1
|
Waliaula PK, Kiarie EG, Diarra MS. Predisposition factors and control strategies of avian pathogenic Escherichia coli in laying hens. Front Vet Sci 2024; 11:1474549. [PMID: 39559543 PMCID: PMC11571327 DOI: 10.3389/fvets.2024.1474549] [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: 08/16/2024] [Accepted: 09/23/2024] [Indexed: 11/20/2024] Open
Abstract
Shift in laying hens housing from conventional cage-based systems to alternatives has impacted their health and performance. Microorganisms colonize young chick in the early stages of their physiological and immune development. These colonizing microbes originate from parent and the environment. Escherichia coli is among the normal gut colonizing bacteria however, some E. coli strains known as avian pathogenic E. coli (APEC), cause local or systemic infections (colibacillosis) responsible of significant economic losses to the poultry industry. Potential APEC strains and other poultry gut microbiota are influenced by several factors such as housing system, and the use of feed additives (prebiotics, probiotics, symbiotic, among others). This review will discuss the status of pullets and layers immunity, gut health, and predisposing factors of colibacillosis. Dietary interventions and some colibacillosis mitigation strategies in pullets and laying hens are reviewed and discussed. With the development of sequencing technologies and the use of feed additives as alternatives to antibiotics, future studies need to understand some of the complex associations between the feed additives, the rearing environment, and their selective pressure on gut microbiota, including E. coli, and their impacts on immune development in pullets and hens.
Collapse
Affiliation(s)
- Paul K. Waliaula
- Department of Animal Biosciences, University of Guelph, Guelph, ON, Canada
- Guelph Research and Development Center, Agriculture and Agri-Food Canada, Guelph, ON, Canada
| | - Elijah G. Kiarie
- Department of Animal Biosciences, University of Guelph, Guelph, ON, Canada
| | - Moussa S. Diarra
- Guelph Research and Development Center, Agriculture and Agri-Food Canada, Guelph, ON, Canada
| |
Collapse
|
2
|
Zhang Z, De X, Sun W, Liu R, Li Y, Yang Z, Liu N, Wu J, Miao Y, Wang J, Wang F, Ge J. Biogenic Selenium Nanoparticles Synthesized by L. brevis 23017 Enhance Aluminum Adjuvanticity and Make Up for its Disadvantage in Mice. Biol Trace Elem Res 2024; 202:4640-4653. [PMID: 38273184 DOI: 10.1007/s12011-023-04042-y] [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: 11/18/2023] [Accepted: 12/24/2023] [Indexed: 01/27/2024]
Abstract
The most popular vaccine adjuvants are aluminum ones, which have significantly reduced the incidence and mortality of many diseases. However, aluminum-adjuvanted vaccines are constrained by their limited capacity to elicit cellular and mucosal immune responses, thus constraining their broader utilization. Biogenic selenium nanoparticles are a low-cost, environmentally friendly, low-toxicity, and highly bioactive form of selenium supplementation. Here, we purified selenium nanoparticles synthesized by Levilactobacillus brevis 23017 (L-SeNP) and characterized them using Fourier-transform infrared spectroscopy, energy-dispersive X-ray spectroscopy, scanning electron microscopy, and transmission electron microscopy. The results indicate that the L-SeNP has a particle size ranging from 30 to 200 nm and is coated with proteins and polysaccharides. Subsequently, we assessed the immune-enhancing properties of L-SeNP in combination with an adjuvant-inactivated Clostridium perfringens type A vaccine using a mouse model. The findings demonstrate that L-SeNP can elevate the IgG and SIgA titers in immunized mice and modulate the Th1/Th2 immune response, thereby enhancing the protective effect of aluminum-adjuvanted vaccines. Furthermore, we observed that L-SeNP increases selenoprotein expression and regulates oxidative stress in immunized mice, which may be how L-SeNP regulates immunity. In conclusion, L-SeNP has the potential to augment the immune response of aluminum adjuvant vaccines and compensate for their limitations in eliciting Th1 and mucosal immune responses.
Collapse
Affiliation(s)
- Zheng Zhang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Xinqi De
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Weijiao Sun
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Runhang Liu
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, 150069, China
| | - Yifan Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Zaixing Yang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Ning Liu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Jingyi Wu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Yaxin Miao
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Jiaqi Wang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Fang Wang
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, 150069, China.
| | - Junwei Ge
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China.
