for ReproductionThe Affiliated Huai'an No. 1 People's Hospital of Nanjing Medical UniversityHuai'anJiangsuChina
| | - Jianhuai Chen
- Department of AndrologyJiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese MedicineNanjingJiangsuChina
| | - Yongsheng Jiang
- Department of AndrologyJiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese MedicineNanjingJiangsuChina
| | - Ziheng Zhang
- Department of AndrologyJiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese MedicineNanjingJiangsuChina
| | - Heng Wang
- Department of AndrologyJiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese MedicineNanjingJiangsuChina
| | - Tao Liu
- Department of AndrologyJiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese MedicineNanjingJiangsuChina
| | - Zhaoxu Yang
- Department of AndrologyJiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese MedicineNanjingJiangsuChina
| | - Guangbo Fu
- Department of UrologyThe Affiliated Huai'an No. 1 People's Hospital of Nanjing Medical UniversityHuai'anJiangsuChina
| | - Yun Chen
- Department of AndrologyJiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese MedicineNanjingJiangsuChina
| |
Collapse
|
13
|
Hizo GH, Rampelotto PH. The Role of Bifidobacterium in Liver Diseases: A Systematic Review of Next-Generation Sequencing Studies. Microorganisms 2023; 11:2999. [PMID: 38138143 PMCID: PMC10745637 DOI: 10.3390/microorganisms11122999] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Revised: 12/06/2023] [Accepted: 12/15/2023] [Indexed: 12/24/2023] Open
Abstract
The physiopathology of liver diseases is complex and can be caused by various factors. Bifidobacterium is a bacterial genus commonly found in the human gut microbiome and has been shown to influence the development of different stages of liver diseases significantly. This study investigated the relationship between the Bifidobacterium genus and liver injury. In this work, we performed a systematic review in major databases using the key terms "Bifidobacterium", "ALD", "NAFLD", "NASH", "cirrhosis", and "HCC" to achieve our purpose. In total, 31 articles were selected for analysis. In particular, we focused on studies that used next-generation sequencing (NGS) technologies. The studies focused on assessing Bifidobacterium levels in the diseases and interventional aimed at examining the therapeutic potential of Bifidobacterium in the mentioned conditions. Overall, the abundance of Bifidobacterium was reduced in hepatic pathologies. Low levels of Bifidobacterium were associated with harmful biochemical and physiological parameters, as well as an adverse clinical outcome. However, interventional studies using different drugs and treatments were able to increase the abundance of the genus and improve clinical outcomes. These results strongly support the hypothesis that changes in the abundance of Bifidobacterium significantly influence both the pathophysiology of hepatic diseases and the related clinical outcomes. In addition, our critical assessment of the NGS methods and related statistical analyses employed in each study highlights concerns with the methods used to define the differential abundance of Bifidobacterium, including potential biases and the omission of relevant information.
Collapse
Affiliation(s)
- Gabriel Henrique Hizo
- Graduate Program in Gastroenterology and Hepatology Sciences, Universidade Federal do Rio Grande do Sul, Porto Alegre 90035-003, Brazil
| | - Pabulo Henrique Rampelotto
- Bioinformatics and Biostatistics Core Facility, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre 91501-907, Brazil
| |
Collapse
|
14
|
Yan J, Zhang R, Kang J, Zhong Y, Abudurexiti A, Tan H, Lei Y, Ma X. Effect of Cichorium glandulosum on intestinal microbiota and bile acid metabolism in db/db mice. Food Sci Nutr 2023; 11:7765-7778. [PMID: 38107125 PMCID: PMC10724598 DOI: 10.1002/fsn3.3694] [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: 05/07/2023] [Revised: 08/31/2023] [Accepted: 09/03/2023] [Indexed: 12/19/2023] Open
Abstract
This study aims to investigate the effects of Chorum glandulosum Boiss. et Huet (CG) on the intestinal microbiota and serum bile acid (BA) in db/db mice. A total of 12 db/db mice were randomly divided into model (MOD), high-dose CG (CGH), and control (CON) groups. The CON and MOD groups received distilled water by gavage for 8 weeks. Whereas, the CGH group received an alcohol extract of CG at a dose of 200 mg/kg/day. Results showed that CG can reduce blood lipid levels. It change the composition of the intestinal microbiota, and increase the relative abundances of Muribaculaceae, Prevotellaceae, Bifidobacterium_pseudolongum, Bacteroidaceae in db/db mice as well. LC-MS metabolomics results showed that CG adjusted the serum BA levels. The results reduced the levels of primary BAs, such as cholic acid (CA) and chenodeoxycholic acid (CDCA). The results decreased the primary BA/secondary BA (PSA/SBA) ratio in db/db mice. Correlation analysis showed that the abundances of Bifidobacterium_pseudolongum and Bacteroidaceae were positively correlated with acetic acid level and negatively correlated with ursocholic acid (UCA), α-muricholic acid (αMCA), triglyceride (TG), and total cholesterol levels (TC), indicating an interaction between the intestinal microbiota and serum BAs. CG may play a positive role in the interaction between the intestinal microbiota and BAs in lipid metabolism.
Collapse
Affiliation(s)
- Junlin Yan
- College of PharmacyXinjiang Medical UniversityXinjiangChina
| | - Rui Zhang
- College of PharmacyXinjiang Medical UniversityXinjiangChina
| | - Jinsen Kang
- College of PharmacyXinjiang Medical UniversityXinjiangChina
| | - Yewei Zhong
- College of PharmacyXinjiang Medical UniversityXinjiangChina
| | | | - Huiwen Tan
- College of PharmacyXinjiang Medical UniversityXinjiangChina
| | - Yi Lei
- College of PharmacyXinjiang Medical UniversityXinjiangChina
| | - Xiaoli Ma
- College of PharmacyXinjiang Medical UniversityXinjiangChina
| |
Collapse
|
15
|
Luo Y, Zhang Y, Yang Y, Wu S, Zhao J, Li Y, Kang X, Li Z, Chen J, Shen X, He F, Cheng R. Bifidobacterium infantis and 2'-fucosyllactose supplementation in early life may have potential long-term benefits on gut microbiota, intestinal development, and immune function in mice. J Dairy Sci 2023; 106:7461-7476. [PMID: 37641283 DOI: 10.3168/jds.2023-23367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Accepted: 05/10/2023] [Indexed: 08/31/2023]
Abstract
The health benefits of nutritional interventions targeting the gut microbiota in early life are transient, such as probiotics, prebiotics, and synbiotics. This study sought to determine whether supplementation with Bifidobacterium infantis 79 (B79), 2'-fucosyllactose (2'-FL), or both (B79+2'FL) would lead to persistent health benefits in neonatal BALB/c mice. We found that at postnatal day (PND) 21, Ki67 and MUC2 expression increased, while total serum IgE content decreased in the B79, 2'-FL, and B79+2'-FL groups. The gut microbiota structure and composition altered as well. The levels of propionic acid, sIgA, and IL-10 increased in the 2'-FL group. Moreover, butyric acid content increased, while IL-6, IL-12p40, and tumor necrosis factor-α decreased in the B79+2'-FL group. At PND 56, Ki67 and MUC2 expression increased, whereas the gut microbiota remained altered in all 3 groups. The serum total IgG level increased only in the B79+2'-FL group. In conclusion, our study suggests that early-life supplementation with B79, 2'-FL, or their combination persistently alters the gut microbiome and promotes intestinal development; the immunomodulatory capacity of B79 and 2'-FL occurs during weaning, and their combination may persist into adulthood.
Collapse
Affiliation(s)
- Yating Luo
- Department of Nutrition and Food Hygiene, West China School of Public Health and West China Fourth Hospital, Sichuan University, 610041 Chengdu, Sichuan, China
| | - Yujie Zhang
- Department of Nutrition and Food Hygiene, West China School of Public Health and West China Fourth Hospital, Sichuan University, 610041 Chengdu, Sichuan, China
| | - Yang Yang
- Department of Nutrition and Food Hygiene, West China School of Public Health and West China Fourth Hospital, Sichuan University, 610041 Chengdu, Sichuan, China
| | - Simou Wu
- Department of Nutrition and Food Hygiene, West China School of Public Health and West China Fourth Hospital, Sichuan University, 610041 Chengdu, Sichuan, China
| | - Jincheng Zhao
- Department of Nutrition and Food Hygiene, West China School of Public Health and West China Fourth Hospital, Sichuan University, 610041 Chengdu, Sichuan, China
| | - Yun Li
- Department of Nutrition and Food Hygiene, West China School of Public Health and West China Fourth Hospital, Sichuan University, 610041 Chengdu, Sichuan, China
| | - Xiaohong Kang
- R&D Center, Inner Mongolia Meng Niu Dairy Industry (Group) Co. Ltd., 011500 Hohhot, Inner Mongolia, China
| | - Zhouyong Li
- R&D Center, Inner Mongolia Meng Niu Dairy Industry (Group) Co. Ltd., 011500 Hohhot, Inner Mongolia, China
| | - Jianguo Chen
- R&D Center, Inner Mongolia Meng Niu Dairy Industry (Group) Co. Ltd., 011500 Hohhot, Inner Mongolia, China; Beijing YuGen Pharmaceutical Co. Ltd., 102600 Beijing, China
| | - Xi Shen
- Department of Nutrition and Food Hygiene, West China School of Public Health and West China Fourth Hospital, Sichuan University, 610041 Chengdu, Sichuan, China
| | - Fang He
- Department of Nutrition and Food Hygiene, West China School of Public Health and West China Fourth Hospital, Sichuan University, 610041 Chengdu, Sichuan, China.
| | - Ruyue Cheng
- Department of Nutrition and Food Hygiene, West China School of Public Health and West China Fourth Hospital, Sichuan University, 610041 Chengdu, Sichuan, China.
