1
|
Zeng Z, Gong S, Quan C, Zhou S, Kulyar MFEA, Iqbal M, Li Y, Li X, Li J. Impact of Bacillus licheniformis from yaks following antibiotic therapy in mouse model. Appl Microbiol Biotechnol 2024; 108:139. [PMID: 38229401 DOI: 10.1007/s00253-023-12866-w] [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: 06/27/2023] [Revised: 10/19/2023] [Accepted: 10/30/2023] [Indexed: 01/18/2024]
Abstract
Gut microorganism (GM) is an integral component of the host microbiome and health system. Abuse of antibiotics disrupts the equilibrium of the microbiome, affecting environmental pathogens and host-associated bacteria alike. However, relatively little research on Bacillus licheniformis alleviates the adverse effects of antibiotics. To test the effect of B. licheniformis as a probiotic supplement against the effects of antibiotics, cefalexin was applied, and the recovery from cefalexin-induced jejunal community disorder and intestinal barrier damage was investigated by pathology, real-time PCR (RT-PCR), and high-throughput sequencing (HTS). The result showed that A group (antibiotic treatment) significantly reduced body weight and decreased the length of jejunal intestinal villi and the villi to crypt (V/C) value, which also caused structural damage to the jejunal mucosa. Meanwhile, antibiotic treatment suppressed the mRNA expression of tight junction proteins ZO-1, claudin, occludin, and Ki67 and elevated MUC2 expression more than the other Groups (P < 0.05 and P < 0.01). However, T group (B. licheniformis supplements after antibiotic treatment) restored the expression of the above genes, and there was no statistically significant difference compared to the control group (P > 0.05). Moreover, the antibiotic treatment increased the relative abundance of 4 bacterial phyla affiliated with 16 bacterial genera in the jejunum community, including the dominant Firmicutes, Proteobacteria, and Cyanobacteria in the jejunum. B. licheniformis supplements after antibiotic treatment reduced the relative abundance of Bacteroidetes and Proteobacteria and increased the relative abundance of Firmicutes, Epsilonbacteraeota, Lactobacillus, and Candidatus Stoquefichus. This study uses mimic real-world exposure scenarios by considering the concentration and duration of exposure relevant to environmental antibiotic contamination levels. We described the post-antibiotic treatment with B. licheniformis could restore intestinal microbiome disorders and repair the intestinal barrier. KEY POINTS: • B. licheniformis post-antibiotics restore gut balance, repair barrier, and aid health • Antibiotics harm the gut barrier, alter structure, and raise disease risk • Long-term antibiotics affect the gut and increase disease susceptibility.
Collapse
Affiliation(s)
- Zhibo Zeng
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
| | - Saisai Gong
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
| | - Chuxian Quan
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
| | - Shimeng Zhou
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
| | | | - Mudassar Iqbal
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
- Faculty of Veterinary and Animal Sciences, The Islamia University of Bahawalpur, Bahawalpur, 63100, Pakistan
| | - Yan Li
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
| | - Xiang Li
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China.
| | - Jiakui Li
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China.
- College of Animals Husbandry and Veterinary Medicine, Tibet Agricultural and Animal Husbandry University, Linzhi, 860000, Tibet, China.
| |
Collapse
|
2
|
Zhang H, Wang J, Shen J, Chen S, Yuan H, Zhang X, Liu X, Yu Y, Li X, Gao Z, Wang Y, Wang J, Song M. Prophylactic supplementation with Bifidobacterium infantis or its metabolite inosine attenuates cardiac ischemia/reperfusion injury. IMETA 2024; 3:e220. [PMID: 39135700 PMCID: PMC11316933 DOI: 10.1002/imt2.220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 06/10/2024] [Accepted: 06/11/2024] [Indexed: 08/15/2024]
Abstract
Emerging evidence has demonstrated the profound impact of the gut microbiome on cardiovascular diseases through the production of diverse metabolites. Using an animal model of myocardial ischemia-reperfusion (I/R) injury, we found that the prophylactic administration of a well-known probiotic, Bifidobacterium infantis (B. infantis), exhibited cardioprotective effects in terms of preserving cardiac contractile function and preventing adverse cardiac remodeling following I/R and that these cardioprotective effects were recapitulated by its metabolite inosine. Transcriptomic analysis further revealed that inosine mitigated I/R-induced cardiac inflammation and cell death. Mechanistic investigations elucidated that inosine suppressed the production of pro-inflammatory cytokines and reduced the numbers of dendritic cells and natural killer cells, achieved through the activation of the adenosine A2A receptor (A2AR) that when inhibited abrogated the cardioprotective effects of inosine. Additionally, in vitro studies using C2C12 myoblasts revealed that inosine attenuated cell death by serving as an alternative carbon source for adenosine triphosphate (ATP) generation through the purine salvage pathway when subjected to oxygen-glucose deprivation/reoxygenation that simulated myocardial I/R injury. Likewise, inosine reversed the I/R-induced decrease in ATP levels in mouse hearts. Taken together, our findings indicate that B. infantis or its metabolite inosine exerts cardioprotective effects against I/R by suppressing cardiac inflammation and attenuating cardiac cell death, suggesting prophylactic therapeutic options for acute ischemic cardiac injury.
Collapse
Affiliation(s)
- Hao Zhang
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Membrane BiologyInstitute of Zoology, Chinese Academy of SciencesBeijingChina
- University of Chinese Academy of SciencesBeijingChina
| | - Jiawan Wang
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Membrane BiologyInstitute of Zoology, Chinese Academy of SciencesBeijingChina
- Department of AnesthesiologyBeijing Chao‐Yang HospitalBeijingChina
| | - Jianghua Shen
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Membrane BiologyInstitute of Zoology, Chinese Academy of SciencesBeijingChina
- University of Chinese Academy of SciencesBeijingChina
- Beijing Institute for Stem Cell and Regenerative MedicineBeijingChina
| | - Siqi Chen
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Membrane BiologyInstitute of Zoology, Chinese Academy of SciencesBeijingChina
- University of Chinese Academy of SciencesBeijingChina
- Beijing Institute for Stem Cell and Regenerative MedicineBeijingChina
| | - Hailong Yuan
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Membrane BiologyInstitute of Zoology, Chinese Academy of SciencesBeijingChina
- Beijing Institute for Stem Cell and Regenerative MedicineBeijingChina
- Joint National Laboratory for Antibody Drug EngineeringHenan UniversityKaifengChina
| | - Xuan Zhang
- University of Chinese Academy of SciencesBeijingChina
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of MicrobiologyChinese Academy of SciencesBeijingChina
| | - Xu Liu
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Membrane BiologyInstitute of Zoology, Chinese Academy of SciencesBeijingChina
- University of Chinese Academy of SciencesBeijingChina
- Beijing Institute for Stem Cell and Regenerative MedicineBeijingChina
| | - Ying Yu
- University of Chinese Academy of SciencesBeijingChina
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of MicrobiologyChinese Academy of SciencesBeijingChina
| | - Xinran Li
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Membrane BiologyInstitute of Zoology, Chinese Academy of SciencesBeijingChina
- University of Chinese Academy of SciencesBeijingChina
- Beijing Institute for Stem Cell and Regenerative MedicineBeijingChina
| | - Zeyu Gao
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Membrane BiologyInstitute of Zoology, Chinese Academy of SciencesBeijingChina
- Beijing Institute for Stem Cell and Regenerative MedicineBeijingChina
- Institute for Stem Cell and Regeneration, Chinese Academy of SciencesBeijingChina
| | - Yaohui Wang
- Joint National Laboratory for Antibody Drug EngineeringHenan UniversityKaifengChina
| | - Jun Wang
- University of Chinese Academy of SciencesBeijingChina
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of MicrobiologyChinese Academy of SciencesBeijingChina
| | - Moshi Song
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Membrane BiologyInstitute of Zoology, Chinese Academy of SciencesBeijingChina
- University of Chinese Academy of SciencesBeijingChina
- Beijing Institute for Stem Cell and Regenerative MedicineBeijingChina
- Institute for Stem Cell and Regeneration, Chinese Academy of SciencesBeijingChina
| |
Collapse
|
3
|
Hwang CH, Kim SH, Lee CH. Bacterial Growth Modulatory Effects of Two Branched-Chain Hydroxy Acids and Their Production Level by Gut Microbiota. J Microbiol Biotechnol 2024; 34:1314-1321. [PMID: 38938006 PMCID: PMC11239411 DOI: 10.4014/jmb.2404.04009] [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: 04/05/2024] [Accepted: 04/25/2024] [Indexed: 06/29/2024]
Abstract
Branched-chain hydroxy acids (BCHAs), produced by lactic acid bacteria, have recently been suggested as bioactive compounds contributing to the systemic metabolism and modulation of the gut microbiome. However, the relationship between BCHAs and gut microbiome remains unclear. In this study, we investigated the effects of BCHAs on the growth of seven different families in the gut microbiota. Based on in vitro screening, both 2-hydroxyisovaleric acid (HIVA) and 2-hydroxyisocaproic acid (HICA) stimulated the growth of Lactobacillaceae and Bifidobacteriaceae, with HIVA showing a significant growth promotion. Additionally, we observed not only the growth promotion of probiotic Lactobacillaceae strains but also growth inhibition of pathogenic B. fragilis in a dosedependent manner. The production of HIVA and HICA varied depending on the family of the gut microbiota and was relatively high in case of Lactobacillaceae and Lachnosporaceae. Furthermore, HIVA and HICA production by each strain positively correlated with their growth variation. These results demonstrated gut microbiota-derived BCHAs as active metabolites that have bacterial growth modulatory effects. We suggest that BCHAs can be utilized as active metabolites, potentially contributing to the treatment of diseases associated with gut dysbiosis.
Collapse
Affiliation(s)
- Chan Hyuk Hwang
- Department of Bioscience and Biotechnology, Konkuk University, Seoul 05029, Republic of Korea
| | - Su-Hyun Kim
- Department of Bioscience and Biotechnology, Konkuk University, Seoul 05029, Republic of Korea
- MetaMass Corp., Seoul 05029, Republic of Korea
| | - Choong Hwan Lee
- Department of Bioscience and Biotechnology, Konkuk University, Seoul 05029, Republic of Korea
- MetaMass Corp., Seoul 05029, Republic of Korea
| |
Collapse
|
4
|
Wu D, Fu K, Zhang W, Li Y, Ji Y, Dai Y, Yang G. Chitosan nanomedicines-engineered bifidobacteria complexes for effective colorectal tumor-targeted delivery of SN-38. Int J Pharm 2024; 659:124283. [PMID: 38810933 DOI: 10.1016/j.ijpharm.2024.124283] [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: 02/15/2024] [Revised: 05/22/2024] [Accepted: 05/26/2024] [Indexed: 05/31/2024]
Abstract
The clinical application of 7-ethyl hydroxy-camptothecin (SN-38) maintains challenges not only due to its poor solubility and stability but also the lack of effective carriers to actively deliver SN-38 to deep tumor sites. Although SN-38-based nanomedicines could improve the solubility and stability from different aspects, the tumor targeting efficiency remains very low. Leveraging the hypoxic taxis of bifidobacteria bifidum (B. bifi) to the deep tumor area, we report SN-38-based nanomedicines-engineered bifidobacterial complexes for effective tumor-targeted delivery. Firstly, SN-38 was covalently coupled with poly-L-glutamic acid (L-PGA) and obtained soluble polymeric prodrug L-PGA-SN38 to improve its solubility and stability. To prolong the drug release, L-PGA-SN38 was mildly complexed with chitosan to form nanomedicines, and nanomedicines engineered B. bifi were further elaborated via electrostatic interaction of the excess of cationic chitosan shell from nanomedicines and anionic teichoic acid from B. bifi. The engineered B. bifi complexes inherited the bioactivity of native B. bifi and exhibited distinctly enhanced accumulation at the tumor site. More importantly, significantly elevated anti-tumor efficacy was achieved after the treatment of CS-L-PGA-SN38 NPs/B. bifi complexes, with favorable tumor suppression up to 80%. Such a B. bifi-mediated delivery system offers a promising platform for effective drug delivery and enhanced drug accumulation in the hypoxia deep tumor with superior anti-tumor efficacy.
Collapse
Affiliation(s)
- Danjun Wu
- College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou 310014, China.
| | - Kaili Fu
- College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou 310014, China
| | - Wangyang Zhang
- College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou 310014, China
| | - Yazhen Li
- College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou 310014, China
| | - Yaning Ji
- College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou 310014, China
| | - Yiwei Dai
- College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou 310014, China
| | - Gensheng Yang
- College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou 310014, China.
| |
Collapse
|
5
|
Raval SD, Archana G. Evaluation of synbiotic combinations of commercial probiotic strains with different prebiotics in in vitro and ex vivo human gut microcosm model. Arch Microbiol 2024; 206:315. [PMID: 38904672 DOI: 10.1007/s00203-024-04030-3] [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: 05/01/2024] [Revised: 05/25/2024] [Accepted: 06/04/2024] [Indexed: 06/22/2024]
Abstract
Exploring probiotics for their crosstalk with the host microbiome through the fermentation of non-digestible dietary fibers (prebiotics) for their potential metabolic end-products, particularly short-chain fatty acids (SCFAs), is important for understanding the endogenous host-gut microbe interaction. This study was aimed at a systematic comparison of commercially available probiotics to understand their synergistic role with specific prebiotics in SCFAs production both in vitro and in the ex vivo gut microcosm model. Probiotic strains isolated from pharmacy products including Lactobacillus sporogenes (strain not labeled), Lactobacillus rhamnosus GG (ATCC53103), Streptococcus faecalis (T-110 JPC), Bacillus mesentericus (TO-AJPC), Bacillus clausii (SIN) and Saccharomyces boulardii (CNCM I-745) were assessed for their probiotic traits including survival, antibiotic susceptibility, and antibacterial activity against pathogenic strains. Our results showed that the microorganisms under study had strain-specific abilities to persist in human gastrointestinal conditions and varied anti-infective efficacy and antibiotic susceptibility. The probiotic strains displayed variation in the utilization of six different prebiotic substrates for their growth under aerobic and anaerobic conditions. Their prebiotic scores (PS) revealed which were the most suitable prebiotic carbohydrates for the growth of each strain and suggested xylooligosaccharide (XOS) was the poorest utilized among all. HPLC analysis revealed a versatile pattern of SCFAs produced as end-products of prebiotic fermentation by the strains which was influenced by growth conditions. Selected synbiotic (prebiotic and probiotic) combinations showing high PS and high total SCFAs production were tested in an ex vivo human gut microcosm model. Interestingly, significantly higher butyrate and propionate production was found only when synbiotics were applied as against when individual probiotic or prebiotics were applied alone. qRT-PCR analysis with specific primers showed that there was a significant increase in the abundance of lactobacilli and bifidobacteria with synbiotic blends compared to pre-, or probiotics alone. In conclusion, this work presents findings to suggest prebiotic combinations with different well-established probiotic strains that may be useful for developing effective synbiotic blends.
Collapse
Affiliation(s)
- Shivani D Raval
- Department of Microbiology and Biotechnology Center, Faculty of Science, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat, 390 002, India
| | - G Archana
- Department of Microbiology and Biotechnology Center, Faculty of Science, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat, 390 002, India.
| |
Collapse
|
6
|
Rätsep M, Kilk K, Zilmer M, Kuusik S, Kuus L, Vallas M, Gerulis O, Štšepetova J, Orav A, Songisepp E. Investigation of Effects of Novel Bifidobacterium longum ssp. longum on Gastrointestinal Microbiota and Blood Serum Parameters in a Conventional Mouse Model. Microorganisms 2024; 12:840. [PMID: 38674784 PMCID: PMC11052112 DOI: 10.3390/microorganisms12040840] [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: 03/06/2024] [Revised: 04/16/2024] [Accepted: 04/17/2024] [Indexed: 04/28/2024] Open
Abstract
Representatives of the genus Bifidobacterium are widely used as probiotics to modulate the gut microbiome and alleviate various health conditions. The action mechanisms of probiotics rely on their direct effect on the gut microbiota and the local and systemic effect of its metabolites. The main purpose of this animal experiment was to assess the biosafety of the Bifidobacterium longum strain BIOCC1719. Additional aims were to characterise the influence of the strain on the intestinal microbiota and the effect on several health parameters of the host during 15- and 30-day oral administration of the strain to mice. The strain altered the gut microbial community, thereby altering luminal short-chain fatty acid metabolism, resulting in a shift in the proportions of acetic, butyric, and propionic acids in the faeces and serum of the test group mice. Targeted metabolic profiling of serum revealed the possible ability of the strain to positively affect the hosts' amino acids and bile acids metabolism, as the cholic acid, deoxycholic acid, aspartate, and glutamate concentration were significantly higher in the test group. The tendency to increase anti-inflammatory polyamines (spermidine, putrescine) and neuroprotective 3-indolepropionic acid metabolism and to lower uremic toxins (P-cresol-SO4, indoxyl-SO4) was registered. Thus, B. longum BIOCC1719 may exert health-promoting effects on the host through modulation of the gut microbiome and the host metabolome via inducing the production of health-promoting bioactive compounds. The health effects of the strain need to be confirmed in clinical trials with human volunteers.