- Heilongjiang Provincial Key Laboratory of Zoonosis, Harbin, 150030, China.
| |
Collapse
|
3
|
Huang Z, Huang X, Huang Y, Liang K, Chen L, Zhong C, Chen Y, Chen C, Wang Z, He F, Qin M, Long C, Tang B, Huang Y, Wu Y, Mo X, Weizhong T, Liu J. Identification of KRAS mutation-associated gut microbiota in colorectal cancer and construction of predictive machine learning model. Microbiol Spectr 2024; 12:e0272023. [PMID: 38572984 PMCID: PMC11064510 DOI: 10.1128/spectrum.02720-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 02/27/2024] [Indexed: 04/05/2024] Open
Abstract
Gut microbiota has demonstrated an increasingly important role in the onset and development of colorectal cancer (CRC). Nonetheless, the association between gut microbiota and KRAS mutation in CRC remains enigmatic. We conducted 16S rRNA sequencing on stool samples from 94 CRC patients and employed the linear discriminant analysis effect size algorithm to identify distinct gut microbiota between KRAS mutant and KRAS wild-type CRC patients. Transcriptome sequencing data from nine CRC patients were transformed into a matrix of immune infiltrating cells, which was then utilized to explore KRAS mutation-associated biological functions, including Gene Ontology items and Kyoto Encyclopedia of Genes and Genomes pathways. Subsequently, we analyzed the correlations among these KRAS mutation-associated gut microbiota, host immunity, and KRAS mutation-associated biological functions. At last, we developed a predictive random forest (RF) machine learning model to predict the KRAS mutation status in CRC patients, based on the gut microbiota associated with KRAS mutation. We identified a total of 26 differential gut microbiota between both groups. Intriguingly, a significant positive correlation was observed between Bifidobacterium spp. and mast cells, as well as between Bifidobacterium longum and chemokine receptor CX3CR1. Additionally, we also observed a notable negative correlation between Bifidobacterium and GOMF:proteasome binding. The RF model constructed using the KRAS mutation-associated gut microbiota demonstrated qualified efficacy in predicting the KRAS phenotype in CRC. Our study ascertained the presence of 26 KRAS mutation-associated gut microbiota in CRC and speculated that Bifidobacterium may exert an essential role in preventing CRC progression, which appeared to correlate with the upregulation of mast cells and CX3CR1 expression, as well as the downregulation of GOMF:proteasome binding. Furthermore, the RF model constructed on the basis of KRAS mutation-associated gut microbiota exhibited substantial potential in predicting KRAS mutation status in CRC patients.IMPORTANCEGut microbiota has emerged as an essential player in the onset and development of colorectal cancer (CRC). However, the relationship between gut microbiota and KRAS mutation in CRC remains elusive. Our study not only identified a total of 26 gut microbiota associated with KRAS mutation in CRC but also unveiled their significant correlations with tumor-infiltrating immune cells, immune-related genes, and biological pathways (Gene Ontology items and Kyoto Encyclopedia of Genes and Genomes pathways). We speculated that Bifidobacterium may play a crucial role in impeding CRC progression, potentially linked to the upregulation of mast cells and CX3CR1 expression, as well as the downregulation of GOMF:Proteasome binding. Furthermore, based on the KRAS mutation-associated gut microbiota, the RF model exhibited promising potential in the prediction of KRAS mutation status for CRC patients. Overall, the findings of our study offered fresh insights into microbiological research and clinical prediction of KRAS mutation status for CRC patients.