| |
Collapse
|
16
|
Yu D, Pei Z, Chen Y, Wang H, Xiao Y, Zhang H, Chen W, Lu W. Bifidobacterium longum subsp. infantis as widespread bacteriocin gene clusters carrier stands out among the Bifidobacterium. Appl Environ Microbiol 2023; 89:e0097923. [PMID: 37681950 PMCID: PMC10537742 DOI: 10.1128/aem.00979-23] [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: 06/17/2023] [Accepted: 07/03/2023] [Indexed: 09/09/2023] Open
Abstract
Bifidobacterium is the dominant genus, particularly in the intestinal tract niche of healthy breast-fed infants, and many of these strains have been proven to elicit positive effects on infant development. In addition to its effective antimicrobial activity against detrimental microorganisms, it helps to improve the intestinal microbiota balance. The isolation and identification of bacteriocins from Bifidobacterium have been limited since the mid-1980s, leading to an underestimation of its ability for bacteriocin production. Here, we employed a silicon-based search strategy to mine 354 putative bacteriocin gene clusters (BGCs), most of which have never been reported, from the genomes of 759 Bifidobacterium strains distributed across 9 species. Consistent with previous reports, most Bifidobacterium strains did not carry or carry only a single BGC; however, Bifidobacterium longum subsp. infantis, in contrast to other Bifidobacterium species, carried numerous BGCs, including lanthipeptides, lasso peptides, thiopeptides, and class IId bacteriocins. The antimicrobial activity of the crude bacteriocins and transcription analysis confirmed its potential for bacteriocin biosynthesis. Additionally, we investigated the association of bacteriocins with the phylogenetic positions of their homologs from other genera and niches. In conclusion, this study re-examines a few Bifidobacterium species traditionally regarded as a poor source of bacteriocins. These bacteriocin genes impart a competitive advantage to Bifidobacterium in colonizing the infant intestinal tract. IMPORTANCE Development of the human gut microbiota commences from birth, with bifidobacteria being among the first colonizers of the newborn intestinal tract and dominating it for a considerable period. To date, the genetic basis for the successful adaptation of bifidobacteria to this particular niche remains unclear since studies have mainly focused on glycoside hydrolase and adhesion-related genes. Bacteriocins are competitive factors that help producers maintain colonization advantages without destroying the niche balance; however, they have rarely been reported in Bifidobacterium. The advancement in sequencing methods and bacteriocin databases enables the use of a silicon-based search strategy for the comprehensive and rapid re-evaluation of the bacteriocin distribution of Bifidobacterium. Our study revealed that B. infantis carries abundant bacteriocin biosynthetic gene clusters for the first time, presenting new evidence regarding the competitive interactions of Bifidobacterium in the infant intestinal tract.
Collapse
Affiliation(s)
- Di Yu
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu, China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
| | - Zhangming Pei
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu, China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
| | - Yutao Chen
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu, China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
| | - Hongchao Wang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu, China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
| | - Yue Xiao
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu, China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
| | - Hao Zhang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu, China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, Jiangsu, China
| | - Wei Chen
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu, China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, Jiangsu, China
| | - Wenwei Lu
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu, China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, Jiangsu, China
| |
Collapse
|
17
|
Mingdong W, Xiang G, Yongjun Q, Mingshuai W, Hao P. Causal associations between gut microbiota and urological tumors: a two-sample mendelian randomization study. BMC Cancer 2023; 23:854. [PMID: 37697271 PMCID: PMC10496293 DOI: 10.1186/s12885-023-11383-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Accepted: 09/06/2023] [Indexed: 09/13/2023] Open
Abstract
BACKGROUND Dysbiosis of gut microbiota has been linked to numerous diseases, including cancer. The unique role of gut microbiota in urological tumors is gaining prominence. However, it is still controversial whether the dysbiosis of gut microbiota should be one of the etiological factors of bladder cancer (BCa), prostate cancer (PCa) or kidney cancer (KCa). MATERIALS AND METHODS The microbiome genome-wide association study (GWAS) from the MiBioGen consortium (18,340 samples of 24 population-based cohorts) was utilized as the exposure data. Additionally, outcomes data (951 BCa cases and 307,092 controls; 1,631 KCa cases and 238,678 controls; 79,148 PCa cases and 61,106 controls) were extracted from the GWAS of the FinnGen and PRACTICAL consortia. To detect the potential causative bacterial traits for BCa, PCa, and KCa, a two-sample Mendelian randomization (MR) analysis was performed, employing the inverse-variance weighted or Wald ratio method. Sensitivity analyses were subsequently conducted to explore the robustness of the primary results. Finally, the reverse MR analysis was undertaken to mitigate the reverse causation. RESULTS This study suggested that Bifidobacterium (p = 0.030), Actinobacteria (p = 0.037 for phylum, 0.041 for class), and Ruminococcustorques group (p = 0.018), exhibited an association with an increased risk of BCa using either the inverse-variance weighted or Wald ratio method. By utilizing the Wald ratio method, Allisonella (p = 0.004, p = 0.038) was associated with a decreased risk of BCa and PCa, respectively. Furthermore, Ruminococcustorques group (p = 0.028) and Erysipelatoclostridium (p = 0.048) were causally linked to an elevated risk of KCa. CONCLUSIONS This MR study supports that genetically predicted gut microbiota is causally related to BCa, PCa and KCa. Additionally, distinct bacterial traits are identified in relation to each tumor type.
Collapse
Affiliation(s)
- Wang Mingdong
- Department of Urology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Gao Xiang
- Department of Otolaryngology Head and Neck Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Quan Yongjun
- Department of Urology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Wang Mingshuai
- Department of Urology, Cancer Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Ping Hao
- Department of Urology, Beijing Tongren Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
18
|
Grozina AA, Ilina LA, Laptev GY, Yildirim EA, Ponomareva ES, Filippova VA, Tyurina DG, Fisinin VI, Kochish II, Griffin DK, Surai PF, Romanov MN. Probiotics as an alternative to antibiotics in modulating the intestinal microbiota and performance of broiler chickens. J Appl Microbiol 2023; 134:lxad213. [PMID: 37715326 DOI: 10.1093/jambio/lxad213] [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: 08/01/2023] [Revised: 09/02/2023] [Accepted: 09/13/2023] [Indexed: 09/17/2023]
Abstract
AIMS Gut bacteria play an important role in poultry nutrition and the immune defense system. Changes in the intestinal microbiome affect the physiological state, metabolism, and innate immunity of poultry. The present study aimed to characterize age-related changes in the gastrointestinal tract microflora in broiler chickens, depending on supplementation of the diet with the in-feed antibiotic Stafac® 110 and a Bacillus subtilis strain-based probiotic. METHODS AND RESULTS In this regard, a comprehensive analysis of the taxonomic structure of the microbial community in the gastrointestinal tract (GIT) of broiler chickens was carried out using a molecular genetic technique of the terminal-restriction fragment length polymorphism analysis and taking into account age dynamics and feeding treatment. A beneficial effect on the microbiological composition and body weight of broilers was observed when using the antibiotic and probiotic in compound feeds. Different bacterial communities were revealed in the duodenum and cecum, and their positive impact on broiler growth was established. The results obtained shed light on the formation of GIT microflora of broiler chickens during the growing period and its changes in response to the use of the antibiotic and the probiotic. CONCLUSIONS We suggest that the implementation of the tested in-feed antibiotic and probiotic can be beneficial in regulating the intestinal microflora microbiological processes in the GIT and improving the feeding efficiency and productivity of broiler chickens.
Collapse
Affiliation(s)
- Alena A Grozina
- Federal Scientific Center "All-Russia Research and Technological Poultry Institute", Russian Academy of Sciences, Sergiev Posad, Moscow Oblast 141311, Russia
| | - Larisa A Ilina
- Federal State Budgetary Educational Institution of Higher Education "St. Petersburg State Agrarian University", Pushkin, St. Petersburg 196601, Russia
- BIOTROF LLC, Pushkin, St. Petersburg 196602, Russia
| | - Georgiy Yu Laptev
- Federal State Budgetary Educational Institution of Higher Education "St. Petersburg State Agrarian University", Pushkin, St. Petersburg 196601, Russia
- BIOTROF LLC, Pushkin, St. Petersburg 196602, Russia
| | - Elena A Yildirim
- Federal State Budgetary Educational Institution of Higher Education "St. Petersburg State Agrarian University", Pushkin, St. Petersburg 196601, Russia
- BIOTROF LLC, Pushkin, St. Petersburg 196602, Russia
| | | | - Valentina A Filippova
- Federal State Budgetary Educational Institution of Higher Education "St. Petersburg State Agrarian University", Pushkin, St. Petersburg 196601, Russia
- BIOTROF LLC, Pushkin, St. Petersburg 196602, Russia
| | | | - Vladimir I Fisinin
- Federal Scientific Center "All-Russia Research and Technological Poultry Institute", Russian Academy of Sciences, Sergiev Posad, Moscow Oblast 141311, Russia
| | - Ivan I Kochish
- K. I. Skryabin Moscow State Academy of Veterinary Medicine and Biotechnology, Moscow 109472, Russia
| | - Darren K Griffin
- School of Biosciences, University of Kent, Canterbury, Kent CT2 7NJ, United Kingdom
| | - Peter F Surai
- Vitagene and Health Research Centre, Bristol BS4 2RS, United Kingdom
- Department of Microbiology and Biochemistry, Faculty of Veterinary Medicine, Trakia University, 6000 Stara Zagora, Bulgaria
- Department of Animal Nutrition, Faculty of Agricultural and Environmental Sciences, Szent Istvan University, H-2103 Gödöllo, Hungary
| | - Michael N Romanov
- School of Biosciences, University of Kent, Canterbury, Kent CT2 7NJ, United Kingdom
- L. K. Ernst Federal Research Center for Animal Husbandry, Dubrovitsy, Podolsk, Moscow Oblast 142132, Russia
| |
Collapse
|
19
|
Ben-Miled H, Semmar N, Castellanos MS, Ben-Mahrez K, Benoit-Biancamano MO, Réjiba S. Effect of honey bee forage plants in Tunisia on diversity and antibacterial potential of lactic acid bacteria and bifidobacteria from Apis mellifera intermissa and its products. Arch Microbiol 2023; 205:295. [PMID: 37480514 DOI: 10.1007/s00203-023-03630-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 07/01/2023] [Accepted: 07/10/2023] [Indexed: 07/24/2023]
Abstract
Lactic acid bacteria and bifidobacteria (LAB and Bifido), isolated from the gastrointestinal tract of Apis mellifera intermissa (BGIT), honey (H), propolis (P) and bee bread (BB) of hives set in different vegetations (wildflowers, caraway, orange blossom, Marrubium vulgare, Eucalyptus and Erica cinerea), were subjected to analysis of their antibacterial potential. Isolates able to inhibit Staphylococcus aureus were selected and identified with MALDI-TOF MS leading to 154 strains representing 12 LAB and Bifido species. Lactiplantibacillus plantarum, Pediococcus pentosaceus and Enterococcus faecalis were predominantly found in all matrices. BGIT showed the highest LAB and Bifido diversity with exclusive occurrences of five species (including Bifidobacterium asteroides and Limosilactobacillus fermentum). Honey was the second origin harboring an important variety of LAB species of which Apilactobacillus kunkeei and Enterococcus mundtii were characteristic of both H and BGIT. Principal components analysis revealed associations between antibacterial activities of LAB and Bifido, matrices and honey bee forage plants. Inhibition trends of S. aureus and Citrobacter freundii were highlighted with: L. plantarum from BGIT, P, H of bees feeding on E. cinerea; Pediococcus pentosaceus from BGIT, P, BB associated with E. cinerea; and Bifidobacterium asteroides from BGIT/orange blossom system. However, Enterococcus faecium associated with BGIT/Eucalyptus system antagonized Escherichia coli, Klebsiella pneumoniae, Pseudomonas aeruginosa and Acinetobacter baumannii. Our findings highlighted noteworthy effects of bee forage plants on the antibacterial activity of LAB and Bifido. Our approach could be useful to identify multiple conditions promoting antibacterial potency of LAB and Bifido under the combined effects of feeding plants and living matrices.