Collapse
Affiliation(s)
- Merle Rätsep
- BioCC OÜ, Riia St. 181A, 50411 Tartu, Estonia (L.K.); (M.V.)
| | - Kalle Kilk
- Department of Biochemistry, Institute of Biomedicine and Translational Medicine, University of Tartu, Ravila St. 19, 50411 Tartu, Estonia; (K.K.)
| | - Mihkel Zilmer
- Department of Biochemistry, Institute of Biomedicine and Translational Medicine, University of Tartu, Ravila St. 19, 50411 Tartu, Estonia; (K.K.)
| | - Sirje Kuusik
- BioCC OÜ, Riia St. 181A, 50411 Tartu, Estonia (L.K.); (M.V.)
| | - Liina Kuus
- BioCC OÜ, Riia St. 181A, 50411 Tartu, Estonia (L.K.); (M.V.)
| | - Mirjam Vallas
- BioCC OÜ, Riia St. 181A, 50411 Tartu, Estonia (L.K.); (M.V.)
| | - Oksana Gerulis
- BioCC OÜ, Riia St. 181A, 50411 Tartu, Estonia (L.K.); (M.V.)
| | - Jelena Štšepetova
- Department of Microbiology, Institute of Biomedicine and Translational Medicine, University of Tartu, Ravila St. 19, 50411 Tartu, Estonia
| | - Aivar Orav
- Tartu Health Care College, Nooruse St. 5, 50411 Tartu, Estonia
| | - Epp Songisepp
- BioCC OÜ, Riia St. 181A, 50411 Tartu, Estonia (L.K.); (M.V.)
| |
Collapse
|
7
|
Renga G, Nunzi E, Stincardini C, Pariano M, Puccetti M, Pieraccini G, Di Serio C, Fraziano M, Poerio N, Oikonomou V, Mosci P, Garaci E, Fianchi L, Pagano L, Romani L. CPX-351 exploits the gut microbiota to promote mucosal barrier function, colonization resistance, and immune homeostasis. Blood 2024; 143:1628-1645. [PMID: 38227935 DOI: 10.1182/blood.2023021380] [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: 06/06/2023] [Revised: 12/18/2023] [Accepted: 01/09/2024] [Indexed: 01/18/2024] Open
Abstract
ABSTRACT CPX-351, a liposomal combination of cytarabine plus daunorubicin, has been approved for the treatment of adults with newly diagnosed, therapy-related acute myeloid leukemia (AML) or AML with myelodysplasia-related changes, because it improves survival and outcome of patients who received hematopoietic stem cell transplant compared with the continuous infusion of cytarabine plus daunorubicin (referred to as "7 + 3" combination). Because gut dysbiosis occurring in patients with AML during induction chemotherapy heavily affects the subsequent phases of therapy, we have assessed whether the superior activity of CPX-351 vs "7 + 3" combination in the real-life setting implicates an action on and by the intestinal microbiota. To this purpose, we have evaluated the impact of CPX-351 and "7 + 3" combination on mucosal barrier function, gut microbial composition and function, and antifungal colonization resistance in preclinical models of intestinal damage in vitro and in vivo and fecal microbiota transplantation. We found that CPX-351, at variance with "7 + 3" combination, protected from gut dysbiosis, mucosal damage, and gut morbidity while increasing antifungal resistance. Mechanistically, the protective effect of CPX-351 occurred through pathways involving both the host and the intestinal microbiota, namely via the activation of the aryl hydrocarbon receptor-interleukin-22 (IL-22)-IL-10 host pathway and the production of immunomodulatory metabolites by anaerobes. This study reveals how the gut microbiota may contribute to the good safety profile, with a low infection-related mortality, of CPX-351 and highlights how a better understanding of the host-microbiota dialogue may contribute to pave the way for precision medicine in AML.
Collapse
Affiliation(s)
- Giorgia Renga
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Emilia Nunzi
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | | | - Marilena Pariano
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Matteo Puccetti
- Department of Pharmaceutical Science, University of Perugia, Perugia, Italy
| | | | - Claudia Di Serio
- Department of Health Sciences, University of Florence, Florence, Italy
| | - Maurizio Fraziano
- Department of Biology, University of Rome "Tor Vergata," Rome, Italy
| | - Noemi Poerio
- Department of Biology, University of Rome "Tor Vergata," Rome, Italy
| | | | - Paolo Mosci
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | | | - Luana Fianchi
- Division of Hematology, Policlinico Gemelli, Università Cattolica Sacro Cuore, Rome, Italy
| | - Livio Pagano
- Division of Hematology, Policlinico Gemelli, Università Cattolica Sacro Cuore, Rome, Italy
| | - Luigina Romani
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
- San Raffaele Sulmona, Sulmona, Italy
| |
Collapse
|
8
|
Garcias-Bonet N, Roik A, Tierney B, García FC, Villela HDM, Dungan AM, Quigley KM, Sweet M, Berg G, Gram L, Bourne DG, Ushijima B, Sogin M, Hoj L, Duarte G, Hirt H, Smalla K, Rosado AS, Carvalho S, Thurber RV, Ziegler M, Mason CE, van Oppen MJH, Voolstra CR, Peixoto RS. Horizon scanning the application of probiotics for wildlife. Trends Microbiol 2024; 32:252-269. [PMID: 37758552 DOI: 10.1016/j.tim.2023.08.012] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 08/24/2023] [Accepted: 08/28/2023] [Indexed: 09/29/2023]
Abstract
The provision of probiotics benefits the health of a wide range of organisms, from humans to animals and plants. Probiotics can enhance stress resilience of endangered organisms, many of which are critically threatened by anthropogenic impacts. The use of so-called 'probiotics for wildlife' is a nascent application, and the field needs to reflect on standards for its development, testing, validation, risk assessment, and deployment. Here, we identify the main challenges of this emerging intervention and provide a roadmap to validate the effectiveness of wildlife probiotics. We cover the essential use of inert negative controls in trials and the investigation of the probiotic mechanisms of action. We also suggest alternative microbial therapies that could be tested in parallel with the probiotic application. Our recommendations align approaches used for humans, aquaculture, and plants to the emerging concept and use of probiotics for wildlife.
Collapse
Affiliation(s)
- Neus Garcias-Bonet
- Red Sea Research Center (RSRC), Division of Biological and Environmental Science and Engineering (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia
| | - Anna Roik
- Helmholtz Institute for Functional Marine Biodiversity (HIFMB), Oldenburg, Germany; Alfred Wegener Institute, Helmholtz Centre for Polar and Marine Research (AWI), Bremerhaven, Germany
| | - Braden Tierney
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
| | - Francisca C García
- Red Sea Research Center (RSRC), Division of Biological and Environmental Science and Engineering (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia
| | - Helena D M Villela
- Red Sea Research Center (RSRC), Division of Biological and Environmental Science and Engineering (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia
| | - Ashley M Dungan
- School of Biosciences, The University of Melbourne, Parkville, VIC, Australia
| | - Kate M Quigley
- Minderoo Foundation, Perth, WA, Australia; James Cook University, Townsville, Australia
| | - Michael Sweet
- Aquatic Research Facility, Nature-based Solutions Research Centre, University of Derby, Derby, UK
| | - Gabriele Berg
- Institute of Environmental Biotechnology, Graz University of Technology, Graz, Austria; University of Potsdam and Leibniz Institute for Agricultural Engineering and Bioeconomy (ATB), Potsdam, Germany
| | - Lone Gram
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Kgs., Lyngby, Denmark
| | - David G Bourne
- College of Science and Engineering, James Cook University, Townsville, QLD 4811, Australia; Australian Institute of Marine Science, PMB 3, Townsville MC, Townsville, QLD 4810, Australia
| | - Blake Ushijima
- Department of Biology and Marine Biology, University of North Carolina Wilmington, Wilmington, NC, USA
| | - Maggie Sogin
- Molecular Cell Biology, University of California, Merced, CA, USA
| | - Lone Hoj
- Australian Institute of Marine Science, PMB 3, Townsville MC, Townsville, QLD 4810, Australia
| | - Gustavo Duarte
- Red Sea Research Center (RSRC), Division of Biological and Environmental Science and Engineering (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia; IMPG, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Heribert Hirt
- Center for Desert Agriculture (CDA), Division of Biological and Environmental Science and Engineering (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia
| | | | - Alexandre S Rosado
- Red Sea Research Center (RSRC), Division of Biological and Environmental Science and Engineering (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia; Computational Bioscience Research Center (CBRC), Division of Biological and Environmental Science and Engineering (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia
| | - Susana Carvalho
- Red Sea Research Center (RSRC), Division of Biological and Environmental Science and Engineering (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia
| | | | - Maren Ziegler
- Department of Animal Ecology and Systematics, Justus Liebig University Giessen, Giessen, Germany
| | - Christopher E Mason
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA; WorldQuant Initiative on Quantitative Prediction, Weill Cornell Medicine, New York, NY, USA
| | - Madeleine J H van Oppen
- School of Biosciences, The University of Melbourne, Parkville, VIC, Australia; Australian Institute of Marine Science, PMB 3, Townsville MC, Townsville, QLD 4810, Australia
| | | | - Raquel S Peixoto
- Red Sea Research Center (RSRC), Division of Biological and Environmental Science and Engineering (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia; Computational Bioscience Research Center (CBRC), Division of Biological and Environmental Science and Engineering (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia.
| |
Collapse
|
9
|
Yinhe S, Lixiang L, Yan L, Xiang G, Yanqing L, Xiuli Z. Bacteroides thetaiotaomicron and its inactivated bacteria ameliorate colitis by inhibiting macrophage activation. Clin Res Hepatol Gastroenterol 2024; 48:102276. [PMID: 38158154 DOI: 10.1016/j.clinre.2023.102276] [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: 11/21/2023] [Revised: 12/16/2023] [Accepted: 12/27/2023] [Indexed: 01/03/2024]
Abstract
BACKGROUND Studies have demonstrated that Bacteroides thetaiotaomicron (BT) has protective effect against colon inflammation in murine models. Macrophages play an important role in gut immunity. However, the specific mechanisms of BT on macrophage are still unelucidated. Thus, our study investigates the anti-inflammatory effect of BT and its heat-treated inactivated bacteria on experimental colitis and macrophages. METHODS A dextran sulfate sodium (DSS)-induced acute colitis model with male C57BL/6 mice, BT (ATCC29148) strain, THP1 cell lines were used in this study. Live and heat-treated inactivated BT (IBT) solution (1 × 10^9cfu/ml) were intragastrically gavaged daily for 14 days. Colonic inflammation was determined by the disease activity index (DAI) score, colon length, histological score, and inflammatory factors. THP1 cells were induced towards M1, then treated with different concentrations of inactivated BT solution and p38 inhibitor. Western blotting, immunohistochemistry, immunofluorescence and qRT-PCR were performed to assess the levels of inflammatory cytokines and molecules of MAPK pathway including IL-6, TNF-α, IL-1β, IL-22, p38 and phosphor-p38 expressions. Moreover, 16S rRNA sequencing of colitis murine fecal samples was applied to investigate the influence of supplementation of BT to the gut microbiota homeostasis. RESULTS Both live and heat-treated inactivated BT decreased the DAI and histological scores as well as levels of inflammatory factors, particularly IL-6 while increasing IL-22 of DSS-induced colitis murine models. The cell experiments showed that inactivated BT downregulates IL-6 expression in THP1 via inhibiting p38 phosphorylation and affecting M1 polarization. Moreover, the 16S rRNA sequencing results showed that BT and IBT gavage could increase beta-diversity of gut flora in DSS-induced colitis mice. Furthermore, the significance test for differences between the groups showed that BT could increase Faecalebaculum, Lactobacillus and Bacteroides, while decreasing Akkermansia. CONCLUSION In summary, our findings imply that BT and its heat-treated inactivated bacteria exert a protective effect by suppressing macrophage-induced IL-6 through the inhibition of p38 MAPK pathway and ameliorating intestinal gut dysbiosis in experimental colitis.
Collapse
Affiliation(s)
- Sikong Yinhe
- Department of Gastroenterology, Qilu Hospital of Shandong University, Jinan, Shandong, PR China; Department of Gastroenterology, Qilu Hospital of Shandong University Qingdao, Qingdao, Shandong, PR China
| | - Li Lixiang
- Department of Gastroenterology, Qilu Hospital of Shandong University, Jinan, Shandong, PR China; Shandong Provincial Clinical Research Center for digestive disease, Shandong, PR China
| | - Li Yan
- Department of Gastroenterology, Qilu Hospital of Shandong University Qingdao, Qingdao, Shandong, PR China
| | - Gu Xiang
- Department of Gastroenterology, Qilu Hospital of Shandong University, Jinan, Shandong, PR China
| | - Li Yanqing
- Department of Gastroenterology, Qilu Hospital of Shandong University, Jinan, Shandong, PR China; Department of Gastroenterology, Qilu Hospital of Shandong University Qingdao, Qingdao, Shandong, PR China; Shandong Provincial Clinical Research Center for digestive disease, Shandong, PR China; Laboratory of Translational Gastroenterology, Qilu Hospital of Shandong University, Jinan, Shandong, PR China; Robot engineering laboratory for precise diagnosis and therapy of GI tumor, Qilu Hospital of Shandong University, Jinan, Shandong, PR China
| | - Zuo Xiuli
- Department of Gastroenterology, Qilu Hospital of Shandong University, Jinan, Shandong, PR China; Department of Gastroenterology, Qilu Hospital of Shandong University Qingdao, Qingdao, Shandong, PR China; Shandong Provincial Clinical Research Center for digestive disease, Shandong, PR China; Laboratory of Translational Gastroenterology, Qilu Hospital of Shandong University, Jinan, Shandong, PR China; Robot engineering laboratory for precise diagnosis and therapy of GI tumor, Qilu Hospital of Shandong University, Jinan, Shandong, PR China.
| |
Collapse
|
10
|
Zhu Q, Qi S, Guo D, Li C, Su M, Wang J, Li Z, Yang D, Sun H, Wang X, Wang M, Wu H, Yu S, Bai W, Zhang Y, Yang X, Jiang L, Liu J, Zhao Y, Xing X, Shi D, Feng L, Sun D. A survey of fecal virome and bacterial community of the diarrhea-affected cattle in northeast China reveals novel disease-associated ecological risk factors. mSystems 2024; 9:e0084223. [PMID: 38108282 PMCID: PMC10804951 DOI: 10.1128/msystems.00842-23] [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: 08/10/2023] [Accepted: 11/06/2023] [Indexed: 12/19/2023] Open
Abstract
Limited information on the virome and bacterial community hampers our ability to discern systemic ecological risk factors that cause cattle diarrhea, which has become a pressing issue in the control of disease. A total of 110 viruses, 1,011 bacterial genera, and 322 complete viral genomes were identified from 70 sequencing samples mixed with 1,120 fecal samples from 58 farms in northeast China. For the diarrheic samples, the identified virome and bacterial community varied in terms of composition, abundance, diversity, and geographic distribution in relation to different disease-associated ecological factors; the abundance of identified viruses and bacteria was significantly correlated with the host factors of clinical status, cattle type, and age, and with environmental factors such as aquaculture model and geographical location (P < 0.05); a significant interaction occurred between viruses and viruses, bacteria and bacteria, as well as between bacteria and viruses (P < 0.05). The abundance of SMB53, Butyrivibrio, Facklamia, Trichococcus, and Turicibacter was significantly correlated with the health status of cattle (P < 0.05). The proportion of BRV, BCoV, BKV, BToV, BoNoV, BoNeV, BoAstV, BEV, BoPV, and BVDV in 1,120 fecal samples varied from 1.61% to 12.05%. A series of significant correlations were observed between the prevalence of individual viruses and the disease-associated ecological factors. A genome-based phylogenetic analysis revealed high variability of 10 bovine enteric viruses. The bovine hungarovirus was initially identified in both dairy and beef cattle in China. This study elucidates the fecal virome and bacterial community signatures of cattle affected by diarrhea, and reveals novel disease-associated ecological risk factors, including cattle type, cattle age, aquaculture model, and geographical location.IMPORTANCEThe lack of data on the virome and bacterial community restricts our capability to recognize ecological risk factors for bovine diarrhea disease, thereby hindering our overall comprehension of the disease's cause. In this study, we found that, for the diarrheal samples, the identified virome and bacterial community varied in terms of composition, abundance, diversity, configuration, and geographic distribution in relation to different disease-associated ecological factors. A series of significant correlations were observed between the prevalence of individual viruses and the disease-associated ecological factors. Our study aims to uncover novel ecological risk factors of bovine diarrheal disease by examining the pathogenic microorganism-host-environment disease ecology, thereby providing a new perspective on the control of bovine diarrheal diseases.