Collapse
Affiliation(s)
- Zigui Huang
- Division of Colorectal & Anal Surgery, Department of Gastrointestinal Surgery, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Xiaoliang Huang
- Division of Colorectal & Anal Surgery, Department of Gastrointestinal Surgery, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Yili Huang
- College of Oncology, Guangxi Medical University, Nanning, China
| | - Kunmei Liang
- College of Oncology, Guangxi Medical University, Nanning, China
| | - Lei Chen
- College of Oncology, Guangxi Medical University, Nanning, China
| | - Chuzhuo Zhong
- College of Oncology, Guangxi Medical University, Nanning, China
| | - Yingxin Chen
- College of Oncology, Guangxi Medical University, Nanning, China
| | - Chuanbin Chen
- Division of Colorectal & Anal Surgery, Department of Gastrointestinal Surgery, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Zhen Wang
- Division of Colorectal & Anal Surgery, Department of Gastrointestinal Surgery, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Fuhai He
- Division of Colorectal & Anal Surgery, Department of Gastrointestinal Surgery, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Mingjian Qin
- Division of Colorectal & Anal Surgery, Department of Gastrointestinal Surgery, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Chenyan Long
- Division of Colorectal & Anal Surgery, Department of Gastrointestinal Surgery, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Binzhe Tang
- Division of Colorectal & Anal Surgery, Department of Gastrointestinal Surgery, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Yongqi Huang
- Division of Colorectal & Anal Surgery, Department of Gastrointestinal Surgery, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Yongzhi Wu
- Division of Colorectal & Anal Surgery, Department of Gastrointestinal Surgery, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Xianwei Mo
- Division of Colorectal & Anal Surgery, Department of Gastrointestinal Surgery, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Tang Weizhong
- Division of Colorectal & Anal Surgery, Department of Gastrointestinal Surgery, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Jungang Liu
- Division of Colorectal & Anal Surgery, Department of Gastrointestinal Surgery, Guangxi Medical University Cancer Hospital, Nanning, China
| |
Collapse
|
4
|
Hou JJ, Ding L, Yang T, Yang YF, Jin YP, Zhang XP, Ma AH, Qin YH. The proteolytic activity in inflammatory bowel disease: insight from gut microbiota. Microb Pathog 2024; 188:106560. [PMID: 38272327 DOI: 10.1016/j.micpath.2024.106560] [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: 08/02/2023] [Revised: 01/20/2024] [Accepted: 01/22/2024] [Indexed: 01/27/2024]
Abstract
Inflammatory bowel disease (IBD) is a chronic, recurrent inflammatory disease caused by the destruction of the intestinal mucosal epithelium that affects a growing number of people worldwide. Although the etiology of IBD is complex and still elucidated, the role of dysbiosis and dysregulated proteolysis is well recognized. Various studies observed altered composition and diversity of gut microbiota, as well as increased proteolytic activity (PA) in serum, plasma, colonic mucosa, and fecal supernatant of IBD compared to healthy individuals. The imbalance of intestinal microecology and intestinal protein hydrolysis were gradually considered to be closely related to IBD. Notably, the pivotal role of intestinal microbiota in maintaining proteolytic balance received increasing attention. In summary, we have speculated a mesmerizing story, regarding the hidden role of PA and microbiota-derived PA hidden in IBD. Most importantly, we provided the diagnosis and therapeutic targets for IBD as well as the formulation of new treatment strategies for other digestive diseases and protease-related diseases.
Collapse
Affiliation(s)
- Jun-Jie Hou
- Department of Gastroenterology, Shaoxing People's Hospital, Shaoxing, PR China
| | - Liang Ding
- Department of Gastroenterology, Shaoxing People's Hospital, Shaoxing, PR China
| | - Tao Yang
- Department of Gastroenterology, Shaoxing People's Hospital, Shaoxing, PR China
| | - Yan-Fei Yang
- Department of Gastroenterology, Shaoxing People's Hospital, Shaoxing, PR China
| | - Yue-Ping Jin
- Department of Gastroenterology, Shaoxing People's Hospital, Shaoxing, PR China
| | - Xiao-Ping Zhang
- Department of Gastroenterology, Shaoxing People's Hospital, Shaoxing, PR China
| | - A-Huo Ma
- Department of Gastroenterology, Shaoxing People's Hospital, Shaoxing, PR China
| | - Yue-Hua Qin
- Department of Gastroenterology, Shaoxing People's Hospital, Shaoxing, PR China.