Collapse
Affiliation(s)
- Houda Ben-Miled
- Biochemistry and Biotechnology Laboratory LR01ES05, Faculty of Sciences of Tunis, University of Tunis El Manar, 2092, Tunis, Tunisia
| | - Nabil Semmar
- Laboratory of Bioinformatics, Biomathematics and Biostatistics (BIMS), Pasteur Institute of Tunis, University of Tunis El Manar, 2092, Tunis, Tunisia
| | - Miguel Sautié Castellanos
- Plateforme IA-Agrosanté, Faculty of Veterinary Medicine, Université de Montréal, 3200 rue Sicotte, Saint-Hyacinthe, QC, J2S 2M2, Canada
| | - Kamel Ben-Mahrez
- Biochemistry and Biotechnology Laboratory LR01ES05, Faculty of Sciences of Tunis, University of Tunis El Manar, 2092, Tunis, Tunisia
| | - Marie-Odile Benoit-Biancamano
- Groupe de Recherche sur les Maladies Infectieuses en Production Animale (GREMIP), Department of Pathology and Microbiology, Faculty of Veterinary Medicine, Université de Montréal, 3200 rue Sicotte, Saint-Hyacinthe, QC, J2S 2M2, Canada
| | - Samia Réjiba
- Biochemistry and Biotechnology Laboratory LR01ES05, Faculty of Sciences of Tunis, University of Tunis El Manar, 2092, Tunis, Tunisia.
- Higher Institute of Biotechnology, Biotechpole of Sidi Thabet, Sidi Thabet, BP-66, 2020, Ariana, Tunis, Tunisia.
- University of Manouba, 2010, Manouba, Tunis, Tunisia.
| |
Collapse
|
20
|
Procaccianti G, Roggiani S, Conti G, Brigidi P, Turroni S, D’Amico F. Bifidobacterium in anticancer immunochemotherapy: friend or foe? MICROBIOME RESEARCH REPORTS 2023; 2:24. [PMID: 38046824 PMCID: PMC10688788 DOI: 10.20517/mrr.2023.23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/08/2023] [Revised: 06/20/2023] [Accepted: 07/03/2023] [Indexed: 12/05/2023]
Abstract
The gut microbiome has received a crescendo of attention in recent years due to myriad influences on human pathophysiology, including cancer. Anticancer therapy research is constantly looking for new hints to improve response to therapy while reducing the risk of relapse. In this scenario, Bifidobacterium, which inhabits the gut microbial ecosystem (especially that of children) and is considered a health-associated microbe, has emerged as a key target to assist anticancer treatments for a better prognosis. However, some researchers have recently hypothesized an unfavorable role of Bifidobacterium spp. in anticancer immunochemotherapy, leading to some confusion in the field. This narrative review summarizes the current knowledge on the role of Bifidobacterium spp. in relation to anticancer treatments, discussing the pros and cons of its presence in the gut microbiome of cancer patients. The current intervention strategies based on the administration of probiotic strains of Bifidobacterium are then discussed. Finally, the need to conduct further studies, especially functional ones, is underlined to provide robust experimental evidence, especially on the underlying molecular mechanisms, and thus resolve the controversies on this microbe for the long-term success of immunochemotherapy.
Collapse
Affiliation(s)
- Giorgia Procaccianti
- Unit of Microbiome Science and Biotechnology, Department of Pharmacy and Biotechnology, University of Bologna, Bologna 40126, Italy
| | - Sara Roggiani
- Unit of Microbiome Science and Biotechnology, Department of Pharmacy and Biotechnology, University of Bologna, Bologna 40126, Italy
- Microbiomics Unit, Department of Medical and Surgical Sciences, University of Bologna, Bologna 40138, Italy
| | - Gabriele Conti
- Unit of Microbiome Science and Biotechnology, Department of Pharmacy and Biotechnology, University of Bologna, Bologna 40126, Italy
- Microbiomics Unit, Department of Medical and Surgical Sciences, University of Bologna, Bologna 40138, Italy
| | - Patrizia Brigidi
- Microbiomics Unit, Department of Medical and Surgical Sciences, University of Bologna, Bologna 40138, Italy
| | - Silvia Turroni
- Unit of Microbiome Science and Biotechnology, Department of Pharmacy and Biotechnology, University of Bologna, Bologna 40126, Italy
| | - Federica D’Amico
- Unit of Microbiome Science and Biotechnology, Department of Pharmacy and Biotechnology, University of Bologna, Bologna 40126, Italy
- Microbiomics Unit, Department of Medical and Surgical Sciences, University of Bologna, Bologna 40138, Italy
| |
Collapse
|
21
|
Gut microbiota affects sensitivity to immune-mediated isoniazid-induced liver injury. Biomed Pharmacother 2023; 160:114400. [PMID: 36805186 DOI: 10.1016/j.biopha.2023.114400] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 02/08/2023] [Accepted: 02/09/2023] [Indexed: 02/19/2023] Open
Abstract
Isoniazid (INH) is a highly effective single and/or combined first-line anti-tuberculosis (anti-TB) therapy drug, and the hepatotoxicity greatly limits its clinical application. INH-induced liver injury (INH-DILI) is a typical immune-mediated idiosyncratic drug-induced liver injury. Existing mechanisms including genetic variations in drug metabolism and immune responses cannot fully explain the differences in susceptibility and sensitivity to INH-DILI, suggesting that other factors may be involved. Accumulating evidence indicates that the development and severity of immune-mediated liver injury is related to gut microbiota. In this study, INH exposure caused liver damage, immune disregulation and microbiota profile alteration. Depletion of gut microbiota ameliorated INH-DILI, and improved INH-DILI-associated immune disorder and inflammatory response. Moreover, hepatotoxicity of INH was ameliorated by fecal microbiota transplantation (FMT) from INH-treated mice. Notably, Bifidobacterium abundance was significantly associated with transaminase levels. In conclusion, our results suggested that the effect of gut microbiota on INH-DILI was related to immunity, and the difference in INH-DILI sensitivity was related to the structure of gut microbiota. Changes in the structure of gut microbiota by continuous exposure of INH resulted in the tolerance to liver injury, and probiotics such as Bifidobacterium might play an important role in INH-DILI and its "adaptation" phenomenon. This work provides novel evidence for elucidating the underlying mechanism of difference in individual's response to INH-DILI and potential approach for intervening anti-TB drug liver injury by modulating gut microbiota.
Collapse
|
22
|
Protective and Therapeutic Capacities of Lactic Acid Bacteria Postmetabolites against Koi Herpesvirus Infection In Vitro. Life (Basel) 2023; 13:life13030739. [PMID: 36983894 PMCID: PMC10054248 DOI: 10.3390/life13030739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 03/01/2023] [Accepted: 03/07/2023] [Indexed: 03/11/2023] Open
Abstract
Background: The accumulation of data on beneficial biological effects of probiotics and their metabolic products favors their potential use in the prevention and treatment of various malaises. Methods: Nine postmetabolites from Lactic acid bacteria (LAB) of human or dairy origin and their antiviral activity were studied using the cytopathic effect inhibition test. The virucidal capacity, their influence on the adsorption stage of Koi herpes virus (KHV) and their preventive role against subsequent viral challenge on intact Common carp brain (CCB) cells were also determined by titration assay. Residual viral infectivity in postmetabolites-treated samples was compared to mock-treated controls and Δlgs were calculated. Results: When administered during KHV replication, the microbial products isolated from Lactiplantibacillus plantarum showed remarkable activity with a selectivity index (SI) between 26.5 and 221.4, as those effects were dependent on the sample-virus incubation time. Postmetabolites from Lactobacillus gasseri and Lactiplantibacillus plantarum also demonstrated significant inhibition of KHV replication with SI of 24 and 16, respectively. The bioactive metabolites isolated from Limosilactobacillus fermentum had a minor effect on the viral replicative cycle. Compounds, produced during the fermentation by lactobacilli, grown on different nutritive media and collected at different time points, significantly inhibited extracellular KHV virions. All investigated postmetabolites remarkably blocked KHV attachment to the host cell (CCB), leading to a drop in viral titers by Δlg = 4.25–5.25, and exerted protective effects on CCB cells before they were subjected to viral infection. Conclusions: Our results open new horizons and promote LAB and their postbiotic products to be used in the prophylaxis and therapy of viral infections.
Collapse
|
23
|
Helmy YA, Taha-Abdelaziz K, Hawwas HAEH, Ghosh S, AlKafaas SS, Moawad MMM, Saied EM, Kassem II, Mawad AMM. Antimicrobial Resistance and Recent Alternatives to Antibiotics for the Control of Bacterial Pathogens with an Emphasis on Foodborne Pathogens. Antibiotics (Basel) 2023; 12:274. [PMID: 36830185 PMCID: PMC9952301 DOI: 10.3390/antibiotics12020274] [Citation(s) in RCA: 32] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Revised: 01/21/2023] [Accepted: 01/27/2023] [Indexed: 01/31/2023] Open
Abstract
Antimicrobial resistance (AMR) is one of the most important global public health problems. The imprudent use of antibiotics in humans and animals has resulted in the emergence of antibiotic-resistant bacteria. The dissemination of these strains and their resistant determinants could endanger antibiotic efficacy. Therefore, there is an urgent need to identify and develop novel strategies to combat antibiotic resistance. This review provides insights into the evolution and the mechanisms of AMR. Additionally, it discusses alternative approaches that might be used to control AMR, including probiotics, prebiotics, antimicrobial peptides, small molecules, organic acids, essential oils, bacteriophage, fecal transplants, and nanoparticles.