Collapse
Affiliation(s)
- Qinghe Zhu
- Key Laboratory of Bovine Disease Control in Northeast China, Ministry of Agriculture and Rural affairs of the People’s Republic of China, Heilongjiang Provincial Key Laboratory of Prevention and Control of Bovine Diseases, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Shanshan Qi
- Key Laboratory of Bovine Disease Control in Northeast China, Ministry of Agriculture and Rural affairs of the People’s Republic of China, Heilongjiang Provincial Key Laboratory of Prevention and Control of Bovine Diseases, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Donghua Guo
- Key Laboratory of Bovine Disease Control in Northeast China, Ministry of Agriculture and Rural affairs of the People’s Republic of China, Heilongjiang Provincial Key Laboratory of Prevention and Control of Bovine Diseases, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Chunqiu Li
- Key Laboratory of Bovine Disease Control in Northeast China, Ministry of Agriculture and Rural affairs of the People’s Republic of China, Heilongjiang Provincial Key Laboratory of Prevention and Control of Bovine Diseases, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Mingjun Su
- Key Laboratory of Bovine Disease Control in Northeast China, Ministry of Agriculture and Rural affairs of the People’s Republic of China, Heilongjiang Provincial Key Laboratory of Prevention and Control of Bovine Diseases, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Jianfa Wang
- Key Laboratory of Bovine Disease Control in Northeast China, Ministry of Agriculture and Rural affairs of the People’s Republic of China, Heilongjiang Provincial Key Laboratory of Prevention and Control of Bovine Diseases, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Zijian Li
- Key Laboratory of Bovine Disease Control in Northeast China, Ministry of Agriculture and Rural affairs of the People’s Republic of China, Heilongjiang Provincial Key Laboratory of Prevention and Control of Bovine Diseases, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Dan Yang
- Key Laboratory of Bovine Disease Control in Northeast China, Ministry of Agriculture and Rural affairs of the People’s Republic of China, Heilongjiang Provincial Key Laboratory of Prevention and Control of Bovine Diseases, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Haibo Sun
- Key Laboratory of Bovine Disease Control in Northeast China, Ministry of Agriculture and Rural affairs of the People’s Republic of China, Heilongjiang Provincial Key Laboratory of Prevention and Control of Bovine Diseases, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Xiaoran Wang
- Key Laboratory of Bovine Disease Control in Northeast China, Ministry of Agriculture and Rural affairs of the People’s Republic of China, Heilongjiang Provincial Key Laboratory of Prevention and Control of Bovine Diseases, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Meijiao Wang
- Key Laboratory of Bovine Disease Control in Northeast China, Ministry of Agriculture and Rural affairs of the People’s Republic of China, Heilongjiang Provincial Key Laboratory of Prevention and Control of Bovine Diseases, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Haoyang Wu
- Key Laboratory of Bovine Disease Control in Northeast China, Ministry of Agriculture and Rural affairs of the People’s Republic of China, Heilongjiang Provincial Key Laboratory of Prevention and Control of Bovine Diseases, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Shiping Yu
- Key Laboratory of Bovine Disease Control in Northeast China, Ministry of Agriculture and Rural affairs of the People’s Republic of China, Heilongjiang Provincial Key Laboratory of Prevention and Control of Bovine Diseases, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Wenfei Bai
- Key Laboratory of Bovine Disease Control in Northeast China, Ministry of Agriculture and Rural affairs of the People’s Republic of China, Heilongjiang Provincial Key Laboratory of Prevention and Control of Bovine Diseases, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Yongchen Zhang
- Key Laboratory of Bovine Disease Control in Northeast China, Ministry of Agriculture and Rural affairs of the People’s Republic of China, Heilongjiang Provincial Key Laboratory of Prevention and Control of Bovine Diseases, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Xu Yang
- Key Laboratory of Bovine Disease Control in Northeast China, Ministry of Agriculture and Rural affairs of the People’s Republic of China, Heilongjiang Provincial Key Laboratory of Prevention and Control of Bovine Diseases, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Limin Jiang
- Key Laboratory of Bovine Disease Control in Northeast China, Ministry of Agriculture and Rural affairs of the People’s Republic of China, Heilongjiang Provincial Key Laboratory of Prevention and Control of Bovine Diseases, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Jiaying Liu
- Key Laboratory of Bovine Disease Control in Northeast China, Ministry of Agriculture and Rural affairs of the People’s Republic of China, Heilongjiang Provincial Key Laboratory of Prevention and Control of Bovine Diseases, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Yingying Zhao
- Key Laboratory of Bovine Disease Control in Northeast China, Ministry of Agriculture and Rural affairs of the People’s Republic of China, Heilongjiang Provincial Key Laboratory of Prevention and Control of Bovine Diseases, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Xiaoxu Xing
- Key Laboratory of Bovine Disease Control in Northeast China, Ministry of Agriculture and Rural affairs of the People’s Republic of China, Heilongjiang Provincial Key Laboratory of Prevention and Control of Bovine Diseases, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Da Shi
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Li Feng
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Dongbo Sun
- Key Laboratory of Bovine Disease Control in Northeast China, Ministry of Agriculture and Rural affairs of the People’s Republic of China, Heilongjiang Provincial Key Laboratory of Prevention and Control of Bovine Diseases, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
| |
Collapse
|
11
|
Rätsep M, Kilk K, Zilmer M, Kuus L, Songisepp E. A Novel Bifidobacterium longum ssp. longum Strain with Pleiotropic Effects. Microorganisms 2024; 12:174. [PMID: 38258000 PMCID: PMC10818833 DOI: 10.3390/microorganisms12010174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 01/11/2024] [Accepted: 01/11/2024] [Indexed: 01/24/2024] Open
Abstract
Postbiotics are gaining increasing interest among the scientific community as well as at the level of food processing enterprises. The aim of this preliminary study was to characterise the metabolic diversity of a novel Bifidobacterium longum strain, BIOCC 1719, of human origin. The change after 24 h cultivation in three media was assessed using a metabolomic approach. Milk-based substrates favoured the activity of the strain, promoting the production of B vitamins, essential amino acids, bile acids, and fatty acids. Vitamins B1, B2, B6, B7, and B12 (with an average increase of 20-30%) were produced in both whole milk and whey; the increased production in the latter was as high as 100% for B7 and 744% for B12. The essential amino acids methionine and threonine were produced (>38%) in both milk and whey, and there was an increased production of leucine (>50%) in milk and lysine (126%) in whey. Increases in the content of docosahexaenoic acid (DHA) by 20%, deoxycholic acid in milk and whey (141% and 122%, respectively), and cholic acid (52%) in milk were recorded. During the preliminary characterisation of the metabolic diversity of the novel B. longum strain, BIOCC 1719, we identified the bioactive compounds produced by the strain during fermentation. This suggests its potential use as a postbiotic ingredient to enrich the human diet.
Collapse
Affiliation(s)
- Merle Rätsep
- BioCC OÜ, Riia St. 181A, 50411 Tartu, Estonia; (M.R.)
| | - Kalle Kilk
- Department of Biochemistry, Institute of Biomedicine and Translational Medicine, University of Tartu, Ravila St. 19, 50411 Tartu, Estonia
| | - Mihkel Zilmer
- Department of Biochemistry, Institute of Biomedicine and Translational Medicine, University of Tartu, Ravila St. 19, 50411 Tartu, Estonia
| | - Liina Kuus
- BioCC OÜ, Riia St. 181A, 50411 Tartu, Estonia; (M.R.)
| | - Epp Songisepp
- BioCC OÜ, Riia St. 181A, 50411 Tartu, Estonia; (M.R.)
| |
Collapse
|
12
|
Abstract
The remarkable diversity of lymphocytes, essential components of the immune system, serves as an ingenious mechanism for maximizing the efficient utilization of limited host defense resources. While cell adhesion molecules, notably in gut-tropic T cells, play a central role in this mechanism, the counterbalancing molecular details have remained elusive. Conversely, we've uncovered the molecular pathways enabling extracellular vesicles secreted by lymphocytes to reach the gut's mucosal tissues, facilitating immunological regulation. This discovery sheds light on immune fine-tuning, offering insights into immune regulation mechanisms.
Collapse
Affiliation(s)
- Yasunari Matsuzaka
- Division of Molecular and Medical Genetics, Center for Gene and Cell Therapy, The Institute of Medical Science, The University of Tokyo, Minato-ku, Tokyo, Japan
- Administrative Section of Radiation Protection, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Japan
| | - Ryu Yashiro
- Administrative Section of Radiation Protection, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Japan
- Department of Mycobacteriology, Leprosy Research Center, National Institute of Infectious Diseases, Higashimurayama, Tokyo, Japan
| |
Collapse
|
13
|
Srivastava S, Basak U, Naghibi M, Vijayakumar V, Parihar R, Patel J, Jadon PS, Pandit A, Dargad RR, Khanna S, Kumar S, Day R. A randomized double-blind, placebo-controlled trial to evaluate the safety and efficacy of live Bifidobacterium longum CECT 7347 (ES1) and heat-treated Bifidobacterium longum CECT 7347 (HT-ES1) in participants with diarrhea-predominant irritable bowel syndrome. Gut Microbes 2024; 16:2338322. [PMID: 38630015 PMCID: PMC11028008 DOI: 10.1080/19490976.2024.2338322] [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: 12/01/2023] [Accepted: 03/29/2024] [Indexed: 04/19/2024] Open
Abstract
To determine the efficacy of the probiotic Bifidobacterium longum CECT 7347 (ES1) and postbiotic heat-treated Bifidobacterium longum CECT 7347 (HT-ES1) in improving symptom severity in adults with diarrhea-predominant irritable bowel syndrome (IBS-D), a randomised, double-blind, placebo-controlled trial with 200 participants split into three groups was carried out. Two capsules of either ES1, HT-ES1 or placebo were administered orally, once daily, for 84 days (12 weeks). The primary outcome was change in total IBS-Symptom Severity Scale (IBS-SSS) score from baseline, compared to placebo. Secondary outcome measures were stool consistency, quality of life, abdominal pain severity and anxiety scores. Safety parameters and adverse events were also monitored. The change in IBS-SSS scores from baseline compared to placebo, reached significance in the ES1 and HT-ES1 group, on Days 28, 56 and 84. The decrease in mean IBS-SSS score from baseline to Day 84 was: ES1 (-173.70 [±75.60]) vs placebo (-60.44 [±65.5]) (p < .0001) and HT-ES1 (-177.60 [±79.32]) vs placebo (-60.44 [±65.5]) (p < .0001). Secondary outcomes included changes in IBS-QoL, APS-NRS, stool consistency and STAI-S and STAI-T scores, with changes from baseline to Day 84 being significant in ES1 and HT-ES1 groups, compared to the placebo group. Both ES1 and HT-ES1 were effective in reducing IBS-D symptom severity, as evaluated by measures such as IBS-SSS, IBS-QoL, APS-NRS, stool consistency, and STAI, in comparison to the placebo. These results are both statistically significant and clinically meaningful, representing, to the best of the authors' knowledge, the first positive results observed for either a probiotic or postbiotic from the same strain, in this particular population.
Collapse
Affiliation(s)
- S Srivastava
- Clinical Development & Science Communications, Vedic Lifesciences Pvt Ltd, Mumbai, India
| | - U Basak
- Clinical Development & Science Communications, Vedic Lifesciences Pvt Ltd, Mumbai, India
| | - M Naghibi
- Medical Department, ADM Health & Wellness, London, UK
| | - V Vijayakumar
- Medical Department, ADM Health & Wellness, London, UK
| | - R Parihar
- Gastroenterology Department, Gastroplus Digestive Disease Centre, Ahmedabad, India
| | - J Patel
- Gastroenterology Department, Apex Gastro Clinic and Hospital, Ahmedabad, India
| | - PS Jadon
- Medicine Department, Jaipur National University Institute for Medical Science & Research Centre, Jaipur, India
| | - A Pandit
- General Surgery Department, United Multispeciality Hospital, Maharashtra, India
| | - RR Dargad
- Medicine Department, Lilavati Hospital & Research Centre, Maharashtra, India
| | - S Khanna
- Gastroenterology Department, Criticare Asia Multispeciality hospital, Maharashtra, India
| | - S Kumar
- Independent Biostatistical Consultant, Delhi, India
| | - R Day
- Medical Department, ADM Health & Wellness, London, UK
| |
Collapse
|
14
|
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
|
15
|
Sharma D, Gajjar D, Seshadri S. Understanding the role of gut microfloral bifidobacterium in cancer and its potential therapeutic applications. MICROBIOME RESEARCH REPORTS 2023; 3:3. [PMID: 38455077 PMCID: PMC10917622 DOI: 10.20517/mrr.2023.51] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 09/28/2023] [Accepted: 10/30/2023] [Indexed: 03/09/2024]
Abstract
Gut microbiota research has gained a tremendous amount of attention from the scientific community because of its contribution to gut homeostasis, human health, and various pathophysiological conditions. The early colonizer of the human gut, i.e., bifidobacteria, has emerged as an efficient probiotic in various diseased conditions, including cancer. This review explores the pros and cons of Bifidobacterium in various malignancies and various therapeutic strategies. We have illustrated the controversial role of bifidobacteria participating in various malignancies as well as described the current knowledge regarding its use in anticancer therapies. Ultimately, this article also addresses the need for further extensive research in elucidating the mechanism of how bifidobacteria is involved and is indirectly affecting the tumor microenvironment. Exhaustive and large-scale research is also required to solve the controversial questions regarding the involvement of bifidobacteria in cancer research.
Collapse
Affiliation(s)
| | | | - Sriram Seshadri
- Institute of Science, Nirma University, 382481 Ahmedabad, Gujarat, India
| |
Collapse
|
16
|
Yin P, Zhang C, Du T, Yi S, Yu L, Tian F, Chen W, Zhai Q. Meta-analysis reveals different functional characteristics of human gut Bifidobacteria associated with habitual diet. Food Res Int 2023; 170:112981. [PMID: 37316017 DOI: 10.1016/j.foodres.2023.112981] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Revised: 05/03/2023] [Accepted: 05/13/2023] [Indexed: 06/16/2023]
Abstract
Dietary habits contribute to the composition and function of the gut microbiota. Different dietary structures, including vegan, vegetarian, and omnivorous diets, affect intestinal Bifidobacteria; however, the relationship between Bifidobacterial function and host metabolism in subjects with different dietary patterns is unclear. Here, we analyzed five metagenomics studies and six 16S sequencing studies, including 206 vegetarians (VG), 249 omnivores (O), and 270 vegans (V), through an unbiased theme-level meta-analysis framework and discovered that diet significantly affects the composition and functionality of intestinal Bifidobacteria. The relative abundance of Bifidobacterium pseudocatenulatum was significantly higher in V than in O and Bifidobacterium longum, Bifidobacterium adolescentis, and B. pseudocatenulatum differed significantly in carbohydrate transport and metabolism in subjects with different diet types. Diets high in fiber were associated with B. longum with increased capacity for carbohydrate catabolism and genes encoding GH29 and GH43_27 were significantly enriched in V. Bifidobacterium adolescentis and B. pseudocatenulatum, associated with O, had a higher prevalence of the genes related to carbohydrate transport and metabolism, which showed the enrichment of GH26 and GH27 families. The same Bifidobacterium species has different functions in subjects with different diet types, resulting in different physiological significance. The diversification and functionalities of Bifidobacterial species in the gut microbiome can be influenced by the host diet and this aspect should be considered when studying host-microbe associations.