| |
Collapse
|
5
|
Lee CG, Cha KH, Kim GC, Im SH, Kwon HK. Exploring probiotic effector molecules and their mode of action in gut-immune interactions. FEMS Microbiol Rev 2023; 47:fuad046. [PMID: 37541953 DOI: 10.1093/femsre/fuad046] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 07/17/2023] [Accepted: 08/03/2023] [Indexed: 08/06/2023] Open
Abstract
Probiotics, live microorganisms that confer health benefits when consumed in adequate amounts, have gained significant attention for their potential therapeutic applications. The beneficial effects of probiotics are believed to stem from their ability to enhance intestinal barrier function, inhibit pathogens, increase beneficial gut microbes, and modulate immune responses. However, clinical studies investigating the effectiveness of probiotics have yielded conflicting results, potentially due to the wide variety of probiotic species and strains used, the challenges in controlling the desired number of live microorganisms, and the complex interactions between bioactive substances within probiotics. Bacterial cell wall components, known as effector molecules, play a crucial role in mediating the interaction between probiotics and host receptors, leading to the activation of signaling pathways that contribute to the health-promoting effects. Previous reviews have extensively covered different probiotic effector molecules, highlighting their impact on immune homeostasis. Understanding how each probiotic component modulates immune activity at the molecular level may enable the prediction of immunological outcomes in future clinical studies. In this review, we present a comprehensive overview of the structural and immunological features of probiotic effector molecules, focusing primarily on Lactobacillus and Bifidobacterium. We also discuss current gaps and limitations in the field and propose directions for future research to enhance our understanding of probiotic-mediated immunomodulation.
Collapse
Affiliation(s)
- Choong-Gu Lee
- Natural Product Informatics Research Center, Korea Institute of Science and Technology, 679, Saimdang-ro, Gangneung 25451, Korea
- Division of Bio-Medical Science and Technology, KIST School, University of Science and Technology, 679, Saimdang-ro, Seoul 02792, Korea
- Department of Convergence Medicine, Wonju College of Medicine, Yonsei University, 20, Ilsan-ro, Wonju 26493, Korea
| | - Kwang Hyun Cha
- Natural Product Informatics Research Center, Korea Institute of Science and Technology, 679, Saimdang-ro, Gangneung 25451, Korea
- Division of Bio-Medical Science and Technology, KIST School, University of Science and Technology, 679, Saimdang-ro, Seoul 02792, Korea
- Department of Convergence Medicine, Wonju College of Medicine, Yonsei University, 20, Ilsan-ro, Wonju 26493, Korea
| | - Gi-Cheon Kim
- Department of Microbiology and Immunology, Institute for Immunology and Immunological Diseases, and Brain Korea 21 PLUS Project for Medical Sciences, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seoul 03722, Korea
| | - Sin-Hyeog Im
- Department of Life Sciences, Pohang University of Science and Technology, 77, Cheongam-ro, Pohang 37673, Korea
- Institute for Convergence Research and Education, Yonsei University, 50-1 Yonsei-ro, Seoul 03722, Korea
- ImmunoBiome Inc, Bio Open Innovation Center, 77, Cheongam-ro, Pohang 37673 , Korea
| | - Ho-Keun Kwon
- Department of Microbiology and Immunology, Institute for Immunology and Immunological Diseases, and Brain Korea 21 PLUS Project for Medical Sciences, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seoul 03722, Korea
| |
Collapse
|
6
|
Xu J, Ye W, Yang TT, Yan T, Cai H, Zhou A, Yang Y. DNA accelerates the protease inhibition of a bacterial serpin chloropin. Front Mol Biosci 2023; 10:1157186. [PMID: 37065444 PMCID: PMC10090351 DOI: 10.3389/fmolb.2023.1157186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Accepted: 03/20/2023] [Indexed: 03/31/2023] Open
Abstract
Serine protease inhibitors (Serpins) are the most widely distributed protease inhibitors in nature and have been identified from all kingdoms of life. Eukaryotic serpins are most abundant with their activities often subject to modulation by cofactors; however, little is known about the regulation of prokaryotic serpins. To address this, here we prepared a recombinant bacteria serpin, termed chloropin, derived from green sulfur bacteria Chlorobium limicola and solved its crystal structure at 2.2 Å resolution. This showed a canonical inhibitory serpin conformation of native chloropin with a surface-exposed reactive loop and a large central beta-sheet. Enzyme activity analysis showed that chloropin could inhibit multiple proteases, such as thrombin and KLK7 with second order inhibition rate constants at 2.5×104 M−1s−1 and 4.5×104 M−1s−1 respectively, consistent with its P1 arginine residue. Heparin could accelerate the thrombin inhibition by ∼17-fold with a bell-shaped dose-dependent curve as seen with heparin-mediated thrombin inhibition by antithrombin. Interestingly, supercoiled DNA could accelerate the inhibition of thrombin by chloropin by 74-fold, while linear DNA accelerated the reaction by 142-fold through a heparin-like template mechanism. In contrast, DNA did not affect the inhibition of thrombin by antithrombin. These results indicate that DNA is likely a natural modulator of chloropin protecting the cell from endogenous or exogenous environmental proteases, and prokaryotic serpins have diverged during evolution to use different surface subsites for activity modulation.