Collapse
Affiliation(s)
- Yosra A. Helmy
- Department of Veterinary Science, College of Agriculture, Food and Environment, University of Kentucky, Lexington, KY 40546, USA
- Department of Zoonoses, Faculty of Veterinary Medicine, Suez Canal University, Ismailia 41522, Egypt
| | - Khaled Taha-Abdelaziz
- Department of Animal and Veterinary Sciences, Clemson University, Clemson, SC 29634, USA
| | - Hanan Abd El-Halim Hawwas
- Department of Zoonoses, Faculty of Veterinary Medicine, Suez Canal University, Ismailia 41522, Egypt
| | - Soumya Ghosh
- Department of Genetics, Faculty of Natural and Agricultural Sciences, University of the Free State, Bloemfontein 9301, South Africa
| | - Samar Sami AlKafaas
- Molecular Cell Biology Unit, Division of Biochemistry, Department of Chemistry, Faculty of Science, Tanta University, Tanta 31511, Egypt
| | | | - Essa M. Saied
- Chemistry Department, Faculty of Science, Suez Canal University, Ismailia 41522, Egypt
- Institute for Chemistry, Humboldt Universität zu Berlin, Brook-Taylor-Str. 2, 12489 Berlin, Germany
| | - Issmat I. Kassem
- Centre for Food Safety, Department of Food Science and Technology, University of Georgia, Griffin, GA 30609, USA
| | - Asmaa M. M. Mawad
- Department of Biology, College of Science, Taibah University, Madinah 42317, Saudi Arabia
- Botany and Microbiology Department, Faculty of Science, Assiut University, Assiut 71516, Egypt
| |
Collapse
|
24
|
Teague KD, Tellez-Isaias G, Chai J, Petrone-Garcia V, Vuong CN, Blanch A, Rasmussen SH, Brown K, Zhao J, Rochell SJ. Dietary soy galactooligosaccharides affect the performance, intestinal function, and gut microbiota composition of growing chicks. Poult Sci 2023; 102:102440. [PMID: 36736136 PMCID: PMC10102551 DOI: 10.1016/j.psj.2022.102440] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 12/14/2022] [Accepted: 12/15/2022] [Indexed: 01/07/2023] Open
Abstract
The objective of this experiment was to investigate the effects of the dietary soy galactooligosaccharides (GOS), raffinose and stachyose, on performance, gastrointestinal health, and systemic stress in young broilers. Birds were fed a GOS-devoid diet based on soy protein isolate (SPI) or the SPI diet with 0.9, 1.8, 2.7, or 3.6% added stachyose and raffinose in a ratio of 4:1 at the expense of corn starch. These 5 treatments were administered to 10 replicate cages of 8 birds. Performance was measured weekly and excreta moisture, N retention, apparent metabolizeable energy, and complete blood cell counts were determined at 14 and 21 d. At 21 d, 2 birds per cage were orally gavaged with fluorescein isothiocyanate-dextran (FITC-d) and serum samples were analyzed for FITC-d as a marker of gut leakage. Additionally, intestinal morphology, crop presumptive lactic acid bacteria (LAB) counts, crop and cecal pH, and cecal microbiota via16S rRNA microbial sequencing were evaluated at 21 d. From 0 to 21 d, feed intake increased linearly (P < 0.01) as dietary GOS increased, whereas BWG increased (P < 0.05) quadratically. Feed conversion ratio increased (P < 0.01) linearly as GOS increased. There were linear increases (P < 0.05) in excreta moisture as dietary GOS increased at 14 and 21 d, as well as dose-dependent responses (P < 0.05) in N retention, AME, and AMEn. There was a quadratic increase (P < 0.05) in crop LAB recovery and a linear decrease (P < 0.01) in ceca pH as GOS increased. At 14 d, a linear increase (P < 0.05) in blood heterophil to lymphocyte ratio was observed as dietary GOS increased. Serum concentrations of FITC-d increased quadratically (P < 0.01) to dietary GOS. Increasing levels of GOS influenced alpha and beta diversities and composition of gut microbiota, including the abundance of Ruminococcus and Bifidobacterium. Results from this trial indicate that soy-derived GOS exert dose-dependent effects on nutrient utilization and intestinal health in young broilers.
Collapse
Affiliation(s)
- K D Teague
- Division of Agriculture, Department of Poultry Science, University of Arkansas, Fayetteville, AR, USA
| | - G Tellez-Isaias
- Division of Agriculture, Department of Poultry Science, University of Arkansas, Fayetteville, AR, USA
| | - Jianmin Chai
- Division of Agriculture, Department of Animal Science, University of Arkansas, Fayetteville, AR, USA; Guangdong Provincial Key Laboratory of Animal Molecular Design and Precise Breeding, School of Life Science and Engineering, Foshan University, Foshan, China
| | - V Petrone-Garcia
- Faculty of Higher Studies Cuautitlan, National Autonomous University of Mexico, Cuautitlan Izcalli, State of Mexico, Mexico
| | - C N Vuong
- Division of Agriculture, Department of Poultry Science, University of Arkansas, Fayetteville, AR, USA
| | - A Blanch
- Hamlet Protein A/S, DK-8700 Horsens, Denmark
| | | | - K Brown
- Hamlet Protein A/S, DK-8700 Horsens, Denmark
| | - Jiangchao Zhao
- Division of Agriculture, Department of Animal Science, University of Arkansas, Fayetteville, AR, USA
| | - S J Rochell
- Division of Agriculture, Department of Poultry Science, University of Arkansas, Fayetteville, AR, USA; Department of Poultry Science, Auburn University, Auburn, AL, USA.
| |
Collapse
|
25
|
He Y, Liang J, Liu Y, Zhou X, Peng C, Long C, Huang P, Feng J, Zhang Z. Combined supplementation with Lactobacillus sp. and Bifidobacterium thermacidophilum isolated from Tibetan pigs improves growth performance, immunity, and microbiota composition in weaned piglets. J Anim Sci 2023; 101:skad220. [PMID: 37358243 PMCID: PMC10347973 DOI: 10.1093/jas/skad220] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 06/23/2023] [Indexed: 06/27/2023] Open
Abstract
Probiotics, such as Lactobacillus and Bifidobacterium, promote growth in piglets by modulating gut microbiota composition and improving the host immune system. A strain of Lactobacillus sp. and Bifidobacterium thermacidophilum were previously isolated from fresh feces of Tibetan pigs. The effects of these isolated strains on growth performance, intestinal morphology, immunity, microbiota composition, and their metabolites were evaluated in weaned piglets. Thirty crossbred piglets were selected and fed either a basal diet (CON), a basal diet supplemented with aureomycin (ANT), or a basal diet supplemented with Lactobacillus sp. and B. thermacidophilum (LB) for 28 d. The piglets in the ANT and LB groups had significantly higher body weight gain than those in the CON group (P < 0.05). Piglets in the ANT and LB groups had regularly arranged villi and microvilli in the small intestine. Furthermore, they had improved immune function, as indicated by decreased serum concentrations of inflammatory cytokines (P < 0.05), improved components of immune cells in the blood, mesenteric lymph nodes, and spleen. Additionally, metagenomic sequencing indicated a significant shift in cecal bacterial composition and alterations in microbiota functional profiles following Lactobacillus sp. and B. thermacidophilum supplementation. Metabolomic results revealed that the metabolites were also altered, and Kyoto Encyclopedia of Genes and Genomes analysis revealed that several significantly altered metabolites were enriched in glycerophospholipid and cholesterol metabolism (P < 0.05). Furthermore, correlation analysis showed that several bacterial members were closely related to the alterations in metabolites, including Bacteroides sp., which were negatively correlated with triglyceride (16:0/18:0/20:4[5Z,8Z,11Z,14Z]), the metabolite that owned the highest variable importance of projection scores. Collectively, our findings suggest that combined supplementation with Lactobacillus sp. and B. thermacidophilum significantly improved the growth performance, immunity, and microbiota composition in weaned piglets, making them prospective alternatives to antibiotics in swine production.
Collapse
Affiliation(s)
- Yiwen He
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China
- Hunan Provincial Key Laboratory of Animal Intestinal Function and Regulation, College of Life Sciences, Hunan International Joint Laboratory of Animal Intestinal Ecology and Health, Hunan Normal University, Changsha 410081, China
| | - Jing Liang
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China
| | - Yonghui Liu
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China
- Hunan Provincial Key Laboratory of Animal Intestinal Function and Regulation, College of Life Sciences, Hunan International Joint Laboratory of Animal Intestinal Ecology and Health, Hunan Normal University, Changsha 410081, China
| | - Xihong Zhou
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China
| | - Can Peng
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China
| | - Ciming Long
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China
| | - Pan Huang
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China
| | - Jie Feng
- Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
| | - Zhigang Zhang
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, School of Life Sciences, Yunnan University, Kunming 650091, China
| |
Collapse
|
26
|
Budden KF, Gellatly SL, Vaughan A, Amorim N, Horvat JC, Hansbro NG, Wood DLA, Hugenholtz P, Dennis PG, Wark PAB, Hansbro PM. Probiotic Bifidobacterium longum subsp. longum Protects against Cigarette Smoke-Induced Inflammation in Mice. Int J Mol Sci 2022; 24:252. [PMID: 36613693 PMCID: PMC9820259 DOI: 10.3390/ijms24010252] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 12/18/2022] [Accepted: 12/20/2022] [Indexed: 12/25/2022] Open
Abstract
Bifidobacterium are prominent gut commensals that produce the short-chain fatty acid (SCFA) acetate, and they are often used as probiotics. Connections between the gut and the lung, termed the gut-lung axis, are regulated by the microbiome. The gut-lung axis is increasingly implicated in cigarette smoke-induced diseases, and cigarette smoke exposure has been associated with depletion of Bifidobacterium species. In this study, we assessed the impact of acetate-producing Bifidobacterium longum subsp. longum (WT) and a mutant strain with an impaired acetate production capacity (MUT) on cigarette smoke-induced inflammation. The mice were treated with WT or MUT B. longum subsp. longum and exposed to cigarette smoke for 8 weeks before assessments of lung inflammation, lung tissue gene expression and cecal SCFAs were performed. Both strains of B. longum subsp. longum reduced lung inflammation, inflammatory cytokine expression and adhesion factor expression and alleviated cigarette smoke-induced depletion in caecum butyrate. Thus, the probiotic administration of B. longum subsp. longum, irrespective of its acetate-producing capacity, alleviated cigarette smoke-induced inflammation and the depletion of cecal butyrate levels.