Collapse
Affiliation(s)
- Pingping Yin
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Chengcheng Zhang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Ting Du
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Shanrong Yi
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Leilei Yu
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Fengwei Tian
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Wei Chen
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China; National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Qixiao Zhai
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China.
| |
Collapse
|
17
|
Najafi S, Sotoodehnejadnematalahi F, Amiri MM, Pourshafie MR, Rohani M. Prophylactic vs. Therapeutic Effect of Probiotics on the Inflammation Mediated by the NF-κB Pathway in Inflammatory Bowel Conditions. Biomedicines 2023; 11:1675. [PMID: 37371769 DOI: 10.3390/biomedicines11061675] [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: 05/09/2023] [Revised: 05/23/2023] [Accepted: 06/07/2023] [Indexed: 06/29/2023] Open
Abstract
Probiotic supplements consumed adequately at the proper time can affect health by modulating inflammatory pathways in gastrointestinal epithelial cells and modifying the resultant inflammatory response. The current study applied in vitro models to investigate the effectiveness of probiotics in modulating inflammatory pathways and altering inflammatory gene expression in gastrointestinal epithelial cells, with the ultimate goal of promoting probiotic consumption as a therapeutic and preventive measure for chronic inflammatory bowel conditions. HT-29 cells were treated with Gram-negative bacteria to evaluate the changes in pathways related to inflammation activities before and after treatment with a Lactobacillus spp. cocktail (L. plantarum, L. rhamnosus, L. brevis, and L. ruteri) and a Bifidobacterium spp. cocktail (B. bifidum, B. langum, and B. breve) using the real-time PCR method and ELISA for IL-1β and IL-6 as pro-inflammatory cytokines. The results showed that the expression of NF-κB signaling pathway genes and IL-1β and IL-6 cytokines increased after exposure to Gram-negative components. In contrast, all probiotic combinations significantly decreased the expression of genes and the secretion of cytokines. However, this decrease was significantly smaller in cells that underwent probiotic treatment after inflammation induction. In addition, cocktails containing combined Lactobacillus and Bifidobacterium demonstrated robust anti-inflammatory activity relative to solo cocktails. Our observations confirm that probiotic consumption could positively impact inflammatory conditions and alleviate inflammatory symptoms; they can be particularly effective as a preventive measure. Our study provides preliminary evidence to support the lifetime consumption of probiotics.
Collapse
Affiliation(s)
- Saeideh Najafi
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran 14778-93855, Iran
| | | | - Mohammad Mehdi Amiri
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran 14155-6619, Iran
| | | | - Mahdi Rohani
- Department of Bacteriology, Pasteur Institute of Iran, Tehran 13169-43551, Iran
| |
Collapse
|
18
|
Du W, Wang X, Hu M, Hou J, Du Y, Si W, Yang L, Xu L, Xu Q. Modulating gastrointestinal microbiota to alleviate diarrhea in calves. Front Microbiol 2023; 14:1181545. [PMID: 37362944 PMCID: PMC10286795 DOI: 10.3389/fmicb.2023.1181545] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Accepted: 05/19/2023] [Indexed: 06/28/2023] Open
Abstract
The calf stage is a critical period for the development of heifers. Newborn calves have low gastrointestinal barrier function and immunity before weaning, making them highly susceptible to infection by various intestinal pathogens. Diarrhea in calves poses a significant threat to the health of young ruminants and may cause serious economic losses to livestock farms. Antibiotics are commonly used to treat diarrhea and promote calf growth, leading to bacterial resistance and increasing antibiotic residues in meat. Therefore, finding new technologies to improve the diarrhea of newborn calves is a challenge for livestock production and public health. The operation of the gut microbiota in the early stages after birth is crucial for optimizing immune function and body growth. Microbiota colonization of newborn animals is crucial for healthy development. Early intervention of the calf gastrointestinal microbiota, such as oral probiotics, fecal microbiota transplantation and rumen microbiota transplantation can effectively relieve calf diarrhea. This review focuses on the role and mechanisms of oral probiotics such as Lactobacillus, Bifidobacterium and Faecalibacterium in relieving calf diarrhea. The aim is to develop appropriate antibiotic alternatives to improve calf health in a sustainable and responsible manner, while addressing public health issues related to the use of antibiotics in livestock.
Collapse
|
19
|
Fatima S, Altwaijry H, Abulmeaty MMA, Abudawood M, Siddiqi NJ, Alrashoudi RH, Alsobaie S. Combined Supplementation of Clostridium butyricum and Bifidobacterium infantis Diminishes Chronic Unpredictable Mild Stress-Induced Intestinal Alterations via Activation of Nrf-2 Signaling Pathway in Rats. Int J Mol Sci 2023; 24:ijms24098264. [PMID: 37175970 PMCID: PMC10178881 DOI: 10.3390/ijms24098264] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Revised: 04/05/2023] [Accepted: 04/06/2023] [Indexed: 05/15/2023] Open
Abstract
Exposure to long-term chronic unpredictable mild stress (CUMS) can cause redox imbalance and inflammation, which may affect the integrity of the gut barrier. The present study was conducted to investigate the effects of a probiotics bacterium mixture, including Clostridium butyricum (C. butyricum) and Bifidobacterium infantis (B. infantis), on the intestinal homeostasis in rats exposed to multiple low-intensity stressors for 28 days. The mechanism of CUMS-induced altered intestinal homeostasis was evaluated by focusing on the nuclear factor-E2-related factor-2 (Nrf-2) pathway. In contrast to the CUMS group, probiotic mixture supplementation significantly (p < 0.01) reversed the stress-induced elevated corticosterone level, protein and lipid oxidation, and increased enzymatic and non-enzymatic antioxidant levels, as well as upregulated Nrf-2/HO-1 pathway. Probiotics supplementation further significantly (p < 0.01) decreased the CUMS-induced inflammation, altered T-lymphocyte levels, and suppressed the protein expression of nuclear factor kappa B (NF-κB) in rat intestines. Improvement in histological changes and intestinal barrier integrity further validate the beneficial effects of probiotic mixtures on CUMS-induced altered intestinal morphology. In conclusion, our results suggest that the combination of C. butyricum and B. infantis significantly attenuated CUMS-induced oxidative stress, inflammation, and T-lymphocyte modulation by upregulating Nrf-2/HO-1 signaling and inhibiting NF-κB expression in rat intestine.
Collapse
Affiliation(s)
- Sabiha Fatima
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Saud University, Riyadh 11433, Saudi Arabia
| | - Haifa Altwaijry
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Saud University, Riyadh 11433, Saudi Arabia
| | - Mahmoud M A Abulmeaty
- Department of Community Health Sciences, College of Applied Medical Sciences, King Saud University, Riyadh 11362, Saudi Arabia
| | - Manal Abudawood
- Chair of Medical and Molecular Genetics Research, Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Saud University, Riyadh 11433, Saudi Arabia
| | - Nikhat J Siddiqi
- Department of Biochemistry, College of Science, King Saud University, Riyadh 11495, Saudi Arabia
| | - Reem Hamoud Alrashoudi
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Saud University, Riyadh 11433, Saudi Arabia
| | - Sarah Alsobaie
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Saud University, Riyadh 11433, Saudi Arabia
| |
Collapse
|
20
|
Calvete-Torre I, Sabater C, Delgado S, Ruas-Madiedo P, Rupérez-García A, Montilla A, Javier Moreno F, Margolles A, Ruiz L. Arabinoxylan-based substrate preferences and predicted metabolic properties of Bifidobacterium longum subspecies as a basis to design differential media. Food Res Int 2023; 167:112711. [PMID: 37087214 DOI: 10.1016/j.foodres.2023.112711] [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: 06/30/2022] [Revised: 03/09/2023] [Accepted: 03/14/2023] [Indexed: 03/30/2023]
Abstract
Arabinoxylan (AX) and arabinoxylo-oligosaccharides (AXOS) derived therefrom are emergent prebiotics with promising health promoting properties, likely linked to its capacity to foster beneficial species in the human gut. Bifidobacteria appear to be one taxa that is frequently promoted following AX or AXOS consumption, and that is known to establish metabolic cross-feeding networks with other beneficial commensal species. Therefore, probiotic bifidobacteria with the capability to metabolize AX-derived prebiotics represent interesting candidates to develop novel probiotic and synbiotic combinations with AX-based prebiotics. In this work we have deepen into the metabolic capabilities of bifidobacteria related to AX and AXOS metabolization through a combination of in silico an in vitro tools. Both approaches revealed that Bifidobacterium longum and, particularly, B. longum subsp. longum, appears as the better equipped to metabolize complex AX substrates, although other related subspecies such as B. longum subsp. infantis, also hold some machinery related to AXOS metabolization. This correlates to the growth profiles exhibited by representative strains of both subspecies in AX or AXOS enriched media. Based on these results, we formulated a differential carbohydrate free medium (CFM) supplemented with a combination of AX and AXOS that enabled to recover a wide diversity of Bifidobacterium species from complex fecal samples, while allowing easy discrimination of AX metabolising strains by the appearance of a precipitation halo. This new media represent an appealing alternative to isolate novel probiotic bifidobacteria, rapidly discriminating their capacity to metabolize structurally complex AX-derived prebiotics. This can be convenient to assist formulation of novel functional foods and supplements, including bifidobacterial species with capacity to metabolize AX-derived prebiotic ingredients.
Collapse
Affiliation(s)
- Ines Calvete-Torre
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias-Consejo Superior de Investigaciones Científicas IPLA, CSIC, Paseo Río Linares s/n, 33300, Villaviciosa, Asturias, Spain; Functionality and Ecology of Beneficial Microbes (MicroHealth) Group, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Asturias, Spain
| | - Carlos Sabater
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias-Consejo Superior de Investigaciones Científicas IPLA, CSIC, Paseo Río Linares s/n, 33300, Villaviciosa, Asturias, Spain; Functionality and Ecology of Beneficial Microbes (MicroHealth) Group, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Asturias, Spain
| | - Susana Delgado
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias-Consejo Superior de Investigaciones Científicas IPLA, CSIC, Paseo Río Linares s/n, 33300, Villaviciosa, Asturias, Spain; Functionality and Ecology of Beneficial Microbes (MicroHealth) Group, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Asturias, Spain
| | - Patricia Ruas-Madiedo
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias-Consejo Superior de Investigaciones Científicas IPLA, CSIC, Paseo Río Linares s/n, 33300, Villaviciosa, Asturias, Spain; Functionality and Ecology of Beneficial Microbes (MicroHealth) Group, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Asturias, Spain
| | - Alicia Rupérez-García
- Instituto de Investigación en Ciencias de la Alimentación CIAL, (CSIC-UAM) CEI (CSIC+UAM), Madrid, Spain
| | - Antonia Montilla
- Instituto de Investigación en Ciencias de la Alimentación CIAL, (CSIC-UAM) CEI (CSIC+UAM), Madrid, Spain
| | - F Javier Moreno
- Instituto de Investigación en Ciencias de la Alimentación CIAL, (CSIC-UAM) CEI (CSIC+UAM), Madrid, Spain
| | - Abelardo Margolles
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias-Consejo Superior de Investigaciones Científicas IPLA, CSIC, Paseo Río Linares s/n, 33300, Villaviciosa, Asturias, Spain; Functionality and Ecology of Beneficial Microbes (MicroHealth) Group, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Asturias, Spain.
| | - Lorena Ruiz
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias-Consejo Superior de Investigaciones Científicas IPLA, CSIC, Paseo Río Linares s/n, 33300, Villaviciosa, Asturias, Spain; Functionality and Ecology of Beneficial Microbes (MicroHealth) Group, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Asturias, Spain.
| |
Collapse
|
21
|
Wang S, Shan Y, Zhang S, Zhang L, Jiao Y, Xue D, Zhang L, Yi H. Lactobacillus paracasei subsp. paracasei X12 Strain Induces Apoptosis in HT-29 Cells through Activation of the Mitochondrial Pathway. Nutrients 2023; 15:2123. [PMID: 37432295 DOI: 10.3390/nu15092123] [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: 03/20/2023] [Revised: 04/18/2023] [Accepted: 04/23/2023] [Indexed: 07/12/2023] Open
Abstract
L. paracasei subsp. paracasei X12 was obtained from traditional cheese produced in northwestern China. In this study, we showed that whole peptidoglycan (WPG), extracted from L. paracasei subsp. paracasei X12, inhibited proliferation and induced apoptosis in HT-29 cells in a dose-dependent manner. In addition, WPG-induced apoptosis was associated with the loss of mitochondrial membrane potential (Ψm), the release of cytochrome c (Cyto-C) from mitochondrialto cytosolic spaces, activation of Caspase 3, and accumulation of intracellular reactive oxygen species (ROS). Finally, semi-quantitative RT-PCR showed that these events were accompanied by upregulation of proapoptotic genes (Bax or Bad) and downregulation of antiapoptotic genes (Bcl-xl). Taken together, our results demonstrated that WPG induced apoptosis in HT-29 cells through activation of the mitochondrial pathway. WPG exerted only minor toxicity upon noncancerous cells and therefore might be used as a natural agent in the treatment of cancer in future.
Collapse
Affiliation(s)
- Shumei Wang
- College of Food Engineering, Heilongjiang Province Key Laboratory of Cold Region Wetland Ecology and Environment Research, Harbin University, Harbin 150086, China
| | - Yi Shan
- College of Food Engineering, Heilongjiang Province Key Laboratory of Cold Region Wetland Ecology and Environment Research, Harbin University, Harbin 150086, China
| | - Shuang Zhang
- College of Food Science, Northeast Agricultural University, Harbin 150030, China
| | - Lanwei Zhang
- College of Food Science and Engineering, Ocean University of China, Qingdao 266000, China
| | - Yuehua Jiao
- Center of Drug Safety Evaluation, Heilongjiang University of Chinese Medicine, Harbin 150040, China
| | - Dijia Xue
- College of Food Science and Engineering, Inner Mongolia Agricultural University, Hohhot 010018, China
| | - Lili Zhang
- College of Food Science, Northeast Agricultural University, Harbin 150030, China
| | - Huaxi Yi
- College of Food Science and Engineering, Ocean University of China, Qingdao 266000, China
| |
Collapse
|
22
|
Ladeira R, Tap J, Derrien M. Exploring Bifidobacterium species community and functional variations with human gut microbiome structure and health beyond infancy. MICROBIOME RESEARCH REPORTS 2023; 2:9. [PMID: 38047280 PMCID: PMC10688807 DOI: 10.20517/mrr.2023.01] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Revised: 03/15/2023] [Accepted: 03/20/2023] [Indexed: 12/05/2023]
Abstract
Aim: The human gut Bifidobacterium community has been studied in detail in infants and following dietary interventions in adults. However, the variability of the distribution of Bifidobacterium species and intra-species functions have been little studied, particularly beyond infancy. Here, we explore the ecology of Bifidobacterium communities in a large public dataset of human gut metagenomes, mostly corresponding to adults. Methods: We selected 9.515 unique gut metagenomes from curatedMetagenomicData. Samples were partitioned by applying Dirichlet's multinomial mixture to Bifidobacterium species. A functional analysis was performed on > 2.000 human-associated Bifidobacterium metagenome-assembled genomes (MAGs) paired with participant gut microbiome and health features. Results: We identified several Bifidobacterium-based partitions in the human gut microbiome differing in terms of the presence and abundance of Bifidobacterium species. The partitions enriched in both B. longum and B. adolescentis were associated with gut microbiome diversity and a higher abundance of butyrate producers and were more prevalent in healthy individuals. B. bifidum MAGs harboring a set of genes potentially related to phages were more prevalent in partitions associated with a lower gut microbiome diversity and were genetically more closely related. Conclusion: This study expands our knowledge of the ecology and variability of the Bifidobacterium community, particularly in adults, and its specific association with the gut microbiota and health. Its findings may guide the rational selection of Bifidobacterium strains for gut microbiome complementation according to the individual's endogenous Bifidobacterium community. Our results also suggest that gut microbiome stratification for particular genera may be relevant for studies of variations of species and associations with the gut microbiome and health.
Collapse
Affiliation(s)
- Ruben Ladeira
- Advanced Health & Science, Danone Global Research & Innovation Center, Gif-sur-Yvette 91190, France
| | - Julien Tap
- Advanced Health & Science, Danone Global Research & Innovation Center, Gif-sur-Yvette 91190, France
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas 78350, France
| | - Muriel Derrien
- Advanced Health & Science, Danone Global Research & Innovation Center, Gif-sur-Yvette 91190, France
| |
Collapse
|
23
|
Arjun OK, Prakash T. Identification of conserved genomic signatures specific to Bifidobacterium species colonising the human gut. 3 Biotech 2023; 13:97. [PMID: 36852175 PMCID: PMC9958220 DOI: 10.1007/s13205-023-03492-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 01/25/2023] [Indexed: 02/26/2023] Open
Abstract
Bifidobacterium species are known for their ability to inhabit various habitats and are often regarded as the first colonisers of the human gut. In the present work, we have used comparative genomics to identify conserved genomic signatures specific to Bifidobacterium species associated with the human gut. Our approach discovered five genomic signatures with varying lengths and confidence. Among the predicted five signatures, a 1790 bp multi-drug resistance (MDR) signature was found to be remarkably specific to only those species that can colonise the human gut. The signature codes for a membrane transport protein belonging to the major facilitator superfamily (MFS) generally involved in MDR. Phylogenetic analyses of the MDR signature suggest a lineage-specific evolution of the MDR signature in bifidobacteria colonising the human gut. Functional annotation led to the discovery of two conserved domains in the protein; a catalytic MFS domain involved in the efflux of drugs and toxins, and a regulatory cystathionine-β-synthase (CBS) domain that can interact with adenosyl-carriers. Molecular docking simulation performed with the modelled tertiary structure of the MDR signature revealed the putative functional role of the covalently linked domains. The MFS domain displayed a high affinity towards various protein synthesis inhibitor antibiotics and human bile acids, whereas the C-terminally linked CBS domain exhibited favourable binding with molecular structures of ATP and AMP. Therefore, we believe that the predicted signature represents a niche-specific survival trait involved in bile and antibiotic resistance, imparting an adaptive advantage to the Bifidobacterium species colonising the human gut. Supplementary Information The online version contains supplementary material available at 10.1007/s13205-023-03492-4.