Collapse
Affiliation(s)
- Jiawei Xu
- Department of Bioengineering, Zunyi Medical University Zhuhai Campus, Zhuhai, Guangdong, China
| | - Wei Ye
- Department of Preventive Dentistry, The Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ting Ting Yang
- Department of Preventive Dentistry, The Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Teng Yan
- Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Haiyan Cai
- Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- *Correspondence: Haiyan Cai, ; Aiwu Zhou, ; Yufeng Yang,
| | - Aiwu Zhou
- Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- *Correspondence: Haiyan Cai, ; Aiwu Zhou, ; Yufeng Yang,
| | - Yufeng Yang
- Department of Bioengineering, Zunyi Medical University Zhuhai Campus, Zhuhai, Guangdong, China
- *Correspondence: Haiyan Cai, ; Aiwu Zhou, ; Yufeng Yang,
| |
Collapse
|
7
|
Smith MM, Melrose J. Pentosan Polysulfate Affords Pleotropic Protection to Multiple Cells and Tissues. Pharmaceuticals (Basel) 2023; 16:437. [PMID: 36986536 PMCID: PMC10132487 DOI: 10.3390/ph16030437] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 02/18/2023] [Accepted: 03/01/2023] [Indexed: 03/16/2023] Open
Abstract
Pentosan polysulfate (PPS), a small semi-synthetic highly sulfated heparan sulfate (HS)-like molecule, shares many of the interactive properties of HS. The aim of this review was to outline the potential of PPS as an interventional therapeutic protective agent in physiological processes affecting pathological tissues. PPS is a multifunctional molecule with diverse therapeutic actions against many disease processes. PPS has been used for decades in the treatment of interstitial cystitis and painful bowel disease, it has tissue-protective properties as a protease inhibitor in cartilage, tendon and IVD, and it has been used as a cell-directive component in bioscaffolds in tissue engineering applications. PPS regulates complement activation, coagulation, fibrinolysis and thrombocytopenia, and it promotes the synthesis of hyaluronan. Nerve growth factor production in osteocytes is inhibited by PPS, reducing bone pain in osteoarthritis and rheumatoid arthritis (OA/RA). PPS also removes fatty compounds from lipid-engorged subchondral blood vessels in OA/RA cartilage, reducing joint pain. PPS regulates cytokine and inflammatory mediator production and is also an anti-tumor agent that promotes the proliferation and differentiation of mesenchymal stem cells and the development of progenitor cell lineages that have proven to be useful in strategies designed to effect repair of the degenerate intervertebral disc (IVD) and OA cartilage. PPS stimulates proteoglycan synthesis by chondrocytes in the presence or absence of interleukin (IL)-1, and stimulates hyaluronan production by synoviocytes. PPS is thus a multifunctional tissue-protective molecule of potential therapeutic application for a diverse range of disease processes.
Collapse
Affiliation(s)
- Margaret M. Smith
- Raymond Purves Laboratory, Institute of Bone and Joint Research, Kolling Institute of Medical Research, Faculty of Health and Science, University of Sydney at Royal North Shore Hospital, St. Leonards, NSW 2065, Australia;
| | - James Melrose
- Raymond Purves Laboratory, Institute of Bone and Joint Research, Kolling Institute of Medical Research, Faculty of Health and Science, University of Sydney at Royal North Shore Hospital, St. Leonards, NSW 2065, Australia;
- Graduate Schools of Biomedical Engineering, University of NSW, Sydney, NSW 2052, Australia
- Sydney Medical School, Northern Campus, Royal North Shore Hospital, St. Leonards, NSW 2065, Australia
| |
Collapse
|
8
|
Sadeghpour Heravi F, Hu H. Bifidobacterium: Host-Microbiome Interaction and Mechanism of Action in Preventing Common Gut-Microbiota-Associated Complications in Preterm Infants: A Narrative Review. Nutrients 2023; 15:709. [PMID: 36771414 PMCID: PMC9919561 DOI: 10.3390/nu15030709] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 01/23/2023] [Accepted: 01/27/2023] [Indexed: 01/31/2023] Open
Abstract
The development and health of infants are intertwined with the protective and regulatory functions of different microorganisms in the gut known as the gut microbiota. Preterm infants born with an imbalanced gut microbiota are at substantial risk of several diseases including inflammatory intestinal diseases, necrotizing enterocolitis, late-onset sepsis, neurodevelopmental disorders, and allergies which can potentially persist throughout adulthood. In this review, we have evaluated the role of Bifidobacterium as commonly used probiotics in the development of gut microbiota and prevention of common diseases in preterm infants which is not fully understood yet. The application of Bifidobacterium as a therapeutical approach in the re-programming of the gut microbiota in preterm infants, the mechanisms of host-microbiome interaction, and the mechanism of action of this bacterium have also been investigated, aiming to provide new insights and opportunities in microbiome-targeted interventions in personalized medicine.