Collapse
Affiliation(s)
- Kurtis F. Budden
- Priority Research Centre for Healthy Lungs and Immune Health Research Program, Hunter Medical Research Institute and The University of Newcastle, Callaghan, NSW 2308, Australia
| | - Shaan L. Gellatly
- Priority Research Centre for Healthy Lungs and Immune Health Research Program, Hunter Medical Research Institute and The University of Newcastle, Callaghan, NSW 2308, Australia
| | - Annalicia Vaughan
- Centre for Inflammation, School of Life Sciences, Faculty of Science, Centenary Institute and University of Technology Sydney, Sydney, NSW 2007, Australia
| | - Nadia Amorim
- Centre for Inflammation, School of Life Sciences, Faculty of Science, Centenary Institute and University of Technology Sydney, Sydney, NSW 2007, Australia
| | - Jay C. Horvat
- Priority Research Centre for Healthy Lungs and Immune Health Research Program, Hunter Medical Research Institute and The University of Newcastle, Callaghan, NSW 2308, Australia
| | - Nicole G. Hansbro
- Centre for Inflammation, School of Life Sciences, Faculty of Science, Centenary Institute and University of Technology Sydney, Sydney, NSW 2007, Australia
| | - David L. A. Wood
- Australian Centre for Ecogenomics, School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD 4102, Australia
| | - Philip Hugenholtz
- Australian Centre for Ecogenomics, School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD 4102, Australia
| | - Paul G. Dennis
- School of Earth and Environmental Sciences, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Peter A. B. Wark
- Priority Research Centre for Healthy Lungs and Immune Health Research Program, Hunter Medical Research Institute and The University of Newcastle, Callaghan, NSW 2308, Australia
| | - Philip M. Hansbro
- Priority Research Centre for Healthy Lungs and Immune Health Research Program, Hunter Medical Research Institute and The University of Newcastle, Callaghan, NSW 2308, Australia
- Centre for Inflammation, School of Life Sciences, Faculty of Science, Centenary Institute and University of Technology Sydney, Sydney, NSW 2007, Australia
| |
Collapse
|
27
|
Kim SH, Yehuala GA, Bang WY, Yang J, Jung YH, Park MK. Safety Evaluation of Bacillus subtilis IDCC1101, Newly Isolated from Cheonggukjang, for Industrial Applications. Microorganisms 2022; 10:microorganisms10122494. [PMID: 36557747 PMCID: PMC9784242 DOI: 10.3390/microorganisms10122494] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 12/11/2022] [Accepted: 12/14/2022] [Indexed: 12/23/2022] Open
Abstract
The present study aimed to evaluate the safety of Bacillus subtilis (BS) IDCC1101, newly isolated from Cheonggukjang in Korea. Genome sequencing of BS IDCC1101 was performed to investigate the presence of secondary metabolites, virulence, antibiotic resistance, and mobile elements. Its phenotypic safety analyses included antibiotic susceptibility, enzyme activity, carbohydrate utilization, production of biogenic amines (BAs) and D-/L-lactate, hemolytic activity, and toxicities in HaCaT cells and rats. The genome of BS IDCC1101 consisted of 4,118,950 bp with 3077 functional genes. Among them, antimicrobial and antifungal secondary metabolites were found, such as fengycin, bacillibactin, and bacilysin. Antibiotic resistance and virulence genes did not exhibit transferability since they did not overlap with mobile elements in the genome. BS IDCC1101 was susceptible to almost all antibiotics suggested for assessment of BS's antibiotic susceptibility by EFSA guidelines, except for streptomycin. BS IDCC1101 showed the utilization of a wide range of 27 carbohydrates, as well as enzyme activities such as alkaline phosphatase, esterase, esterase lipase, naphthol-AS-BI-phosphohydrolase, α-galactosidase, β-galactosidase, α-glucosidase, and β-glucosidase activities. Additionally, BS IDCC1101 did not exhibit the production of D-/L-lactate and hemolytic activities. Its toxicity in HaCaT cells and rats was also not detected. Thus, these genotypic and phenotypic findings indicate that BS IDCC1101 can be safely used for industrial applications.
Collapse
Affiliation(s)
- Su-Hyeon Kim
- School of Food Science and Biotechnology, Kyungpook National University, Daegu 41566, Republic of Korea
- Food and Bio-Industry Research Institute, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Gashaw Assefa Yehuala
- School of Food Science and Biotechnology, Kyungpook National University, Daegu 41566, Republic of Korea
- Department of Food Engineering, College of Biological and Chemical Engineering, Addis Ababa Science and Technology University, Addis Ababa 16417, Ethiopia
| | - Won Yeong Bang
- Ildong Bioscience, Pyeongtaek-si 17957, Republic of Korea
| | - Jungwoo Yang
- Ildong Bioscience, Pyeongtaek-si 17957, Republic of Korea
| | - Young Hoon Jung
- School of Food Science and Biotechnology, Kyungpook National University, Daegu 41566, Republic of Korea
- Food and Bio-Industry Research Institute, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Mi-Kyung Park
- School of Food Science and Biotechnology, Kyungpook National University, Daegu 41566, Republic of Korea
- Food and Bio-Industry Research Institute, Kyungpook National University, Daegu 41566, Republic of Korea
- Correspondence: ; Tel.: +82-53-950-5776
| |
Collapse
|
28
|
Yan J, Zhao J, Ning X, Qin Y, Xing Y, Wang Y, Jia Q, Huang B, Ma R, Lei C, Zhou M, Yu Z, Zhang Y, Guo WF, Sun S. Alterations of the gut microbiota in patients with immunoglobulin light chain amyloidosis. Front Immunol 2022; 13:973760. [PMID: 36341382 PMCID: PMC9628213 DOI: 10.3389/fimmu.2022.973760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 09/28/2022] [Indexed: 11/13/2022] Open
Abstract
Background Emerging evidence revealed that gut microbial dysbiosis is implicated in the development of plasma cell dyscrasias and amyloid deposition diseases, but no data are available on the relationship between gut microbiota and immunoglobulin light chain (AL) amyloidosis. Methods To characterize the gut microbiota in patients with AL amyloidosis, we collected fecal samples from patients with AL amyloidosis (n=27) and age-, gender-, and BMI-matched healthy controls (n=27), and conducted 16S rRNA MiSeq sequencing and amplicon sequence variants (ASV)-based analysis. Results There were significant differences in gut microbial communities between the two groups. At the phylum level, the abundance of Actinobacteriota and Verrucomicrobiota was significantly higher, while Bacteroidota reduced remarkably in patients with AL amyloidosis. At the genus level, 17 genera, including Bifidobacterium, Akkermansia, and Streptococcus were enriched, while only 4 genera including Faecalibacterium, Tyzzerella, Pseudomonas, and Anaerostignum decreased evidently in patients with AL amyloidosis. Notably, 5 optimal ASV-based microbial markers were identified as the diagnostic model of AL amyloidosis and the AUC value of the train set and the test set was 0.8549 (95% CI 0.7310-0.9789) and 0.8025 (95% CI 0.5771-1), respectively. With a median follow-up of 19.0 months, further subgroup analysis also demonstrated some key gut microbial markers were related to disease severity, treatment response, and even prognosis of patients with AL amyloidosis. Conclusions For the first time, we demonstrated the alterations of gut microbiota in AL amyloidosis and successfully established and validated the microbial-based diagnostic model, which boosted more studies about microbe-based strategies for diagnosis and treatment in patients with AL amyloidosis in the future.
Collapse
Affiliation(s)
- Jipeng Yan
- Department of Nephrology, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Jin Zhao
- Department of Nephrology, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Xiaoxuan Ning
- Department of Geriatrics, Xijing Hospital, Fourth Military Medical University, Xi’an, China
- *Correspondence: Shiren Sun, ; Xiaoxuan Ning,
| | - Yunlong Qin
- Department of Nephrology, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Yan Xing
- Department of Nephrology, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Yuwei Wang
- Department of Nephrology, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Qing Jia
- Department of Nephrology, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Boyong Huang
- Department of Geriatrics, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Rui Ma
- Department of Geriatrics, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Changhui Lei
- Xijing Hypertrophic Cardiomyopathy Center, Department of Ultrasound, Xijing Hospital, Xi’an, China
| | - Meilan Zhou
- Department of Nephrology, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Zixian Yu
- Department of Nephrology, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Yumeng Zhang
- Department of Nephrology, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Wei-Feng Guo
- School of Electrical Engineering, Zhengzhou University, Zhengzhou, China
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Shiren Sun
- Department of Nephrology, Xijing Hospital, Fourth Military Medical University, Xi’an, China
- *Correspondence: Shiren Sun, ; Xiaoxuan Ning,
| |
Collapse
|
29
|
Kono K, Murakami Y, Ebara A, Okuma K, Tokuno H, Odachi A, Ogasawara K, Hidaka E, Mori T, Satoh K, Kimoto S, Masuyama H, Takeda M, Managi S. Fluctuations in Intestinal Microbiota Following Ingestion of Natto Powder Containing Bacillus subtilis var. natto SONOMONO Spores: Considerations Using a Large-Scale Intestinal Microflora Database. Nutrients 2022; 14:nu14183839. [PMID: 36145213 PMCID: PMC9505718 DOI: 10.3390/nu14183839] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 09/01/2022] [Accepted: 09/08/2022] [Indexed: 11/16/2022] Open
Abstract
Improving the intestinal microbiota using probiotics, prebiotics, and synbiotics has attracted attention as a method of disease prevention and treatment. This is the first study to discuss the effects of food intake on the intestinal microbiota using a large Japanese intestinal microbiota database. Here, as a case study, we determined changes in the intestinal microbiota caused by ingestion of a processed natto food containing B. subtilisvar. natto SONOMONO spores, SONOMONO NATTO POWDER CAPSULESTM, by analyzing 16S rRNA sequence data generated using next-generation sequencing techniques. The results showed that the relative abundance of Bifidobacterium and Blautia as well as the relative abundance of Bifidobacterium were increased in males and females in the ingesting group, respectively. Additionally, the effects of SONOMONO NATTO POWDER CAPSULESTM intake on Bifidobacterium and Blautia abundance depended on the relative abundance of Bifidobacterium at baseline. Finally, analysis of a large Japanese intestinal microbiota database suggested that the bacterial genera that fluctuated with the ingestion of SONOMONO NATTO POWDER CAPSULESTM may be associated with lifestyle-related diseases such as diabetes.