Collapse
Affiliation(s)
- O. K. Arjun
- School of Biosciences and Bioengineering, Indian Institute of Technology Mandi, Mandi, Himachal Pradesh 175005 India
| | - Tulika Prakash
- School of Biosciences and Bioengineering, Indian Institute of Technology Mandi, Mandi, Himachal Pradesh 175005 India
| |
Collapse
|
24
|
Infant Fecal Fermentations with Galacto-Oligosaccharides and 2′-Fucosyllactose Show Differential Bifidobacterium longum Stimulation at Subspecies Level. CHILDREN 2023; 10:children10030430. [PMID: 36979988 PMCID: PMC10047592 DOI: 10.3390/children10030430] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 01/26/2023] [Accepted: 02/21/2023] [Indexed: 02/25/2023]
Abstract
The objective of the current study was to evaluate the potential of 2′-FL and GOS, individually and combined, in beneficially modulating the microbial composition of infant and toddler (12–18 months) feces using the micro-Matrix bioreactor. In addition, the impacts of GOS and 2′-FL, individually and combined, on the outgrowth of fecal bifidobacteria at (sub)species level was investigated using the baby M-SHIME® model. For young toddlers, significant increases in the genera Bifidobacterium, Veillonella, and Streptococcus, and decreases in Enterobacteriaceae, Clostridium XIVa, and Roseburia were observed in all supplemented fermentations. In addition, GOS, and combinations of GOS and 2′-FL, increased Collinsella and decreased Salmonella, whereas 2′-FL, and combined GOS and 2′-FL, decreased Dorea. Alpha diversity increased significantly in infants with GOS and/or 2′-FL, as well as the relative abundances of the genera Veillonella and Akkermansia with 2′-FL, and Lactobacillus with GOS. Combinations of GOS and 2′-FL significantly stimulated Veillonella, Lactobacillus, Bifidobacterium, and Streptococcus. In all supplemented fermentations, Proteobacteria decreased, with the most profound decreases accomplished by the combination of GOS and 2′-FL. When zooming in on the different (sub)species of Bifidobacterium, GOS and 2’-FL were shown to be complementary in stimulating breast-fed infant-associated subspecies of Bifidobacterium longum in a dose-dependent manner: GOS stimulated Bifidobacterium longum subsp. longum, whereas 2′-FL supported outgrowth of Bifidobacterium longum subsp. infantis.
Collapse
|
25
|
Luo S, Hou Y, Xie L, Zhang H, Liu C, Chen T. Effects of microwave on the potential microbiota modulating effects of agro-industrial by-product fibers among different individuals. Lebensm Wiss Technol 2023. [DOI: 10.1016/j.lwt.2023.114621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/27/2023]
|
26
|
Ma B, Gavzy SJ, Saxena V, Song Y, Piao W, Lwin HW, Lakhan R, Iyyathurai J, Li L, France M, Paluskievicz C, Shirkey MW, Hittle L, Munawwar A, Mongodin EF, Bromberg JS. Strain-specific alterations in gut microbiome and host immune responses elicited by tolerogenic Bifidobacterium pseudolongum. Sci Rep 2023; 13:1023. [PMID: 36658194 PMCID: PMC9852428 DOI: 10.1038/s41598-023-27706-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 01/06/2023] [Indexed: 01/20/2023] Open
Abstract
The beneficial effects attributed to Bifidobacterium are largely attributed to their immunomodulatory capabilities, which are likely to be species- and even strain-specific. However, their strain-specificity in direct and indirect immune modulation remain largely uncharacterized. We have shown that B. pseudolongum UMB-MBP-01, a murine isolate strain, is capable of suppressing inflammation and reducing fibrosis in vivo. To ascertain the mechanism driving this activity and to determine if it is specific to UMB-MBP-01, we compared it to a porcine tropic strain B. pseudolongum ATCC25526 using a combination of cell culture and in vivo experimentation and comparative genomics approaches. Despite many shared features, we demonstrate that these two strains possess distinct genetic repertoires in carbohydrate assimilation, differential activation signatures and cytokine responses signatures in innate immune cells, and differential effects on lymph node morphology with unique local and systemic leukocyte distribution. Importantly, the administration of each B. pseudolongum strain resulted in major divergence in the structure, composition, and function of gut microbiota. This was accompanied by markedly different changes in intestinal transcriptional activities, suggesting strain-specific modulation of the endogenous gut microbiota as a key to immune modulatory host responses. Our study demonstrated a single probiotic strain can influence local, regional, and systemic immunity through both innate and adaptive pathways in a strain-specific manner. It highlights the importance to investigate both the endogenous gut microbiome and the intestinal responses in response to probiotic supplementation, which underpins the mechanisms through which the probiotic strains drive the strain-specific effect to impact health outcomes.
Collapse
Affiliation(s)
- Bing Ma
- Institute of Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.
| | - Samuel J Gavzy
- Department of Surgery, University of Maryland Medical Center, Baltimore, MD, 21201, USA
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Vikas Saxena
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Yang Song
- Institute of Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Wenji Piao
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Hnin Wai Lwin
- Institute of Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Ram Lakhan
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Jegan Iyyathurai
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Lushen Li
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Michael France
- Institute of Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Christina Paluskievicz
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Marina W Shirkey
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Lauren Hittle
- Institute of Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Arshi Munawwar
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Emmanuel F Mongodin
- Institute of Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
- Division of Lung Diseases, National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Jonathan S Bromberg
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.
- Department of Surgery, University of Maryland Medical Center, Baltimore, MD, 21201, USA.
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.
| |
Collapse
|
27
|
Postbiotics in Human Health: A Narrative Review. Nutrients 2023; 15:nu15020291. [PMID: 36678162 PMCID: PMC9863882 DOI: 10.3390/nu15020291] [Citation(s) in RCA: 30] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 01/03/2023] [Accepted: 01/03/2023] [Indexed: 01/10/2023] Open
Abstract
In the 21st century, compressive health and functional foods are advocated by increasingly more people in order to eliminate sub-health conditions. Probiotics and postbiotics have gradually become the focus of scientific and nutrition communities. With the maturity and wide application of probiotics, the safety concerns and other disadvantages are non-negligible as we review here. As new-era products, postbiotics continue to have considerable potential as well as plentiful drawbacks to optimize. "Postbiotic" has been defined as a "preparation of inanimate microorganisms and/or their components that confers a health benefit on the host". Here, the evolution of the concept "postbiotics" is reviewed. The underlying mechanisms of postbiotic action are discussed. Current insight suggests that postbiotics exert efficacy through protective modulation, fortifying the epithelial barrier and modulation of immune responses. Finally, we provide an overview of the comparative advantages and the current application in the food industry at pharmaceutical and biomedical levels.
Collapse
|
28
|
Jung DR, Yoo HY, Kim MJ, Singh V, Park SH, Jeong M, Park BJ, Shin JH. Comparative analysis of scalp and gut microbiome in androgenetic alopecia: A Korean cross-sectional study. Front Microbiol 2022; 13:1076242. [PMID: 36578576 PMCID: PMC9791053 DOI: 10.3389/fmicb.2022.1076242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Accepted: 11/21/2022] [Indexed: 12/14/2022] Open
Abstract
Androgenetic alopecia (AGA) is a non-scarring and progressive form of hair loss occurring in both men and women. Although genetic predisposition and sex steroid hormones are the main causes, many factors remain unknown, and various extrinsic factors can negatively affect the lifespan of hair. We investigated skin-gut axis microorganisms as potential exogenous factors causing AGA, through comparative analyses of the scalp and gut microbiome in individuals with and without AGA in a Korean cohort. Using 16S rRNA gene sequencing, we characterized the scalp and gut microbiomes of 141 individuals divided into groups by sex and presence of AGA. Alpha diversity indices in the scalp microbiome were generally higher in individuals with AGA than in healthy controls. These indices showed a strong negative correlation with scalp-inhabitant bacteria (Cutibacterium and Staphylococcus), indicating that the appearance of non-inhabitant bacteria increases as hair loss progresses. No significant differences in diversity were observed between the gut microbiomes. However, bacterial functional differences, such as bile acid synthesis and bacterial invasion of epithelial cells, which are related to intestinal homeostasis, were observed. The networks of the scalp and gut microbiome were more complex and denser with higher values of the network topology statistic coefficient values (i.e., transitivity, density, and degree centrality) and more unique associations in individuals with AGA than in healthy controls. Our findings reveal a link between skin-gut microorganisms and AGA, indicating the former's potential involvement in the latter's development. Additionally, these results provide evidence for the development of cosmetics and therapeutics using microorganisms and metabolites involved in AGA.
Collapse
Affiliation(s)
- Da-Ryung Jung
- Department of Applied Biosciences, Kyungpook National University, Daegu, South Korea
| | - Hye-Young Yoo
- Skin and Natural Products Lab, Kolmar Korea Co., Ltd., Seoul, South Korea
| | - Min-Ji Kim
- Department of Applied Biosciences, Kyungpook National University, Daegu, South Korea
| | - Vineet Singh
- Department of Applied Biosciences, Kyungpook National University, Daegu, South Korea
| | - Sung-Ha Park
- Skin and Natural Products Lab, Kolmar Korea Co., Ltd., Seoul, South Korea
| | - Minsoo Jeong
- Department of Applied Biosciences, Kyungpook National University, Daegu, South Korea
| | - Byoung-Jun Park
- Skin and Natural Products Lab, Kolmar Korea Co., Ltd., Seoul, South Korea,*Correspondence: Byoung-Jun Park,
| | - Jae-Ho Shin
- Department of Applied Biosciences, Kyungpook National University, Daegu, South Korea,Department of Integrative Biotechnology, Kyungpook National University, Daegu, South Korea,NGS Core Facility, Kyungpook National University, Daegu, South Korea,Jae-Ho Shin,
| |
Collapse
|
29
|
Dang G, Wang W, Zhong R, Wu W, Chen L, Zhang H. Pectin supplement alleviates gut injury potentially through improving gut microbiota community in piglets. Front Microbiol 2022; 13:1069694. [PMID: 36569061 PMCID: PMC9780600 DOI: 10.3389/fmicb.2022.1069694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 11/16/2022] [Indexed: 12/14/2022] Open
Abstract
As pectin is widely used as a food and feed additive due to its tremendous prebiotic potentials for gut health. Yet, the underlying mechanisms associated with its protective effect remain unclear. Twenty-four piglets (Yorkshire × Landrace, 6.77 ± 0.92 kg) were randomly divided into three groups with eight replicates per treatment: (1) Control group (CON), (2) Lipopolysaccharide-challenged group (LPS), (3) Pectin-LPS group (PECL). Piglets were administrated with LPS or saline on d14 and 21 of the experiment. Piglets in each group were fed with corn-soybean meal diets containing 5% citrus pectin or 5% microcrystalline cellulose. Our result showed that pectin alleviated the morphological damage features by restoring the goblet numbers which the pig induced by LPS in the cecum. Besides, compared with the LPS group, pectin supplementation elevated the mRNA expression of tight junction protein [Claudin-1, Claudin-4, and zonula occludens-1 (ZO-1)], mucin (Muc-2), and anti-inflammatory cytokines [interleukin 10 (IL-10), and IL-22]. Whereas pectin downregulated the expression of proinflammatory cytokines (IL-1β, IL-6, IL-18), tumor necrosis factor-&alpha (TNF-α), and NF-κB. What is more, pectin supplementation also significantly increased the abundance of beneficial bacteria (Lactobacillus, Clostridium_sensu_stricto_1, Blautia, and Subdoligranulum), and significantly reduced the abundance of harmful bacteria, such as Streptococcus. Additionally, pectin restored the amount of short-chain fatty acids (SCFAs) after being decreased by LPS (mainly Acetic acid, Propionic acid, and Butyric acid) to alleviate gut injury and improve gut immunity via activating relative receptors (GPR43, GPR109, AhR). Mantel test and correlation analysis also revealed associations between intestinal microbiota and intestinal morphology, and intestinal inflammation in piglets. Taken together, dietary pectin supplementation enhances the gut barrier and improves immunity to ameliorate LPS-induced injury by optimizing gut microbiota and their metabolites.
Collapse
Affiliation(s)
- Guoqi Dang
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China,Precision Livestock and Nutrition Unit, Gembloux Agro-Bio Tech, TERRA Teaching and Research Centre, Liège University, Gembloux, Belgium
| | - Wenxing Wang
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Ruqing Zhong
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Weida Wu
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Liang Chen
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China,*Correspondence: Liang Chen,
| | - Hongfu Zhang
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China,Hongfu Zhang,
| |
Collapse
|
30
|
Karbowiak M, Gałek M, Szydłowska A, Zielińska D. The Influence of the Degree of Thermal Inactivation of Probiotic Lactic Acid Bacteria and Their Postbiotics on Aggregation and Adhesion Inhibition of Selected Pathogens. Pathogens 2022; 11:1260. [PMID: 36365011 PMCID: PMC9692860 DOI: 10.3390/pathogens11111260] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 10/25/2022] [Accepted: 10/25/2022] [Indexed: 07/25/2023] Open
Abstract
The study aimed to evaluate the effect of thermal inactivation of potentially probiotic lactic acid bacteria (LAB) strains isolated from food on their ability to compete with pathogenic microorganisms. Five strains of LAB, previously isolated from food and characterized, one commercial reference strain of Lactiplantibacillus plantarum 299v, and two indicator strains of Staphylococcus aureus 25923 and Listeriamonocytogenes 15313 were used in the study. The experiment consisted in applying a stress factor (high temperature: 80 °C, at a different time: 5, 15, and 30 min) to the tested LAB cells to investigate the in vitro properties such as hydrophobicity abilities (against p-xylene and n-hexadecane), auto-aggregation, co-aggregation with pathogens, and inhibition of pathogens adhesion to the porcine gastric mucin. The bacterial strains showed various hydrophobicity to p-xylene (36-73%) and n-hexadecane (11-25%). The affinity for solvents expanded with increasing thermal inactivation time. All LAB isolates were able to auto-aggregate (ranging from 17 to 49%). Bacterial strains subjected to 5 and 15 min of thermal inactivation had the highest auto-aggregation ability in comparison to viable and heat-killed cells for 30 min. The LAB strains co-aggregated with pathogens to different degrees; among them, the highest scores of co-aggregation were observed for L. monocytogenes, reaching 27% (with 15 min of heat-killed LAB cells). All LAB strains reduced the adherence of pathogenic bacteria in the competition test, moreover, heat-killed cells (especially 15 min inactivated) were more efficient than viable cells. The properties of selected LAB strains as moderately heat-stressed forms analyzed in the study increased the prevention of colonization and elimination of pathogenic bacteria in the in vitro model of gastrointestinal tract. The thermal inactivation process may therefore preserve and modifies some characteristics of bacterial cells.