Collapse
Affiliation(s)
| | - Honghua Hu
- Macquarie Medical School, Macquarie University, Sydney, NSW 2109, Australia
- Innovation Center of Translational Pharmacy, Jinhua Institute of Zhejiang University, Jinhua 321016, China
| |
Collapse
|
9
|
Mazziotta C, Tognon M, Martini F, Torreggiani E, Rotondo JC. Probiotics Mechanism of Action on Immune Cells and Beneficial Effects on Human Health. Cells 2023; 12:184. [PMID: 36611977 PMCID: PMC9818925 DOI: 10.3390/cells12010184] [Citation(s) in RCA: 266] [Impact Index Per Article: 133.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 12/12/2022] [Accepted: 12/29/2022] [Indexed: 01/03/2023] Open
Abstract
Immune cells and commensal microbes in the human intestine constantly communicate with and react to each other in a stable environment in order to maintain healthy immune activities. Immune system-microbiota cross-talk relies on a complex network of pathways that sustain the balance between immune tolerance and immunogenicity. Probiotic bacteria can interact and stimulate intestinal immune cells and commensal microflora to modulate specific immune functions and immune homeostasis. Growing evidence shows that probiotic bacteria present important health-promoting and immunomodulatory properties. Thus, the use of probiotics might represent a promising approach for improving immune system activities. So far, few studies have been reported on the beneficial immune modulatory effect of probiotics. However, many others, which are mainly focused on their metabolic/nutritional properties, have been published. Therefore, the mechanisms behind the interaction between host immune cells and probiotics have only been partially described. The present review aims to collect and summarize the most recent scientific results and the resulting implications of how probiotic bacteria and immune cells interact to improve immune functions. Hence, a description of the currently known immunomodulatory mechanisms of probiotic bacteria in improving the host immune system is provided.
Collapse
Affiliation(s)
- Chiara Mazziotta
- Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy
- Center for Studies on Gender Medicine, Department of Medical Sciences, University of Ferrara, 64/b, Fossato di Mortara Street, 44121 Ferrara, Italy
| | - Mauro Tognon
- Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy
| | - Fernanda Martini
- Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy
- Center for Studies on Gender Medicine, Department of Medical Sciences, University of Ferrara, 64/b, Fossato di Mortara Street, 44121 Ferrara, Italy
- Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, 44121 Ferrara, Italy
| | - Elena Torreggiani
- Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy
| | - John Charles Rotondo
- Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy
- Center for Studies on Gender Medicine, Department of Medical Sciences, University of Ferrara, 64/b, Fossato di Mortara Street, 44121 Ferrara, Italy
| |
Collapse
|
10
|
Mills S, Yang B, Smith GJ, Stanton C, Ross RP. Efficacy of Bifidobacterium longum alone or in multi-strain probiotic formulations during early life and beyond. Gut Microbes 2023; 15:2186098. [PMID: 36896934 PMCID: PMC10012958 DOI: 10.1080/19490976.2023.2186098] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Accepted: 02/24/2023] [Indexed: 03/11/2023] Open
Abstract
The significance of Bifidobacterium to human health can be appreciated from its early colonization of the neonatal gut, where Bifidobacterium longum represents the most abundant species. While its relative abundance declines with age, it is further reduced in several diseases. Research into the beneficial properties of B. longum has unveiled a range of mechanisms, including the production of bioactive molecules, such as short-chain fatty acids, polysaccharides, and serine protease inhibitors. From its intestinal niche, B. longum can have far-reaching effects in the body influencing immune responses in the lungs and even skin, as well as influencing brain activity. In this review, we present the biological and clinical impacts of this species on a range of human conditions beginning in neonatal life and beyond. The available scientific evidence reveals a strong rationale for continued research and further clinical trials that investigate the ability of B. longum to treat or prevent a range of diseases across the human lifespan.