Collapse
Affiliation(s)
- Kanako Kono
- Symbiosis Solutions Inc., Tokyo 101-0064, Japan
| | - Yasufumi Murakami
- Department of Biological Science and Technology, Tokyo University of Science, Tokyo 162-8601, Japan
| | - Aya Ebara
- Symbiosis Solutions Inc., Tokyo 101-0064, Japan
| | - Kana Okuma
- Symbiosis Solutions Inc., Tokyo 101-0064, Japan
| | | | | | - Kazuya Ogasawara
- Sonomono Inc., Fukuoka 810-0023, Japan
- Department of Agricultural and Resource Economics, Faculty of Agriculture, Kyushu University, Fukuoka 819-0395, Japan
| | | | - Teruaki Mori
- National Hospital Organization Nishi-Beppu National Hospital, Oita 874-0840, Japan
| | - Kazuko Satoh
- Department of Nursing, Faculty of Medicine, Oita University, Oita 879-5593, Japan
| | | | - Hiroaki Masuyama
- Symbiosis Solutions Inc., Tokyo 101-0064, Japan
- Correspondence: ; Tel.: +81-3-6275-0878
| | - Midori Takeda
- Urban Institute & Department of Civil Engineering, Kyushu University, Fukuoka 819-0395, Japan
| | - Shunsuke Managi
- Urban Institute & Department of Civil Engineering, Kyushu University, Fukuoka 819-0395, Japan
| |
Collapse
|
30
|
Zhou K, Deng N, Yi X, Cai Y, Peng M, Xiao N. Baohe pill decoction for diarrhea induced by high-fat and high-protein diet is associated with the structure of lactase-producing bacterial community. Front Cell Infect Microbiol 2022; 12:1004845. [PMID: 36093186 PMCID: PMC9458856 DOI: 10.3389/fcimb.2022.1004845] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 08/10/2022] [Indexed: 01/30/2023] Open
Abstract
Background This study investigated the effects of Baohe pill decoction on the diversity and community composition of lactase-producing bacteria in the intestinal contents of mice with diarrhea induced by high-fat and high-protein diet, which provided an experimental basis for the study on the therapeutic mechanism of Baohe pill decoction. Materials and methods The Traditional Chinese Medicine Systems Pharmacology (TCMSP), DisGeNET, UniProt, National Center for Biotechnology Information (NCBI), and GeneCards databases were used to collect the potential targets with active ingredients of Baohe pill decoction, diarrhea, and lactase, and then construct correlation networks. Fifteen Kunming mice were randomly divided into the control group (CN), natural recovery group (NR), and Baohe pill decoction treatment group (BHP), with five mice in each group. After constructing a mouse diarrhea model by HFHPD induction, BHP was gavaged with Baohe pill decoction, and the other groups were gavaged with distilled water of equal. The intestinal contents were collected from ileal to jejunal and analyzed using metagenomic sequencing to characterize the intestinal content of lactase-producing bacteria in mice. Results The core active ingredients related to diarrhea in Baohe pill decoction were quercetin, luteolin, kaempferol, forsythin, and wogonin. And there was no intersection between the potential targets with the active ingredient of Baohe pill, lactase, and diarrhea. After the intervention of Baohe pill decoction, the Observed species, Chao1 index, and Operational Taxonomic Units (OTU) number increased in BHP (P > 0.05), while the Pielous evenness and Shannon index decreased (P > 0.05). In Beta diversity, the community structure of the NR was significantly different from CN and BHP (P < 0.05), and the community structure of the CN was not significant difference from BHP (P > 0.05). Compared to NR, the relative abundance of Bifidobacterium and Amycolatopsis increased, while the relative abundance of Lachnoclostridium, Sinorhizobium, Cedecea, and Escherichia decreased in BHP, but none of the significant differences (P > 0.05). Conclusion The therapeutic effect of Baohe pill decoction on diarrhea induced by HFHPD does not appear to involve the body’s lactase gene targets directly, but is associated with the change of the construction of lactase-producing bacterial communities.
Collapse
Affiliation(s)
- Kang Zhou
- College of Pharmacy, Hunan University of Chinese Medicine, Changsha, China
| | - Na Deng
- College of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China
| | - Xin Yi
- College of Pharmacy, Hunan University of Chinese Medicine, Changsha, China
| | - Ying Cai
- College of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China
| | - Maijiao Peng
- College of Pharmacy, Hunan University of Chinese Medicine, Changsha, China
- *Correspondence: Maijiao Peng, ; Nenqun Xiao,
| | - Nenqun Xiao
- College of Pharmacy, Hunan University of Chinese Medicine, Changsha, China
- *Correspondence: Maijiao Peng, ; Nenqun Xiao,
| |
Collapse
|
31
|
Hazan S. Microbiome-Based Hypothesis on Ivermectin's Mechanism in COVID-19: Ivermectin Feeds Bifidobacteria to Boost Immunity. Front Microbiol 2022; 13:952321. [PMID: 35898916 PMCID: PMC9309549 DOI: 10.3389/fmicb.2022.952321] [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: 05/25/2022] [Accepted: 06/10/2022] [Indexed: 01/24/2023] Open
Abstract
Ivermectin is an anti-parasitic agent that has gained attention as a potential COVID-19 therapeutic. It is a compound of the type Avermectin, which is a fermented by-product of Streptomyces avermitilis. Bifidobacterium is a member of the same phylum as Streptomyces spp., suggesting it may have a symbiotic relation with Streptomyces. Decreased Bifidobacterium levels are observed in COVID-19 susceptibility states, including old age, autoimmune disorder, and obesity. We hypothesize that Ivermectin, as a by-product of Streptomyces fermentation, is capable of feeding Bifidobacterium, thereby possibly preventing against COVID-19 susceptibilities. Moreover, Bifidobacterium may be capable of boosting natural immunity, offering more direct COVID-19 protection. These data concord with our study, as well as others, that show Ivermectin protects against COVID-19.
Collapse
|
32
|
Liu Z, Wen S, Wu G, Wu H. Heterologous expression and characterization of Anaeromyces robustus xylanase and its use in bread making. Eur Food Res Technol 2022. [DOI: 10.1007/s00217-022-04047-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
|
33
|
Van Daele E, Kamphorst K, Vlieger AM, Hermes G, Milani C, Ventura M, Belzer C, Smidt H, van Elburg RM, Knol J. Effect of antibiotics in the first week of life on faecal microbiota development. Arch Dis Child Fetal Neonatal Ed 2022; 107:fetalneonatal-2021-322861. [PMID: 35534183 PMCID: PMC9606546 DOI: 10.1136/archdischild-2021-322861] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 03/17/2022] [Indexed: 12/15/2022]
Abstract
BACKGROUND Infants are frequently exposed to antibiotics (AB) in the first week of life for suspected bacterial infections. Little is known about the effect of AB on the developing intestinal microbiota. Therefore, we studied intestinal microbiota development with and without AB exposure in the first week of life in term born infants. METHODS We analysed the faecal microbiota from birth until 2.5 years of age by 16S rRNA gene amplicon sequencing in a cohort with 56 term born infants, exposed to AB in the first week of life (AB+) (AB for 2-3 days (AB2, n=20), AB for 7 days (AB7, n=36)), compared with 126 healthy controls (AB-). The effects of AB and duration were examined in relation to delivery and feeding mode. RESULTS AB+ was associated with significantly increased relative abundance of Enterobacteriaceae at 3 weeks and 1 year and a decrease of Bifidobacteriaceae, from 1 week until 3 months of age only in vaginally delivered, but not in C-section born infants. Similar deviations were noted in AB7, but not in AB2. After AB, breastfed infants had lower relative abundance of potentially pathogenic Enterobacteriaceae compared with formula fed infants and recovered 2 weeks faster towards controls. CONCLUSIONS AB exposure in the first week of life alters faecal microbiota development with deviations in the relative abundance of individual taxa until 1 year of age. These alterations can have long-term health consequences, which emphasises the need for future studies aiming at restoring intestinal microbiota after AB administration.
Collapse
Affiliation(s)
- Emmy Van Daele
- Laboratory of Microbiology, Wageningen University & Research, Wageningen, The Netherlands
| | - Kim Kamphorst
- Pediatrics, Amsterdam Gastroenterology, Metabolism & Nutrition, Amsterdam Reproduction & Development, Amsterdam UMC Locatie AMC, Amsterdam, The Netherlands
- Pediatrics, St. Antonius Hospital, Nieuwegein, The Netherlands
| | - Arine M Vlieger
- Pediatrics, St. Antonius Hospital, Nieuwegein, The Netherlands
| | - Gerben Hermes
- Laboratory of Microbiology, Wageningen University & Research, Wageningen, The Netherlands
| | - Christian Milani
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma Department of Chemical Life Sciences and Environmental Sustainability, Parma, Emilia-Romagna, Italy
- Interdepartmental Research Centre "Microbiome Research Hub", University of Parma, Parma, Emilia-Romagna, Italy
| | - Marco Ventura
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma Department of Chemical Life Sciences and Environmental Sustainability, Parma, Emilia-Romagna, Italy
- Interdepartmental Research Centre "Microbiome Research Hub", University of Parma, Parma, Emilia-Romagna, Italy
| | - Clara Belzer
- Laboratory of Microbiology, Wageningen University & Research, Wageningen, The Netherlands
| | - Hauke Smidt
- Laboratory of Microbiology, Wageningen University & Research, Wageningen, The Netherlands
| | - Ruurd M van Elburg
- Pediatrics, Amsterdam Gastroenterology, Metabolism & Nutrition, Amsterdam Reproduction & Development Amsterdam, Amsterdam UMC Locatie AMC, Amsterdam, North Holland, The Netherlands
| | - Jan Knol
- Laboratory of Microbiology, Wageningen University & Research, Wageningen, The Netherlands
- Nutricia Research BV, Utrecht, The Netherlands
| |
Collapse
|
34
|
[Research progress of antibacterial modification of orthopaedic implants surface]. ZHONGGUO XIU FU CHONG JIAN WAI KE ZA ZHI = ZHONGGUO XIUFU CHONGJIAN WAIKE ZAZHI = CHINESE JOURNAL OF REPARATIVE AND RECONSTRUCTIVE SURGERY 2022; 36:511-516. [PMID: 35426294 PMCID: PMC9011072 DOI: 10.7507/1002-1892.202112109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
OBJECTIVE To summarize the related research progress of antibacterial modification of orthopaedic implants surface in recent years. METHODS The domestic and foreign related literature in recent years was extensively consulted, the research progress on antibacterial modification of orthopaedic implants surface was discussed from two aspects of characteristics of infection in orthopedic implants and surface anti-infection modification. RESULTS The orthopaedic implants infections are mainly related to aspects of bacterial adhesion, decreased host immunity, and surface biofilm formation. At present, the main antimicrobial coating methods of orthopaedic implants are antibacterial adhesion coating, antibiotic coating, inorganic antimicrobial coating, composite antimicrobial coating, nitric oxide coating, immunomodulation, three-dimensional printing, polymer antimicrobial coating, and "smart" coating. CONCLUSION The above-mentioned antibacterial coating methods of orthopedic implants can not only inhibit bacterial adhesion, but also solve the problems of low immunity and biofilm formation. However, its mechanism of action and modification are still controversial and require further research.