Collapse
|
31
|
Ghosh S, Sarkar B, Kaushik A, Mostafavi E. Nanobiotechnological prospects of probiotic microflora: Synthesis, mechanism, and applications. THE SCIENCE OF THE TOTAL ENVIRONMENT 2022; 838:156212. [PMID: 35623529 DOI: 10.1016/j.scitotenv.2022.156212] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 05/20/2022] [Accepted: 05/20/2022] [Indexed: 06/15/2023]
Abstract
Nanotechnology-driven solutions have almost touched every aspect of life, such as therapeutics, cosmetics, agriculture, and the environment. Physical and chemical methods for the synthesis of nanoparticles involve hazardous reaction conditions and toxic reducing as well as stabilizing agents. Hence, environmentally benign green routes are preferred to synthesize nanoparticles with tunable size and shape. Bacteria, fungi, algae, and medicinal plants are employed to synthesize gold, silver, copper, zinc, and other nanoparticles. However, very little literature is available on exploring probiotic bacteria for the synthesis of nanoparticles. In view of the background, this review gives the most comprehensive report on the nanobiotechnological potential of probiotic bacteria like Bacillus licheniformis, Bifidobacterium animalis, Brevibacterium linens, Lactobacillus acidophilus, Lactobacillus casei, and others for the synthesis of gold (AuNPs), selenium (SeNPs), silver (AgNPs), platinum (PtNPs), tellurium nanoparticles (TeNPs), zinc oxide (ZnONPs), copper oxide (CuONPs), iron oxide (Fe3O4NPs), and titanium oxide nanoparticles (TiO2NPs). Both intracellular and extracellular synthesis are involved as potential routes for biofabrication of polydispersed nanoparticles that are spherical, rod, or hexagonal in shape. Capsular exopolysaccharide associated carbohydrates such as galactose, glucose, mannose, and rhamnose, cell membrane-associated diglycosyldiacylglycerol (DGDG), 1,2-di-O-acyl-3-O-[O-α-D-galactopyranosyl-(1 → 2)-α-d-glucopyranosyl]glycerol, triglycosyl diacylglycerol (TGDG), NADH-dependent enzymes, amino acids such as cysteine, tyrosine, and tryptophan, S-layer proteins (SLP), lacto-N-triose, and lactic acid play a significant role in synthesis and stabilization of the nanoparticles. The biogenic nanoparticles can be recovered by rational treatment with sodium dodecyl sulfate (SDS) and/or sodium hydroxide (NaOH). Eventually, diverse applications like antibacterial, antifungal, anticancer, antioxidant, and other associated activities of the bacteriogenic nanoparticles are also elaborated. Being more biocompatible and effective, probiotic-generated nanoparticles can be explored as novel nutraceuticals for their ability to ensure sustained release and bioavailability of the loaded bioactive ingredients for diagnosis, targeted drug delivery, and therapy.
Collapse
Affiliation(s)
- Sougata Ghosh
- Department of Microbiology, School of Science, RK University, Rajkot, Gujarat, India
| | | | - Ajeet Kaushik
- NanoBioTech Laboratory, Health Systems Engineering, Department of Environmental Engineering, Florida Polytechnic University, Lakeland, FL 33805, USA; School of Engineering, University of Petroleum and Energy Studies (UPES), Dehradun, Uttarakhand, India
| | - Ebrahim Mostafavi
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA; Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
32
|
Chai HH, Ham JS, Kim TH, Lim D. Identifying ligand-binding specificity of the oligopeptide receptor OppA from Bifidobacterium longum KACC91563 by structure-based molecular modeling. ARAB J CHEM 2022. [DOI: 10.1016/j.arabjc.2022.104198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
|
33
|
Therapeutic effect of heat-killed Lactobacillus plantarum L-137 on the gut health and growth of broilers. Acta Trop 2022; 232:106537. [PMID: 35623400 DOI: 10.1016/j.actatropica.2022.106537] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 05/20/2022] [Accepted: 05/22/2022] [Indexed: 11/21/2022]
Abstract
Although some studies on the effects of para-probiotics on the immune system and intestinal health have been conducted independently of research on antibiotics ass growth promoters. This study investigated the effects of heat-killed Lactobacillus plantarumL-137 (L-137) and antibiotics as preventive and/or therapeutic substances for broilers against subclinical necrotic enteritis caused by Clostridium perfringens (CP). In total, 300 1-day-old broilers (46.13 ± 1.38 g) were randomly stocked at 10 birds pen-1 in five replicates and divided into six groups, namely T1 and T2, positive and negative control of CP challenge; T3 and T4, prevention with basal diet plus 10 and 50 mg/kg L-137; T5 and T6, prevention and treatment with basal diet plus 50 mg/kg of L-137 and bacitracin at 50 ppm, respectively. Broilers administered L-137 in T4, T5 and bacitracin in T6 showed an improved (p < 0.05) villus height/crypt depth ratio than control groups, suggesting that it might significantly boost growth performance. In contrast to bacitracin, a high dosage of L-137 significantly increased (p < 0.05) the spleen index value and the cytokine levels, as well as the expression of intestinal β-defensin genes on day 28. During the 42-day production period, broilers in T4 and T5 showed a significantly enhanced (p < 0.05) expression of cytokines, AvBD-1 and AvBD-7 on day 42 compared to the control and bacitracin groups. In particular, broilers given the L-137 diets demonstrated no cumulative mortality following CP exposure, compared to a 2% mortality in T6. Our findings provide insight into eco-friendly alternatives to antibiotics for maximizing growth performance, feed efficiency and long-term disease protection in chickens; however, this has to be proven in larger-scale commercial experiments.
Collapse
|
34
|
Wang M, Zhang Z, Sun H, He S, Liu S, Zhang T, Wang L, Ma G. Research progress of anthocyanin prebiotic activity: A review. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 102:154145. [PMID: 35567994 DOI: 10.1016/j.phymed.2022.154145] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 04/22/2022] [Accepted: 05/01/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND Anthocyanins are a kind of flavonoids and natural water-soluble pigments, which endow fruits, vegetables, and plants with multiple colors. They are important source of new products with prebiotic activity. However, there is no systematic review documenting prebiotic activity of anthocyanins and their structural analogues. This study aims to fill this gap in literature. PURPOSE The objective of this review is to summarize and evaluate the prebiotic activity of anthocyanin's, and discuss the physical and molecular modification methods to improve their biological activities. STUDY DESIGN AND METHODS In this review, the databases (PubMed, Google Scholar, Web of Science, Researchgate and Elsevier) were searched profoundly with keywords (anthocyanin's, prebiotics, probiotics, physical embedding and molecular modification). RESULTS A total of 34 articles were considered for reviewing. These studies approved that anthocyanins play an important role in promoting the proliferation of probiotics, inhibiting the growth of harmful bacteria and improving the intestinal environment. In addition, physical embedding and molecular modification have also been proved to be effective methods to improve the prebiotic activity of anthocyanins. Anthocyanins could promote the production of short chain fatty acids, accelerate self degradation and improve microbial related enzyme activities to promote the proliferation of probiotics. They inhibited the growth of harmful bacteria by inhibiting the expression of harmful bacteria genes, interfering with the role of metabolism related enzymes and affecting respiratory metabolism. They promoted the formation of a complete intestinal barrier and regulated the intestinal environment to keep the body healthy. Physical embedding, including microencapsulation and colloidal embedding, greatly improved the stability of anthocyanins. On the other hand, molecular modification, especially enzymatic modification, significantly improved the biological activities (antioxidant, prebiotic activity and so on) of anthocyanins. CONCLUSION All these research results displayed by this review indicate that anthocyanins are a useful tool for developing prebiotic products. The better activities of the new anthocyanins formed by embedding and modification may make them become more effective raw materials. Our review provides a scientific basis for the future research and application of anthocyanins.
Collapse
Affiliation(s)
- Muwen Wang
- School of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, P.R. China
| | - Zuoyong Zhang
- School of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, P.R. China
| | - Hanju Sun
- School of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, P.R. China.
| | - Shudong He
- School of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, P.R. China.
| | - Shuyun Liu
- School of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, P.R. China
| | - Tao Zhang
- School of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, P.R. China
| | - Lei Wang
- School of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, P.R. China
| | - Gang Ma
- School of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, P.R. China
| |
Collapse
|
35
|
Jang HM, Kim JK, Joo MK, Shin YJ, Lee KE, Lee CK, Kim HJ, Kim DH. Enterococcus faecium and Pediococcus acidilactici deteriorate Enterobacteriaceae-induced depression and colitis in mice. Sci Rep 2022; 12:9389. [PMID: 35672451 PMCID: PMC9174183 DOI: 10.1038/s41598-022-13629-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 05/11/2022] [Indexed: 01/14/2023] Open
Abstract
Gut dysbiosis is closely associated with the outbreak of inflammatory bowel disease (IBD) and psychiatric disorder. The Enterobacteriaceae population was higher in the feces of patients with inflammatory bowel disease (IBD-F) than in those of healthy control volunteers (HC-F). The Enterococcaceae and Lactobacillaceae populations were higher in the feces of IBD patients with depression (IBD/D+-F) vs. the feces of IBD patients without depression (IBD/D--F). Therefore, we examined the effects of Klebsiella oxytoca, Escherichia coli, Cronobacter sakazakii, Enterococcus faecium, and Pediococcus acidolactici overpopulated in IBD/D+-F and their byproducts LPS and exopolysaccharide (EPS) on the occurrence of depression and colitis in mice. Oral gavages of Klebsiella oxytoca, Escherichia coli, and Cronobacter sakazakii belonging to Enterobacteriaceae, singly or together, caused dose-dependently colitis and depression-like behaviors in germ-free and specific-pathogen-free mice. Although Enterococcus faecium and Pediococcus acidolactici did not significantly cause colitis and depression-like behaviors, they significantly deteriorated Klebsiella oxytoca- or Escherichia coli-induced colitis, neuroinflammation, and anxiety/depression-like behaviors and increased blood LPS, corticosterone, and IL-6 levels. The EPSs from Enterococcus faecium and Pediococcus acidolactici also worsened Klebsiella oxytoca LPS-induced colitis, neuroinflammation, and depression-like behaviors in mice and increased the translocation of fluorescein isothiocyanate-conjugated LPS into the hippocampus. However, Bifidobacterium longum, which was lower in IBD/D+-F vs. IBD/D--F, or its EPS suppressed them. In conclusion, Enterococcus faecium and Pediococcus acidolactici, known as a probiotic strain, and their EPSs may be a risk factor for the outbreak of depression and IBD.
Collapse
Affiliation(s)
- Hyo-Min Jang
- Neurobiota Research Center, College of Pharmacy, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul, 02447, Korea
| | - Jeon-Kyung Kim
- Neurobiota Research Center, College of Pharmacy, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul, 02447, Korea
- College of Pharmacy, Jeonbuk National University, 26, Jeonju, 54896, Korea
| | - Min-Kyung Joo
- Neurobiota Research Center, College of Pharmacy, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul, 02447, Korea
| | - Yoon-Jung Shin
- Neurobiota Research Center, College of Pharmacy, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul, 02447, Korea
| | - Kyung-Eon Lee
- Neurobiota Research Center, College of Pharmacy, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul, 02447, Korea
| | - Chang Kyun Lee
- Department of Internal Medicine, Kyung Hee University School of Medicine, Seoul, 02447, Korea
| | - Hyo-Jong Kim
- Department of Internal Medicine, Kyung Hee University School of Medicine, Seoul, 02447, Korea
| | - Dong-Hyun Kim
- Neurobiota Research Center, College of Pharmacy, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul, 02447, Korea.
| |
Collapse
|
36
|
Louis P, Duncan SH, Sheridan PO, Walker AW, Flint HJ. Microbial lactate utilisation and the stability of the gut microbiome. GUT MICROBIOME (CAMBRIDGE, ENGLAND) 2022; 3:e3. [PMID: 39295779 PMCID: PMC11406415 DOI: 10.1017/gmb.2022.3] [Citation(s) in RCA: 41] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 04/05/2022] [Accepted: 04/07/2022] [Indexed: 09/21/2024]
Abstract
The human large intestinal microbiota thrives on dietary carbohydrates that are converted to a range of fermentation products. Short-chain fatty acids (acetate, propionate and butyrate) are the dominant fermentation acids that accumulate to high concentrations in the colon and they have health-promoting effects on the host. Although many gut microbes can also produce lactate, it usually does not accumulate in the healthy gut lumen. This appears largely to be due to the presence of a relatively small number of gut microbes that can utilise lactate and convert it to propionate, butyrate or acetate. There is increasing evidence that these microbes play important roles in maintaining a healthy gut environment. In this review, we will provide an overview of the different microbes involved in lactate metabolism within the gut microbiota, including biochemical pathways utilised and their underlying energetics, as well as regulation of the corresponding genes. We will further discuss the potential consequences of perturbation of the microbiota leading to lactate accumulation in the gut and associated disease states and how lactate-utilising bacteria may be employed to treat such diseases.
Collapse
Affiliation(s)
- Petra Louis
- Gut Health Group, Rowett Institute, University of Aberdeen, Aberdeen, UK
| | | | | | | | - Harry James Flint
- Gut Health Group, Rowett Institute, University of Aberdeen, Aberdeen, UK
| |
Collapse
|
37
|
Derrien M, Turroni F, Ventura M, van Sinderen D. Insights into endogenous Bifidobacterium species in the human gut microbiota during adulthood. Trends Microbiol 2022; 30:940-947. [DOI: 10.1016/j.tim.2022.04.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 04/14/2022] [Accepted: 04/19/2022] [Indexed: 01/25/2023]
|
38
|
The Influence of Gut Dysbiosis in the Pathogenesis and Management of Ischemic Stroke. Cells 2022; 11:cells11071239. [PMID: 35406804 PMCID: PMC8997586 DOI: 10.3390/cells11071239] [Citation(s) in RCA: 64] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 03/21/2022] [Accepted: 03/25/2022] [Indexed: 12/12/2022] Open
Abstract
Recent research on the gut microbiome has revealed the influence of gut microbiota (GM) on ischemic stroke pathogenesis and treatment outcomes. Alterations in the diversity, abundance, and functions of the gut microbiome, termed gut dysbiosis, results in dysregulated gut–brain signaling, which induces intestinal barrier changes, endotoxemia, systemic inflammation, and infection, affecting post-stroke outcomes. Gut–brain interactions are bidirectional, and the signals from the gut to the brain are mediated by microbially derived metabolites, such as trimethylamine N-oxide (TMAO) and short-chain fatty acids (SCFAs); bacterial components, such as lipopolysaccharide (LPS); immune cells, such as T helper cells; and bacterial translocation via hormonal, immune, and neural pathways. Ischemic stroke affects gut microbial composition via neural and hypothalamic–pituitary–adrenal (HPA) pathways, which can contribute to post-stroke outcomes. Experimental and clinical studies have demonstrated that the restoration of the gut microbiome usually improves stroke treatment outcomes by regulating metabolic, immune, and inflammatory responses via the gut–brain axis (GBA). Therefore, restoring healthy microbial ecology in the gut may be a key therapeutic target for the effective management and treatment of ischemic stroke.
Collapse
|
39
|
Mathur H, Linehan K, Flynn J, Byrne N, Dillon P, Conneely M, Grimaud G, Hill C, Stanton C, Ross RP. Emulsion-Based Postbiotic Formulation Is Comparable to Viable Cells in Eliciting a Localized Immune Response in Dairy Cows With Chronic Mastitis. Front Microbiol 2022; 13:759649. [PMID: 35391729 PMCID: PMC8981918 DOI: 10.3389/fmicb.2022.759649] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 01/05/2022] [Indexed: 11/13/2022] Open
Abstract
Bovine mastitis is a disease with a multi-etiological nature, defined as an infection and inflammation of the udder. Mastitis represents a significant ongoing concern in the dairy industry, leading to substantial losses in profits and revenue for farmers worldwide. The predominant causes of bovine mastitis include the pathogens Staphylococcus aureus, Streptococcus dysgalactiae, Streptococcus uberis, and Escherichia coli. Antibiotic administration is currently the main treatment option for mastitis. However, there is a pressing need for alternative therapies to treat and prevent the disease, especially with the emergence of antibiotic-resistant, mastitis-causing pathogens, resulting in antibiotic treatment failure. One such example is live bio-therapeutics (also known as probiotics), such as Lactococcus lactis DPC3147. The efficacy of this live bio-therapeutic has been demonstrated in several previous trials by our group. The most recent of these trials showed that an emulsion-based formulation of this strain was as effective as a commercial antibiotic formulation in treating sub-clinical and clinical cases of bovine mastitis. Here, we report the results of a follow-up field trial, in which we sought to gain insight into the mechanism of action of such live bio-therapeutics, focussing on chronic mastitis cases. We treated 28 cows with chronic mastitis with two separate emulsion-based formulations containing either viable L. lactis DPC3147 cells (15 cows) or heat-killed L. lactis DPC3147 cells (13 cows). We then evaluated the efficacies of the two formulations (two treatment groups) in terms of stimulating a localized immune response (quantified by measuring IL-8 concentrations in milk collected from udders affected by mastitis) and efficacies in terms of cure rates (quantified by reductions in somatic cell counts and absence of pathogens). We demonstrate that the presence of heat-inactivated bacteria (a postbiotic) was as effective as the live bio-therapeutic in eliciting a localized immune response in cows with chronic mastitis. The response to heat-killed cells (postbiotic) reported herein could have beneficial implications for farmers with regard to prolonging the shelf life of such emulsion-based formulations containing heat-killed cells of L. lactis DPC3147 for curing cows with mastitis.