Collapse
Affiliation(s)
- Susan Mills
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Bo Yang
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
| | | | - Catherine Stanton
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- Food Biosciences Department, Teagasc Food Research Centre, Co Cork, Ireland
| | - R. Paul Ross
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| |
Collapse
|
11
|
Adjuvant role of probiotics in allergen-specific immunotherapy. Clin Immunol 2022; 245:109164. [DOI: 10.1016/j.clim.2022.109164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 10/04/2022] [Accepted: 10/06/2022] [Indexed: 11/19/2022]
|
12
|
Torres-Maravilla E, Holowacz S, Delannoy J, Lenoir L, Jacouton E, Gervason S, Meynier M, Boucard AS, Carvalho FA, Barbut F, Bermúdez-Humarán LG, Langella P, Waligora-Dupriet AJ. Serpin-positive Bifidobacterium breve CNCM I-5644 improves intestinal permeability in two models of irritable bowel syndrome. Sci Rep 2022; 12:19776. [PMID: 36396717 PMCID: PMC9672316 DOI: 10.1038/s41598-022-21746-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 09/30/2022] [Indexed: 11/19/2022] Open
Abstract
Probiotic supplementation can help to mitigate the pathogenesis of irritable bowel syndrome (IBS) by reinforcing the intestinal barrier, and reducing both inflammation and proteolytic activity. Here, a combination of in vitro tests was performed on 33 Bifidobacterium strains as probiotic candidates for IBS. In addition to the classical tests performed, the detection of the serine protease inhibitor (serpin) enzyme capable of decreasing the high proteolytic activity found in IBS patients was included. Three serpin-positive strains were selected: Bifidobacterium breve CNCM I-5644, Bifidobacterium longum subsp. infantis CNCM I-5645 and B. longum CNCM I-5646 for their immunomodulation properties and protection of intestinal epithelial integrity in vitro. Furthermore, we found that B. breve CNCM I-5644 strain prevented intestinal hyperpermeability by upregulating Cingulin and Tight Junction Protein 1 mRNA levels and reducing pro-inflammatory markers. The ability of CNCM I-5644 strain to restore intestinal hyperpermeability (FITC-dextran) was shown in the murine model of low-grade inflammation induced by dinitrobenzene sulfonic acid (DNBS). This effect of this strain was corroborated in a second model of IBS, the neonatal maternal separation model in mice. Altogether, these data suggest that serpin-positive B. breve CNCM I-5644 may partially prevent disorders associated with increased barrier permeability such as IBS.
Collapse
Affiliation(s)
- Edgar Torres-Maravilla
- grid.460789.40000 0004 4910 6535INRAE, AgroParisTech, Micalis Institute, Université Paris-Saclay, 78350 Jouy-en-Josas, France ,grid.7429.80000000121866389Université Paris Cité, INSERM, 3PHM, F-75006 Paris, France
| | - Sophie Holowacz
- PiLeJe Laboratoire, 37 Quai de Grenelle, 75015 Paris Cedex 15, France
| | - Johanne Delannoy
- grid.7429.80000000121866389Université Paris Cité, INSERM, 3PHM, F-75006 Paris, France
| | - Loïc Lenoir
- PiLeJe Laboratoire, 37 Quai de Grenelle, 75015 Paris Cedex 15, France
| | - Elsa Jacouton
- PiLeJe Laboratoire, 37 Quai de Grenelle, 75015 Paris Cedex 15, France
| | - Sandie Gervason
- grid.494717.80000000115480420INSERM UMR 1107 NeuroDol, University of Clermont Auvergne, 63001 Clermont-Ferrand, France
| | - Maëva Meynier
- grid.494717.80000000115480420INSERM UMR 1107 NeuroDol, University of Clermont Auvergne, 63001 Clermont-Ferrand, France
| | - Anne-Sophie Boucard
- grid.460789.40000 0004 4910 6535INRAE, AgroParisTech, Micalis Institute, Université Paris-Saclay, 78350 Jouy-en-Josas, France
| | - Frédéric A. Carvalho
- grid.494717.80000000115480420INSERM UMR 1107 NeuroDol, University of Clermont Auvergne, 63001 Clermont-Ferrand, France
| | - Frédéric Barbut