Collapse
|
35
|
Athalye-Jape G, Esvaran M, Patole S, Simmer K, Nathan E, Doherty D, Keil A, Rao S, Chen L, Chandrasekaran L, Kok C, Schuster S, Conway P. Effect of single versus multistrain probiotic in extremely preterm infants: a randomised trial. BMJ Open Gastroenterol 2022; 9:bmjgast-2021-000811. [PMID: 35185013 PMCID: PMC8860036 DOI: 10.1136/bmjgast-2021-000811] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 01/12/2022] [Indexed: 11/03/2022] Open
Abstract
OBJECTIVE Evidence indicates that multistrain probiotics benefit preterm infants more than single-strain (SS) probiotics. We assessed the effects of SS versus triple-strain (TS) probiotic supplementation (PS) in extremely preterm (EP) infants. DESIGN EP infants (gestational age (GA) <28 weeks) were randomly allocated to TS or SS probiotic, assuring blinding. Reference (REF) group was EP infants in the placebo arm of our previous probiotic trial. PS was commenced with feeds and continued until 37 weeks' corrected GA. Primary outcome was time to full feed (TFF: 150 mL/kg/day). Secondary outcomes included short-chain fatty acids and faecal microbiota collected at T1 (first week) and T2 (after 3 weeks of PS) using 16S ribosomal RNA gene sequencing. RESULTS 173 EP (SS: 86, TS: 87) neonates with similar GA and birth weight (BW) were randomised. Median TFF was comparable (11 (IQR 8-16) vs 10 (IQR 8-16) days, p=0.92). Faecal propionate (SS, p<0.001, and TS, p=0.0009) and butyrate levels (TS, p=0.029) were significantly raised in T2 versus T1 samples. Secondary clinical outcomes were comparable. At T2, alpha diversity was comparable (p>0.05) between groups, whereas beta-diversity analysis revealed significant differences between PS and REF groups (both p=0.001). Actinobacteria were higher (both p<0.01), and Proteobacteria, Firmicutes and Bacteroidetes were lower in PS versus REF. Gammaproteobacteria, Clostridia and Negativicutes were lower in both PS versus REF. CONCLUSION TFF in EP infants was similar between SS and TS probiotics. Both probiotics were effective in reducing dysbiosis (higher bifidobacteria and lower Gammaproteobacteria). Long-term significance of increased propionate and butyrate needs further studies. TRIAL REGISTRATION NUMBER ACTRN 12615000940572.
Collapse
Affiliation(s)
- Gayatri Athalye-Jape
- Neonatology directorate, King Edward Memorial Hospital for Women Perth, Subiaco, Western Australia, Australia
| | - Meera Esvaran
- Faculty of Science, University of New South Wales, Sydney, New South Wales, Australia
| | - Sanjay Patole
- Neonatal Clinical Care Unit, King Edward Memorial Hospital, Subiaco, Western Australia, Australia
| | - Karen Simmer
- Neonatal Clinical Care Unit, King Edward Memorial Hospital, Subiaco, Western Australia, Australia
| | - Elizabeth Nathan
- Biostatistics, Women and Infants Research Foundation Western Australia, Subiaco, Western Australia, Australia
| | - Dorota Doherty
- Biostatistics, Women and Infants Research Foundation Western Australia, Subiaco, Western Australia, Australia
| | - Anthony Keil
- Microbiology, PathWest Laboratory Medicine Western Australia, Nedlands, Western Australia, Australia
| | - Shripada Rao
- Neonatal Clinical Care Unit, Perth Children's Hospital, Nedlands, Western Australia, Australia
| | - Liwei Chen
- Genomics and Bioinformatics, Nanyang Technological University, Singapore
| | | | - Chooi Kok
- Neonatal Clinical Care Unit, King Edward Memorial Hospital, Subiaco, Western Australia, Australia
| | - Stephan Schuster
- Genomics and Bioinformatics, Nanyang Technological University, Singapore
| | - Patricia Conway
- Genomics and Bioinformatics, Nanyang Technological University, Singapore
| |
Collapse
|
36
|
Kober AKMH, Riaz Rajoka MS, Mehwish HM, Villena J, Kitazawa H. Immunomodulation Potential of Probiotics: A Novel Strategy for Improving Livestock Health, Immunity, and Productivity. Microorganisms 2022; 10:microorganisms10020388. [PMID: 35208843 PMCID: PMC8878146 DOI: 10.3390/microorganisms10020388] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 01/23/2022] [Accepted: 02/03/2022] [Indexed: 02/06/2023] Open
Abstract
Over the past decade, the use of probiotics as feed supplements in animal production has increased considerably due to the ban on antibiotic growth promoters in livestock. This review provides an overview of the current situation, limitation, and prospects for probiotic formulations applied to livestock. Recently, the use of probiotics in livestock has been suggested to significantly improve their health, immunity, growth performance, nutritional digestibility, and intestinal microbial balance. Furthermore, it was reported that the use of probiotics in animals was helpful in equilibrating their beneficial microbial population and microbial turnover via stimulating the host immune response through specific secretions and competitive exclusion of potentially pathogenic bacteria in the digestive tract. Recently, there has been great interest in the understanding of probiotics targeted diet and its ability to compete with harmful microbes and acquire their niches. Therefore, the present review explores the most commonly used probiotic formulations in livestock feed and their effect on animal health. In summary, this article provides an in-depth knowledge about the formulation of probiotics as a step toward a better alternative to antibiotic healthy growth strategies.
Collapse
Affiliation(s)
- A. K. M. Humayun Kober
- Laboratory of Animal Food Function, Graduate School of Agricultural Science, Tohoku University, Sendai 980-8572, Japan; (M.S.R.R.); (H.M.M.)
- Livestock Immunology Unit, International Education and Research Centre for Food and Agricultural Immunology, Graduate School of Agricultural Science, Tohoku University, Sendai 980-8572, Japan
- Department of Dairy and Poultry Science, Chittagong Veterinary and Animal Sciences University, Khulshi, Chittagong 4225, Bangladesh
- Correspondence: or (A.K.M.H.K.); (H.K.); Tel.: +880-1712-164794 (A.K.M.H.K.); +81-22-757-4372 (H.K.)
| | - Muhammad Shahid Riaz Rajoka
- Laboratory of Animal Food Function, Graduate School of Agricultural Science, Tohoku University, Sendai 980-8572, Japan; (M.S.R.R.); (H.M.M.)
- Livestock Immunology Unit, International Education and Research Centre for Food and Agricultural Immunology, Graduate School of Agricultural Science, Tohoku University, Sendai 980-8572, Japan
| | - Hafiza Mahreen Mehwish
- Laboratory of Animal Food Function, Graduate School of Agricultural Science, Tohoku University, Sendai 980-8572, Japan; (M.S.R.R.); (H.M.M.)
- Livestock Immunology Unit, International Education and Research Centre for Food and Agricultural Immunology, Graduate School of Agricultural Science, Tohoku University, Sendai 980-8572, Japan
| | - Julio Villena
- Laboratory of Immunobiotechnology, Reference Centre for Lactobacilli (CERELA-CONICET), Tucuman 4000, Argentina;
| | - Haruki Kitazawa
- Laboratory of Animal Food Function, Graduate School of Agricultural Science, Tohoku University, Sendai 980-8572, Japan; (M.S.R.R.); (H.M.M.)
- Livestock Immunology Unit, International Education and Research Centre for Food and Agricultural Immunology, Graduate School of Agricultural Science, Tohoku University, Sendai 980-8572, Japan
- Correspondence: or (A.K.M.H.K.); (H.K.); Tel.: +880-1712-164794 (A.K.M.H.K.); +81-22-757-4372 (H.K.)
| |
Collapse
|
37
|
Lee H, Li Z, Christensen B, Peng Y, Li X, Hernell O, Lönnerdal B, Slupsky CM. Metabolic Phenotype and Microbiome of Infants Fed Formula Containing Lactobacillus paracasei Strain F-19. Front Pediatr 2022; 10:856951. [PMID: 35558362 PMCID: PMC9087039 DOI: 10.3389/fped.2022.856951] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Accepted: 03/16/2022] [Indexed: 12/24/2022] Open
Abstract
Early childhood nutrition drives the development of the gut microbiota. In contrast to breastfeeding, feeding infant formula has been shown to impact both the gut microbiota and the serum metabolome toward a more unfavorable state. It is thought that probiotics may alter the gut microbiota and hence create a more favorable metabolic outcome. To investigate the impact of supplementation with Lactobacillus paracasei spp. paracasei strain F-19 on the intestinal microbiota and the serum metabolome, infants were fed a formula containing L. paracasei F19 (F19) and compared to a cohort of infants fed the same standard formula without the probiotic (SF) and a breast-fed reference group (BF). The microbiome, as well as serum metabolome, were compared amongst groups. Consumption of L. paracasei F19 resulted in lower community diversity of the gut microbiome relative to the SF group that made it more similar to the BF group at the end of the intervention (4 months). It also significantly increased lactobacilli and tended to increase bifidobacteria, also making it more similar to the BF group. The dominant genus in the microbiome of all infants was Bifidobacterium throughout the intervention, which was maintained at 12 months. Although the serum metabolome of the F19 group was more similar to the group receiving the SF than the BF group, increases in serum TCA cycle intermediates and decreases in several amino acids in the metabolome of the F19 group were observed, which resulted in a metabolome that trended toward the BF group. Overall, L. paracasei F19 supplementation did not override the impact of formula-feeding but did impact the microbiome and the serum metabolome in a way that may mitigate some unfavorable metabolic impacts of formula-feeding.