Collapse
Affiliation(s)
- Harsh Mathur
- Teagasc Food Research Centre, Moorepark, Fermoy, Ireland
| | - Kevin Linehan
- Teagasc Food Research Centre, Moorepark, Fermoy, Ireland
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- School of Microbiology, University College Cork, Cork, Ireland
| | - James Flynn
- Dairy Production Research Centre, Teagasc, Moorepark, Fermoy, Ireland
| | - Noel Byrne
- Teagasc Animal and Grassland Research and Innovation Centre, Moorepark, Fermoy, Ireland
| | - Pat Dillon
- Teagasc Animal and Grassland Research and Innovation Centre, Moorepark, Fermoy, Ireland
| | - Muireann Conneely
- Teagasc Animal and Grassland Research and Innovation Centre, Moorepark, Fermoy, Ireland
| | - Ghjuvan Grimaud
- Teagasc Food Research Centre, Moorepark, Fermoy, Ireland
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Colin Hill
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- School of Microbiology, University College Cork, Cork, Ireland
| | - Catherine Stanton
- Teagasc Food Research Centre, Moorepark, Fermoy, Ireland
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - R. Paul Ross
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- School of Microbiology, University College Cork, Cork, Ireland
| |
Collapse
|
40
|
Su J, Ren Y, Liu L, Hu Y, Shi H, Ren J, Xie C. Decreased serum iron concentration and total iron binding capacity are associated with serious Crohn's disease. Sci Rep 2022; 12:3923. [PMID: 35273280 PMCID: PMC8913652 DOI: 10.1038/s41598-022-07948-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2021] [Accepted: 02/14/2022] [Indexed: 11/17/2022] Open
Abstract
This study aimed to investigate whether serum indicators related to iron stores in the body are associated with clinical and endoscopic disease severity. Eighty-four patients with Crohn’s disease (CD) and twenty-four healthy volunteers were included. The indicators related to iron stores were detected within one week after endoscopic and CT enterography examinations. Patients were divided into three groups according to the CDAI(Crohn's disease activity index)scores. Serum iron levels were decreased in all groups (p < 0.05), and the values of remission group were higher than those of moderate group (p < 0.001). The total iron binding capacity(TIBC)values of the moderate group were lower than those of the controls and the other groups (p < 0.05). None of the indicators differed significantly among the patients classified by SES-CD (p > 0.05). Underweight, decreased serum iron and TIBC were independent risk factors for moderate clinical disease. Combined detection of decreased serum iron and TIBC was helpful in differentiating severe patients. The sensitivity and specificity were 32.7% and 100%, respectively (AUC = 0.812, p < 0.01). Decreases in serum iron and TIBC were associated with the clinical activity of CD. Combined detection of the two indicators was conducive to screening serious disease.
Collapse
Affiliation(s)
- Jingling Su
- Department of Gastroenterology, Zhongshan Hospital Xiamen University, Xiamen, 361000, Fujian Province, China
| | - Yandan Ren
- Department of Gastroenterology, Zhongshan Hospital Xiamen University, Xiamen, 361000, Fujian Province, China
| | - Lupeng Liu
- Department of Gastroenterology, Zhongshan Hospital Xiamen University, Xiamen, 361000, Fujian Province, China
| | - Yiqun Hu
- Department of Gastroenterology, Zhongshan Hospital Xiamen University, Xiamen, 361000, Fujian Province, China
| | - Huaxiu Shi
- Department of Gastroenterology, Zhongshan Hospital Xiamen University, Xiamen, 361000, Fujian Province, China
| | - Jianlin Ren
- Department of Gastroenterology, Zhongshan Hospital Xiamen University, Xiamen, 361000, Fujian Province, China
| | - Chenxi Xie
- Department of Gastroenterology, Zhongshan Hospital Xiamen University, Xiamen, 361000, Fujian Province, China.
| |
Collapse
|
41
|
Human milk oligosaccharide-sharing by a consortium of infant derived Bifidobacterium species. Sci Rep 2022; 12:4143. [PMID: 35264656 PMCID: PMC8907170 DOI: 10.1038/s41598-022-07904-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Accepted: 02/24/2022] [Indexed: 11/08/2022] Open
Abstract
Bifidobacteria are associated with a host of health benefits and are typically dominant in the gut microbiota of healthy, breast-fed infants. A key adaptation, facilitating the establishment of these species, is their ability to consume particular sugars, known as human milk oligosaccharides (HMO), which are abundantly found in breastmilk. In the current study, we aimed to characterise the co-operative metabolism of four commercial infant-derived bifidobacteria (Bifidobacterium bifidum R0071, Bifidobacterium breve M-16V, Bifidobacterium infantis R0033, and Bifidobacterium infantis M-63) when grown on HMO. Three different HMO substrates (2'-fucosyllactose alone and oligosaccharides isolated from human milk representing non-secretor and secretor status) were employed. The four-strain combination resulted in increased bifidobacterial numbers (> 21%) in comparison to single strain cultivation. The relative abundance of B. breve increased by > 30% during co-cultivation with the other strains despite demonstrating limited ability to assimilate HMO in mono-culture. HPLC analysis revealed strain-level variations in HMO consumption. Metabolomics confirmed the production of formate, acetate, 1,2-propanediol, and lactate with an overall increase in such metabolites during co-cultivation. These results support the concept of positive co-operation between multiple bifidobacterial strains during HMO utilisation which may result in higher cell numbers and a potentially healthier balance of metabolites.
Collapse
|
42
|
Anto L, Blesso CN. Interplay Between Diet, the Gut Microbiome, and Atherosclerosis: Role of Dysbiosis and Microbial Metabolites on Inflammation and Disordered Lipid Metabolism. J Nutr Biochem 2022; 105:108991. [DOI: 10.1016/j.jnutbio.2022.108991] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 12/21/2021] [Accepted: 02/22/2022] [Indexed: 12/16/2022]
|
43
|
Vemuri R, Ruggiero A, Whitfield JM, Dugan GO, Cline JM, Block MR, Guo H, Kavanagh K. Hypertension promotes microbial translocation and dysbiotic shifts in the fecal microbiome of nonhuman primates. Am J Physiol Heart Circ Physiol 2022; 322:H474-H485. [PMID: 35148233 PMCID: PMC8897002 DOI: 10.1152/ajpheart.00530.2021] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Accumulating evidence indicates a link between gut barrier dysfunction and hypertension. However, it is unclear whether hypertension causes gut barrier dysfunction or vice versa and whether the gut microbiome plays a role. To understand this relationship, we first cross-sectionally examined 153 nonhuman primates [NHPs; Chlorocebus aethiops sabaeus; mean age, 16 ± 0.4 yr; 129 (84.3%) females] for cardiometabolic risk factors and gut barrier function biomarkers. This analysis identified blood pressure and age as specific factors that independently associated with microbial translocation. We then longitudinally tracked male, age-matched spontaneously hypertensive NHPs (Macaca mulatta) to normotensives (n = 16), mean age of 5.8 ± 0.5 yr, to confirm hypertension-related gut barrier dysfunction and to explore the role of microbiome by comparing groups at baseline, 12, and 27 mo. Collectively, hypertensive animals in both studies showed evidence of gut barrier dysfunction (i.e., microbial translocation), as indicated by higher plasma levels of lipopolysaccharide-binding protein (LBP)-1, when compared with normotensive animals. Furthermore, plasma LBP-1 levels were correlated with diastolic blood pressure, independent of age and other health markers, suggesting specificity of the effect of hypertension on microbial translocation. In over 2 yr of longitudinal assessment, hypertensive animals had escalating plasma levels of LBP-1 and greater bacterial gene expression in mesenteric lymph nodes compared with normotensive animals, confirming microbes translocated across the intestinal barrier. Concomitantly, we identified distinct shifts in the gut microbial signature of hypertensive versus normotensive animals at 12 and 27 mo. These results suggest that hypertension contributes to microbial translocation in the gut and eventually unhealthy shifts in the gut microbiome, possibly contributing to poor health outcomes, providing further impetus for the management of hypertension.NEW & NOTEWORTHY Hypertension specifically had detrimental effects on microbial translocation when age and metabolic syndrome criteria were evaluated as drivers of cardiovascular disease in a relevant nonhuman primate model. Intestinal barrier function exponentially decayed over time with chronic hypertension, and microbial translocation was confirmed by detection of more microbial genes in regional draining lymph nodes. Chronic hypertension resulted in fecal microbial dysbiosis and elevations of the biomarker NT-proBNP. This study provides insights on the barrier dysfunction, dysbiosis, and hypertension in controlled studies of nonhuman primates. Our study includes a longitudinal component comparing naturally occurring hypertensive to normotensive primates to confirm microbial translocation and dysbiotic microbiome development. Hypertension is an underappreciated driver of subclinical endotoxemia that can drive chronic inflammatory diseases.
Collapse
Affiliation(s)
- Ravichandra Vemuri
- 1Section on Comparative Medicine, Department of Pathology, Wake Forest School of Medicine, Winston‐Salem, North Carolina
| | - Alistaire Ruggiero
- 1Section on Comparative Medicine, Department of Pathology, Wake Forest School of Medicine, Winston‐Salem, North Carolina
| | - Jordyn M. Whitfield
- 1Section on Comparative Medicine, Department of Pathology, Wake Forest School of Medicine, Winston‐Salem, North Carolina
| | - Greg O. Dugan
- 1Section on Comparative Medicine, Department of Pathology, Wake Forest School of Medicine, Winston‐Salem, North Carolina
| | - J. Mark Cline
- 1Section on Comparative Medicine, Department of Pathology, Wake Forest School of Medicine, Winston‐Salem, North Carolina
| | - Masha R. Block
- 1Section on Comparative Medicine, Department of Pathology, Wake Forest School of Medicine, Winston‐Salem, North Carolina
| | - Hao Guo
- 2Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina,3Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Kylie Kavanagh
- 1Section on Comparative Medicine, Department of Pathology, Wake Forest School of Medicine, Winston‐Salem, North Carolina,4Department of Biomedicine, University of Tasmania, Hobart, Tasmania, Australia
| |
Collapse
|
44
|
Nazhand A, Durazzo A, Lucarini M, Guerra F, Souto SB, Souto EB, Santini A. Nutraceuticals and functional beverages: Focus on Prebiotics and Probiotics active beverages. FUTURE FOODS 2022. [DOI: 10.1016/b978-0-323-91001-9.00026-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
|
45
|
Venardou B, O’Doherty JV, Garcia-Vaquero M, Kiely C, Rajauria G, McDonnell MJ, Ryan MT, Sweeney T. Evaluation of the Antibacterial and Prebiotic Potential of Ascophyllum nodosum and Its Extracts Using Selected Bacterial Members of the Pig Gastrointestinal Microbiota. Mar Drugs 2021; 20:41. [PMID: 35049896 PMCID: PMC8778111 DOI: 10.3390/md20010041] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 12/22/2021] [Accepted: 12/27/2021] [Indexed: 11/28/2022] Open
Abstract
Ascophyllum nodosum and its extracts are promising antibacterial and prebiotic dietary supplements for pigs. The objectives of this study were to evaluate the effects of the increasing concentrations of: (1) two whole biomass samples of A. nodosum with different harvest seasons, February (ANWB-F) and November (ANWB-N), in a weaned pig faecal batch fermentation assay, and (2) A. nodosum extracts produced using four different extraction conditions of a hydrothermal-assisted extraction methodology (ANE1-4) and conventional extraction methods with water (ANWE) and ethanol (ANEE) as solvent in individual pure culture growth assays using a panel of beneficial and pathogenic bacterial strains. In the batch fermentation assay, ANWB-F reduced Bifidobacterium spp. counts (p < 0.05) while ANWB-N increased total bacterial counts and reduced Bifidobacterium spp. and Enterobacteriaceae counts (p < 0.05). Of the ANE1-4, produced from ANWB-F, ANWE and ANEE that were evaluated in the pure culture growth assays, the most interesting extracts were the ANE1 that reduced Salmonella Typhimurium, enterotoxigenic Escherichia coli and B. thermophilum counts and the ANE4 that stimulated B. thermophilum growth (p < 0.05). In conclusion, the extraction method and conditions influenced the bioactivities of the A. nodosum extracts with ANE1 and ANE4 exhibiting distinct antibacterial and prebiotic properties in vitro, respectively, that merit further exploration.
Collapse
Affiliation(s)
- Brigkita Venardou
- School of Veterinary Medicine, University College Dublin, Belfield, D04 V1W8 Dublin 4, Ireland; (B.V.); (M.T.R.)
| | - John V. O’Doherty
- School of Agriculture and Food Science, University College Dublin, Belfield, D04 V1W8 Dublin 4, Ireland; (J.V.O.); (M.G.-V.); (C.K.); (G.R.); (M.J.M.)
| | - Marco Garcia-Vaquero
- School of Agriculture and Food Science, University College Dublin, Belfield, D04 V1W8 Dublin 4, Ireland; (J.V.O.); (M.G.-V.); (C.K.); (G.R.); (M.J.M.)
| | - Claire Kiely
- School of Agriculture and Food Science, University College Dublin, Belfield, D04 V1W8 Dublin 4, Ireland; (J.V.O.); (M.G.-V.); (C.K.); (G.R.); (M.J.M.)
| | - Gaurav Rajauria
- School of Agriculture and Food Science, University College Dublin, Belfield, D04 V1W8 Dublin 4, Ireland; (J.V.O.); (M.G.-V.); (C.K.); (G.R.); (M.J.M.)
| | - Mary J. McDonnell
- School of Agriculture and Food Science, University College Dublin, Belfield, D04 V1W8 Dublin 4, Ireland; (J.V.O.); (M.G.-V.); (C.K.); (G.R.); (M.J.M.)
| | - Marion T. Ryan
- School of Veterinary Medicine, University College Dublin, Belfield, D04 V1W8 Dublin 4, Ireland; (B.V.); (M.T.R.)
| | - Torres Sweeney
- School of Veterinary Medicine, University College Dublin, Belfield, D04 V1W8 Dublin 4, Ireland; (B.V.); (M.T.R.)
| |
Collapse
|
46
|
Roussel C, De Paepe K, Galia W, de Bodt J, Chalancon S, Denis S, Leriche F, Vandekerkove P, Ballet N, Blanquet-Diot S, Van de Wiele T. Multi-targeted properties of the probiotic saccharomyces cerevisiae CNCM I-3856 against enterotoxigenic escherichia coli (ETEC) H10407 pathogenesis across human gut models. Gut Microbes 2021; 13:1953246. [PMID: 34432600 PMCID: PMC8405159 DOI: 10.1080/19490976.2021.1953246] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Enterotoxigenic Escherichia coli (ETEC) is one of the most common causes of acute traveler's diarrhea. Adhesins and enterotoxins constitute the major ETEC virulence traits. With the dramatic increase in antibiotic resistance, probiotics are considered a wholesome alternative to prevent or treat ETEC infections. Here, we examined the antimicrobial properties of the probiotic Saccharomyces cerevisiae CNCM I-3856 against ETEC H10407 pathogenesis upon co-administration in the TNO gastrointestinal Model (TIM-1), simulating the physicochemical and enzymatic conditions of the human upper digestive tract and preventive treatment in the Mucosal Simulator of the Human Intestinal Microbial Ecosystem (M-SHIME), integrating microbial populations of the ileum and ascending colon. Interindividual variability was assessed by separate M-SHIME experiments with microbiota from six human individuals. The probiotic did not affect ETEC survival along the digestive tract. However, ETEC pathogenicity was significantly reduced: enterotoxin encoding virulence genes were repressed, especially in the TIM-1 system, and a lower enterotoxin production was noted. M-SHIME experiments revealed that 18-days probiotic treatment stimulate the growth of Bifidobacterium and Lactobacillus in different gut regions (mucosal and luminal, ileum and ascending colon) while a stronger metabolic activity was noted in terms of short-chain fatty acids (acetate, propionate, and butyrate) and ethanol production. Moreover, the probiotic pre-treated microbiota displayed a higher robustness in composition following ETEC challenge compared to the control condition. We thus demonstrated the multi-inhibitory properties of the probiotic S. cerevisiae CNCM I-3856 against ETEC in the overall simulated human digestive tract, regardless of the inherent variability across individuals in the M-SHIME.
Collapse
Affiliation(s)
- Charlène Roussel
- Université Clermont Auvergne, UMR UCA-INRA 454 MEDIS, Microbiology Digestive Environment and Health, Clermont-Ferrand, France,CMET, Center for Microbial Ecology and Technology, Department of Biotechnology, Faculty of Bioscience Engineering, Ghent University, Ghent, Belgium
| | - Kim De Paepe
- CMET, Center for Microbial Ecology and Technology, Department of Biotechnology, Faculty of Bioscience Engineering, Ghent University, Ghent, Belgium
| | - Wessam Galia
- UMR 5557 Microbial Ecology, Research Group On Bacterial Opportunistic Pathogens And Environment, CNRS, VetAgro Sup, Lyon, France
| | - Jana de Bodt
- CMET, Center for Microbial Ecology and Technology, Department of Biotechnology, Faculty of Bioscience Engineering, Ghent University, Ghent, Belgium
| | - Sandrine Chalancon
- Université Clermont Auvergne, UMR UCA-INRA 454 MEDIS, Microbiology Digestive Environment and Health, Clermont-Ferrand, France
| | - Sylvain Denis
- Université Clermont Auvergne, UMR UCA-INRA 454 MEDIS, Microbiology Digestive Environment and Health, Clermont-Ferrand, France
| | | | | | - Nathalie Ballet
- Lesaffre International, Lesaffre Group, Marcq-en-Baroeul, France
| | - Stéphanie Blanquet-Diot
- CONTACT Stéphanie Blanquet-Diot Université Clermont Auvergne, UMR UCA-INRA 454 MEDIS, Microbiology Digestive Environment and Health, Clermont-Ferrand, France
| | - Tom Van de Wiele
- CMET, Center for Microbial Ecology and Technology, Department of Biotechnology, Faculty of Bioscience Engineering, Ghent University, Ghent, Belgium
| |
Collapse
|
47
|
Fernández-Ciganda S, Fraga M, Zunino P. Probiotic Lactobacilli Administration Induces Changes in the Fecal Microbiota of Preweaned Dairy Calves. Probiotics Antimicrob Proteins 2021; 14:804-815. [PMID: 34390476 DOI: 10.1007/s12602-021-09834-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/04/2021] [Indexed: 01/06/2023]
Abstract
Early microbial colonization is a determinant factor in animal health, and probiotic administration has been demonstrated to modulate intestinal microbiota and promote health in dairy calves. The objective of this study was to evaluate changes in calves' fecal microbiota after the administration of two probiotic lactobacilli strains that had previously exhibited beneficial effects in calves' health in relation to neonatal calf diarrhea. An in vivo assay was performed with 30 newborn male Holstein calves that were divided into three groups. Two groups were orally administered with two different lactobacilli strains (Lactobacillus johnsonii TP1.6 or Limosilactobacillus reuteri TP1.3B), and the third was the control group. Calves (5 to 9 days old) were administered with freeze-dried bacteria once a day for 10 consecutive days. Feces samples were taken before the first administration (day 0) and then again on days 10 and 21, and the V4 region of the bacterial 16S ribosomal gene was sequenced with an Illumina MiSeq 250 paired-end platform. The administration of both strains significantly affected the total bacterial community composition, and the effect lasted for 11 days after the last dose. In particular, amplicon sequence variants related to Bifidobacterium and Akkermansia genera were significantly higher in both treated groups. Therefore, modulation of the intestinal microbiota is a potential mechanism of action behind the beneficial effects of these probiotic strains.
Collapse
Affiliation(s)
- Sofía Fernández-Ciganda
- Department of Microbiology, Instituto de Investigaciones Biológicas Clemente Estable, Montevideo, Uruguay. .,Unidad de Investigación de Salud Animal, Instituto Nacional de Investigación Agropecuaria (INIA-LE), Colonia, Uruguay.
| | - Martín Fraga
- Unidad de Investigación de Salud Animal, Instituto Nacional de Investigación Agropecuaria (INIA-LE), Colonia, Uruguay
| | - Pablo Zunino
- Department of Microbiology, Instituto de Investigaciones Biológicas Clemente Estable, Montevideo, Uruguay
| |
Collapse
|
48
|
Moreira-Rosário A, Marques C, Pinheiro H, Araújo JR, Ribeiro P, Rocha R, Mota I, Pestana D, Ribeiro R, Pereira A, de Sousa MJ, Pereira-Leal J, de Sousa J, Morais J, Teixeira D, Rocha JC, Silvestre M, Príncipe N, Gatta N, Amado J, Santos L, Maltez F, Boquinhas A, de Sousa G, Germano N, Sarmento G, Granja C, Póvoa P, Faria A, Calhau C. Gut Microbiota Diversity and C-Reactive Protein Are Predictors of Disease Severity in COVID-19 Patients. Front Microbiol 2021; 12:705020. [PMID: 34349747 PMCID: PMC8326578 DOI: 10.3389/fmicb.2021.705020] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Accepted: 06/10/2021] [Indexed: 12/15/2022] Open
Abstract
The risk factors for coronavirus disease 2019 (COVID-19) severity are still poorly understood. Considering the pivotal role of the gut microbiota on host immune and inflammatory functions, we investigated the association between changes in the gut microbiota composition and the clinical severity of COVID-19. We conducted a multicenter cross-sectional study prospectively enrolling 115 COVID-19 patients categorized according to: (1) the WHO Clinical Progression Scale-mild, 19 (16.5%); moderate, 37 (32.2%); or severe, 59 (51.3%), and (2) the location of recovery from COVID-19-ambulatory, 14 (household isolation, 12.2%); hospitalized in ward, 40 (34.8%); or hospitalized in the intensive care unit, 61 (53.0%). Gut microbiota analysis was performed through 16S rRNA gene sequencing, and the data obtained were further related to the clinical parameters of COVID-19 patients. The risk factors for COVID-19 severity were identified by univariate and multivariable logistic regression models. In comparison to mild COVID-19 patients, the gut microbiota of moderate and severe patients have: (a) lower Firmicutes/Bacteroidetes ratio; (b) higher abundance of Proteobacteria; and (c) lower abundance of beneficial butyrate-producing bacteria such as the genera Roseburia and Lachnospira. Multivariable regression analysis showed that the Shannon diversity index [odds ratio (OR) = 2.85, 95% CI = 1.09-7.41, p = 0.032) and C-reactive protein (OR = 3.45, 95% CI = 1.33-8.91, p = 0.011) are risk factors for severe COVID-19 (a score of 6 or higher in the WHO Clinical Progression Scale). In conclusion, our results demonstrated that hospitalized patients with moderate and severe COVID-19 have microbial signatures of gut dysbiosis; for the first time, the gut microbiota diversity is pointed out as a prognostic biomarker of COVID-19 severity.
Collapse
Affiliation(s)
- André Moreira-Rosário
- Faculdade de Ciências M dicas, NOVA Medical School, Universidade NOVA de Lisboa, Lisbon, Portugal.,Faculdade de Ciências Médicas, CINTESIS - Center for Health Technology and Services Research, NOVA Medical School, Universidade NOVA de Lisboa, Lisboa, Portugal
| | - Cláudia Marques
- Faculdade de Ciências M dicas, NOVA Medical School, Universidade NOVA de Lisboa, Lisbon, Portugal.,Faculdade de Ciências Médicas, CINTESIS - Center for Health Technology and Services Research, NOVA Medical School, Universidade NOVA de Lisboa, Lisboa, Portugal
| | - Hélder Pinheiro
- Faculdade de Ciências M dicas, NOVA Medical School, Universidade NOVA de Lisboa, Lisbon, Portugal.,Department of Infectious Diseases, Hospital Curry Cabral, Centro Hospitalar Universitário de Lisboa Central, Lisboa, Portugal
| | - João Ricardo Araújo
- Faculdade de Ciências M dicas, NOVA Medical School, Universidade NOVA de Lisboa, Lisbon, Portugal.,Faculdade de Ciências Médicas, CINTESIS - Center for Health Technology and Services Research, NOVA Medical School, Universidade NOVA de Lisboa, Lisboa, Portugal
| | - Pedro Ribeiro
- Centro de Medicina Laboratorial Germano de Sousa, Lisboa, Portugal
| | - Rita Rocha
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,Instituto de Biologia Molecular e Celular (IBMC), Universidade do Porto, Porto, Portugal
| | - Inês Mota
- Faculdade de Ciências M dicas, NOVA Medical School, Universidade NOVA de Lisboa, Lisbon, Portugal.,Faculdade de Ciências Médicas, CINTESIS - Center for Health Technology and Services Research, NOVA Medical School, Universidade NOVA de Lisboa, Lisboa, Portugal
| | - Diogo Pestana
- Faculdade de Ciências M dicas, NOVA Medical School, Universidade NOVA de Lisboa, Lisbon, Portugal.,Faculdade de Ciências Médicas, CINTESIS - Center for Health Technology and Services Research, NOVA Medical School, Universidade NOVA de Lisboa, Lisboa, Portugal
| | - Rita Ribeiro
- Centro de Medicina Laboratorial Germano de Sousa, Lisboa, Portugal
| | - Ana Pereira
- Centro de Medicina Laboratorial Germano de Sousa, Lisboa, Portugal
| | - Maria José de Sousa
- Faculdade de Ciências M dicas, NOVA Medical School, Universidade NOVA de Lisboa, Lisbon, Portugal.,Centro de Medicina Laboratorial Germano de Sousa, Lisboa, Portugal
| | | | - José de Sousa
- Centro de Medicina Laboratorial Germano de Sousa, Lisboa, Portugal
| | - Juliana Morais
- Faculdade de Ciências M dicas, NOVA Medical School, Universidade NOVA de Lisboa, Lisbon, Portugal.,Faculdade de Ciências Médicas, CINTESIS - Center for Health Technology and Services Research, NOVA Medical School, Universidade NOVA de Lisboa, Lisboa, Portugal.,Faculdade de Ciências Médicas, Comprehensive Health Research Centre (CHRC), NOVA Medical School, Universidade NOVA de Lisboa, Lisboa, Portugal
| | - Diana Teixeira
- Faculdade de Ciências M dicas, NOVA Medical School, Universidade NOVA de Lisboa, Lisbon, Portugal.,Faculdade de Ciências Médicas, Comprehensive Health Research Centre (CHRC), NOVA Medical School, Universidade NOVA de Lisboa, Lisboa, Portugal
| | - Júlio César Rocha
- Faculdade de Ciências M dicas, NOVA Medical School, Universidade NOVA de Lisboa, Lisbon, Portugal.,Faculdade de Ciências Médicas, CINTESIS - Center for Health Technology and Services Research, NOVA Medical School, Universidade NOVA de Lisboa, Lisboa, Portugal
| | - Marta Silvestre
- Faculdade de Ciências M dicas, NOVA Medical School, Universidade NOVA de Lisboa, Lisbon, Portugal.,Faculdade de Ciências Médicas, CINTESIS - Center for Health Technology and Services Research, NOVA Medical School, Universidade NOVA de Lisboa, Lisboa, Portugal
| | - Nuno Príncipe
- Department of Emergency and Intensive Care Medicine, Centro Hospitalar Universitário de São João, Porto, Portugal
| | - Nuno Gatta
- Department of Emergency and Intensive Care Medicine, Centro Hospitalar Universitário de São João, Porto, Portugal
| | - José Amado
- Department of Emergency and Intensive Care Medicine, Centro Hospitalar Universitário de São João, Porto, Portugal
| | - Lurdes Santos
- Infectious Diseases Service, ID Intensive Care Unit, Faculdade de Medicina, Centro Hospitalar Universitário de São João, Universidade do Porto, Porto, Portugal
| | - Fernando Maltez
- Department of Infectious Diseases, Hospital Curry Cabral, Centro Hospitalar Universitário de Lisboa Central, Lisboa, Portugal
| | - Ana Boquinhas
- Department of Emergency, CUF Infante Santo Hospital, Lisboa, Portugal
| | - Germano de Sousa
- Centro de Medicina Laboratorial Germano de Sousa, Lisboa, Portugal
| | - Nuno Germano
- Polyvalent Intensive Care Unit, Hospital Curry Cabral, Centro Hospitalar Universitário de Lisboa Central, Lisboa, Portugal
| | - Gonçalo Sarmento
- Department of Internal Medicine, Centro Hospitalar de Entre o Douro e Vouga, Santa Maria da Feira, Portugal
| | - Cristina Granja
- Faculdade de Ciências Médicas, CINTESIS - Center for Health Technology and Services Research, NOVA Medical School, Universidade NOVA de Lisboa, Lisboa, Portugal.,Department of Anesthesiology, Centro Hospitalar Universitário de São João, Porto, Portugal.,Department of Surgery and Physiology, Faculdade de Medicina, Universidade do Porto, Porto, Portugal
| | - Pedro Póvoa
- Faculdade de Ciências M dicas, NOVA Medical School, Universidade NOVA de Lisboa, Lisbon, Portugal.,Polyvalent Intensive Care Unit, Hospital São Francisco Xavier, Centro Hospitalar Lisboa Ocidental, Lisboa, Portugal.,Center for Clinical Epidemiology, Research Unit of Clinical Epidemiology, OUH Odense University Hospital, Odense, Denmark
| | - Ana Faria
- Faculdade de Ciências M dicas, NOVA Medical School, Universidade NOVA de Lisboa, Lisbon, Portugal.,Faculdade de Ciências Médicas, Comprehensive Health Research Centre (CHRC), NOVA Medical School, Universidade NOVA de Lisboa, Lisboa, Portugal
| | - Conceição Calhau
- Faculdade de Ciências M dicas, NOVA Medical School, Universidade NOVA de Lisboa, Lisbon, Portugal.,Faculdade de Ciências Médicas, CINTESIS - Center for Health Technology and Services Research, NOVA Medical School, Universidade NOVA de Lisboa, Lisboa, Portugal
| |
Collapse
|
49
|
Milanović V, Cardinali F, Belleggia L, Garofalo C, Pasquini M, Tavoletti S, Riolo P, Ruschioni S, Isidoro N, Osimani A, Aquilanti L. Exploitation of Tenebrio molitor larvae as biological factories for human probiotics, an exploratory study. J Funct Foods 2021. [DOI: 10.1016/j.jff.2021.104490] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
|
50
|
Venardou B, O'Doherty JV, McDonnell MJ, Mukhopadhya A, Kiely C, Ryan MT, Sweeney T. Evaluation of the in vitro effects of the increasing inclusion levels of yeast β-glucan, a casein hydrolysate and its 5 kDa retentate on selected bacterial populations and strains commonly found in the gastrointestinal tract of pigs. Food Funct 2021; 12:2189-2200. [PMID: 33589892 DOI: 10.1039/d0fo02269a] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Previously, the 5 kDa retentate (5kDaR) of a casein hydrolysate (CH) and yeast β-glucan (YBG) were identified as promising anti-inflammatory dietary supplements for supporting intestinal health in pigs post-weaning. However, their direct effects on intestinal bacterial populations are less well-known. The main objectives of this study were to determine if the increasing concentrations of the CH, 5kDaR and YBG individually, can: (1) alter the bacterial and short-chain fatty acid profiles in a weaned pig faecal batch fermentation assay, and (2) directly influence the growth of selected beneficial (Lactobacillus plantarum, L. reuteri, Bifidobacterium thermophilum) and pathogenic (Enterotoxigenic Escherichia coli, Salmonella Typhimurium) bacterial strains in individual pure culture growth assays. The potential of CH as a comparable 5kDaR substitute was also evaluated. The 5kDaR increased lactobacilli counts and butyrate concentration in the batch fermentation assay (P < 0.05) and increased L. plantarum (linear, P < 0.05), L. reuteri (quadratic, P < 0.05) and B. thermophilum (linear, P < 0.05) counts and reduced S. typhimurium (quadratic, P = 0.058) counts in the pure culture growth assays. CH increased butyrate concentration (P < 0.05) in the batch fermentation assay. YBG reduced Prevotella spp. counts (P < 0.05) and butyrate concentration (P < 0.05) in the batch fermentation assay. Both CH and YBG had no major effects in the pure culture growth assays. In conclusion, the 5kDaR had the most beneficial effects associated with increased counts of Lactobacillus and Bifidobacterium genera and butyrate production and reduced S. typhimurium counts in vitro indicating its potential to promote gastrointestinal health.
Collapse
Affiliation(s)
- Brigkita Venardou
- School of Veterinary Medicine, University College Dublin, Belfield, Dublin 4, Ireland.
| | - John V O'Doherty
- School of Agriculture and Food Science, University College Dublin, Belfield, Dublin 4, Ireland
| | - Mary J McDonnell
- School of Agriculture and Food Science, University College Dublin, Belfield, Dublin 4, Ireland
| | - Anindya Mukhopadhya
- School of Veterinary Medicine, University College Dublin, Belfield, Dublin 4, Ireland. and Food for Health Ireland, University College Dublin, Belfield, Dublin 4, Ireland
| | - Claire Kiely
- School of Agriculture and Food Science, University College Dublin, Belfield, Dublin 4, Ireland
| | - Marion T Ryan
- School of Veterinary Medicine, University College Dublin, Belfield, Dublin 4, Ireland.
| | - Torres Sweeney
- School of Veterinary Medicine, University College Dublin, Belfield, Dublin 4, Ireland. and Food for Health Ireland, University College Dublin, Belfield, Dublin 4, Ireland
| |
Collapse
|