Collapse
Affiliation(s)
- Hanna Lee
- Department of Food Science and Technology, University of California, Davis, Davis, CA, United States
| | - Zailing Li
- Department of Pediatrics, Peking University Third Hospital, Beijing, China
| | | | - Yongmei Peng
- Department of Child Health Care, Children's Hospital, Fudan University, Shanghai, China
| | - Xiaonan Li
- Department of Child Health Care, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Olle Hernell
- Department of Clinical Sciences, Pediatrics, Umeå University, Umeå, Sweden
| | - Bo Lönnerdal
- Department of Nutrition, University of California, Davis, Davis, CA, United States
| | - Carolyn M Slupsky
- Department of Food Science and Technology, University of California, Davis, Davis, CA, United States.,Department of Nutrition, University of California, Davis, Davis, CA, United States
| |
Collapse
|
38
|
KDP, a Lactobacilli Product from Kimchi, Enhances Mucosal Immunity by Increasing Secretory IgA in Mice and Exhibits Antimicrobial Activity. Nutrients 2021; 13:nu13113936. [PMID: 34836191 PMCID: PMC8618749 DOI: 10.3390/nu13113936] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 10/31/2021] [Accepted: 11/01/2021] [Indexed: 12/24/2022] Open
Abstract
The potential of KDP, a lactic acid bacterial strain of Lactobacillus sakei, to enhance the production of mucosal specific immunoglobulin A (IgA) in mice and thereby enhance gut mucosal immunity was examined. KDP is composed of dead cells isolated from the Korean traditional food kimchi. Female BALB/c mice orally received 0.25 mg KDP once daily for 5 weeks and were co-administrated ovalbumin (OVA) for negative control and cholera toxin for positive control. Mice administered KDP exhibited increased secretory IgA (sIgA) contents in the small intestine, Peyer’s patches, serum, colon, and lungs as examined by ELISA. KDP also significantly increased the gene expression of Bcl-6, IL-10, IL-12p40, IL-21, and STAT4. In addition, KDP acted as a potent antioxidant, as indicated by its significant inhibitory effects in the range of 16.5–59.4% for DPPH, nitric oxide, maximum total antioxidant capacity, and maximum reducing power. Finally, KDP exhibited potent antimicrobial activity as evidenced by a significant decrease in the growth of 7 samples of gram-negative and gram-positive bacteria and Candida albicans. KDP’s adjuvant effect is shown to be comparable to that of cholera toxin. We conclude that KDP can significantly enhance the intestine’s secretory immunity to OVA, as well as act as a potent antioxidant and antimicrobial agent. These results suggest that orally administered KDP should be studied in clinical trials for antigen-specific IgA production.
Collapse
|
39
|
Nezametdinova VZ, Yunes RA, Dukhinova MS, Alekseeva MG, Danilenko VN. The Role of the PFNA Operon of Bifidobacteria in the Recognition of Host's Immune Signals: Prospects for the Use of the FN3 Protein in the Treatment of COVID-19. Int J Mol Sci 2021; 22:ijms22179219. [PMID: 34502130 PMCID: PMC8430577 DOI: 10.3390/ijms22179219] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 08/21/2021] [Accepted: 08/24/2021] [Indexed: 12/11/2022] Open
Abstract
Bifidobacteria are some of the major agents that shaped the immune system of many members of the animal kingdom during their evolution. Over recent years, the question of concrete mechanisms underlying the immunomodulatory properties of bifidobacteria has been addressed in both animal and human studies. A possible candidate for this role has been discovered recently. The PFNA cluster, consisting of five core genes, pkb2, fn3, aaa-atp, duf58, tgm, has been found in all gut-dwelling autochthonous bifidobacterial species of humans. The sensory region of the species-specific serine-threonine protein kinase (PKB2), the transmembrane region of the microbial transglutaminase (TGM), and the type-III fibronectin domain-containing protein (FN3) encoded by the I gene imply that the PFNA cluster might be implicated in the interaction between bacteria and the host immune system. Moreover, the FN3 protein encoded by one of the genes making up the PFNA cluster, contains domains and motifs of cytokine receptors capable of selectively binding TNF-α. The PFNA cluster could play an important role for sensing signals of the immune system. Among the practical implications of this finding is the creation of anti-inflammatory drugs aimed at alleviating cytokine storms, one of the dire consequences resulting from SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Venera Z. Nezametdinova
- Laboratory of Bacterial Genetics, The Vavilov Institute of General Genetics, 117971 Moscow, Russia; (V.Z.N.); (R.A.Y.); (M.G.A.)
| | - Roman A. Yunes
- Laboratory of Bacterial Genetics, The Vavilov Institute of General Genetics, 117971 Moscow, Russia; (V.Z.N.); (R.A.Y.); (M.G.A.)
| | - Marina S. Dukhinova
- International Institute ‘Solution Chemistry of Advanced Materials and Technologies’, ITMO University, 197101 Saint-Petersburg, Russia;
| | - Maria G. Alekseeva
- Laboratory of Bacterial Genetics, The Vavilov Institute of General Genetics, 117971 Moscow, Russia; (V.Z.N.); (R.A.Y.); (M.G.A.)
| | - Valery N. Danilenko
- Laboratory of Bacterial Genetics, The Vavilov Institute of General Genetics, 117971 Moscow, Russia; (V.Z.N.); (R.A.Y.); (M.G.A.)
- Correspondence:
| |
Collapse
|
40
|
Choi IY, Kim J, Kim SH, Ban OH, Yang J, Park MK. Safety Evaluation of Bifidobacterium breve IDCC4401 Isolated from Infant Feces for Use as a Commercial Probiotic. J Microbiol Biotechnol 2021; 31:949-955. [PMID: 34024895 PMCID: PMC9706084 DOI: 10.4014/jmb.2103.03041] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 05/10/2021] [Accepted: 05/21/2021] [Indexed: 12/15/2022]
Abstract
Previously, our research group isolated Bifidobacterium breve IDCC4401 from infant feces as a potential probiotic. For this study, we evaluated the safety of B. breve IDCC4401 using genomic and phenotypic analyses. Whole genome sequencing was performed to identify genomic characteristics and investigate the potential presence of genes encoding virulence, antibiotic resistance, and mobile genetic elements. Phenotypic analyses including antibiotic susceptibility, enzyme activity, production of biogenic amines (BAs), and proportion of D-/L-lactate were evaluated using E-test, API ZYM test, high-performance liquid chromatography (HPLC), and D-/L-lactic acid assay respectively. The genome of B. breve IDCC4401 consists of 2,426,499 bp with a GC content of 58.70% and 2,016 coding regions. Confirmation of the genome as B. breve was provided by its 98.93% similarity with B. breve DSM20213. Furthermore, B. breve IDCC4401 genes encoding virulence and antibiotic resistance were not identified. Although B. breve IDCC4401 showed antibiotic resistance against vancomycin, we confirmed that this was an intrinsic feature since the antibiotic resistance gene was not present. B. breve IDCC4401 showed leucine arylamidase, cystine arylamidase, α-galactosidase, β-galactosidase, and α-glucosidase activities, whereas it did not show production of harmful enzymes such as β-glucosidase and β-glucuronidase. In addition, B. breve IDCC4401 did not produce any tyramine, histamine, putrescine, cadaverine, or 2-phenethylamine, which are frequently detected BAs during fermentation. B. breve IDCC4401 produced 95.08% of L-lactate and 4.92% of Dlactate. Therefore, our findings demonstrate the safety of B. breve IDCC 4401 as a potential probiotic for use in the food industry.
Collapse
Affiliation(s)
- In Young Choi
- School of Food Science and Biotechnology, Kyungpook National University, Daegu 41566, Republic of Korea,Food and Bio-Industry Research Institute, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Jinhee Kim
- Drug Information Platform Center, Korea Research Institute of Chemical Technology, Daejeon 34114, Republic of Korea
| | - Su-Hyeon Kim
- School of Food Science and Biotechnology, Kyungpook National University, Daegu 41566, Republic of Korea,Food and Bio-Industry Research Institute, Kyungpook National University, Daegu 41566, Republic of Korea
| | - O-Hyun Ban
- Ildong BioScience, Pyeongtaek 17957, Republic of Korea
| | - Jungwoo Yang
- Ildong BioScience, Pyeongtaek 17957, Republic of Korea
| | - Mi-Kyung Park
- School of Food Science and Biotechnology, Kyungpook National University, Daegu 41566, Republic of Korea,Food and Bio-Industry Research Institute, Kyungpook National University, Daegu 41566, Republic of Korea,Corresponding author Phone: +82-53-950-5776 Fax: +82-53-950-6772 E-mail:
| |
Collapse
|
41
|
Ohnuma T, Taku T, Nagatani T, Horii A, Imaoka S, Tanaka T. Chemo-enzymatic synthesis of Lacto-N-biose I catalyzed by β-1,3-galactosidase from Bacillus circulans using 4,6-dimethoxy-1,3,5-triazin-2-yl β-galactopyranoside as a glycosyl donor. Biosci Biotechnol Biochem 2021; 85:1716-1719. [PMID: 33877301 DOI: 10.1093/bbb/zbab071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Accepted: 04/16/2021] [Indexed: 11/12/2022]
Abstract
Chemo-enzymatic synthesis of lacto-N-biose I (LNB) catalyzed by β-1,3-galactosidase from Bacillus circulans (BgaC) has been developed using 4,6-dimethoxy-1,3,5-triazin-2-yl β-galactopyranoside (DMT-β-Gal) and GlcNAc as the donor and acceptor substrates, respectively. BgaC transferred the Gal moiety to the acceptor, giving rise to LNB. The maximum yield of LNB was obtained at the acceptor : donor substrate ratio of 1:30.
Collapse
Affiliation(s)
- Takayuki Ohnuma
- Department of Advanced Bioscience, Kindai University, Nara, Japan.,Agricultural Technology and Innovation Research Institute (ATIRI), Kindai University, Nara, Japan
| | - Tomoki Taku
- Department of Advanced Bioscience, Kindai University, Nara, Japan
| | - Takeshi Nagatani
- Department of Advanced Bioscience, Kindai University, Nara, Japan
| | - Atsushi Horii
- Department of Advanced Bioscience, Kindai University, Nara, Japan
| | - Shun Imaoka
- Department of Advanced Bioscience, Kindai University, Nara, Japan
| | - Tomonari Tanaka
- Department of Biobased Materials Science, Graduate School of Science and Technology, Kyoto Institute of Technology, Matsugasaki, Sakyo-ku, Kyoto, Japan
| |
Collapse
|