- grid.7429.80000000121866389Université Paris Cité, INSERM, 3PHM, F-75006 Paris, France ,grid.50550.350000 0001 2175 4109National Reference Laboratory for C. Difficile, Hôpital Saint-Antoine, Assistance Publique-Hôpitaux de Paris, 75012 Paris, France
| | - Luis G. Bermúdez-Humarán
- grid.460789.40000 0004 4910 6535INRAE, AgroParisTech, Micalis Institute, Université Paris-Saclay, 78350 Jouy-en-Josas, France
| | - Philippe Langella
- grid.460789.40000 0004 4910 6535INRAE, AgroParisTech, Micalis Institute, Université Paris-Saclay, 78350 Jouy-en-Josas, France
| | | |
Collapse
|
13
|
Yadav R, Kumar A, Bano N, Singh P, Pandey A, Dhar YV, Bag SK, Pande V, Sharma P, Singh SP, Iqbal HM, Sanyal I. Co-expression of Cocculus hirsutus trypsin inhibitor with Cry protein reduces resistant development in targeted insects along with complete mortality. INDUSTRIAL CROPS AND PRODUCTS 2022; 188:115674. [DOI: 10.1016/j.indcrop.2022.115674] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
|
14
|
The double-edged sword of gut bacteria in celiac disease and implications for therapeutic potential. Mucosal Immunol 2022; 15:235-243. [PMID: 35031683 DOI: 10.1038/s41385-021-00479-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 11/22/2021] [Accepted: 12/18/2021] [Indexed: 02/04/2023]
Abstract
Celiac disease (CeD) is an immune-mediated disease, triggered by gluten ingestion, in genetically susceptible individuals. The gluten-free diet (GFD) is the only current treatment for CeD, but is difficult to follow, has high non-adherence rates, and does not always lead to symptomatic or mucosal remission. Microbially-mediated mechanisms have been proposed to contribute to disease pathogenesis, and clinical studies support an association, but mechanistic insight has been difficult to obtain. Recent advances using translational approaches have provided clues to the mechanisms through which bacteria could contribute to CeD pathogenesis. In this review we discuss these bacterially mediated mechanisms, which include the modulation of pathogenic or protective pathways. Targeting these pathways through microbial therapeutics could provide adjuvant therapies to the GFD.
Collapse
|
15
|
Probiotic Molecules That Inhibit Inflammatory Diseases. APPLIED SCIENCES-BASEL 2022. [DOI: 10.3390/app12031147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Consumption of probiotics for health purposes has increased vastly in the past few decades, and yet the scientific evidence to support health benefits from probiotics is only beginning to emerge. As more probiotics are studied, we are beginning to understand the mechanisms of action by which they benefit human health, as well as to identify the bacterial molecules responsible for these benefits. A new era of therapeutics is on the horizon in which purified molecules from probiotics will be used to prevent and treat diseases. In this review, we summarize the active molecules from probiotic bacteria that have been shown to affect innate and adaptive immunity and have health benefits in experimental settings. We focus particularly on the cellular and molecular mechanisms of the probiotic Bacillus subtilis and its active molecule, exopolysaccharide (ESPBs).
Collapse
|
16
|
Duboux S, Van Wijchen M, Kleerebezem M. The Possible Link Between Manufacturing and Probiotic Efficacy; a Molecular Point of View on Bifidobacterium. Front Microbiol 2022; 12:812536. [PMID: 35003044 PMCID: PMC8741271 DOI: 10.3389/fmicb.2021.812536] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 12/06/2021] [Indexed: 11/13/2022] Open
Abstract
Probiotics for food or supplement use have been studied in numerous clinical trials, addressing a broad variety of diseases, and conditions. However, discrepancies were observed in the clinical outcomes stemming from the use of lactobacillaceae and bifidobacteria strains. These differences are often attributed to variations in the clinical trial protocol like trial design, included target population, probiotic dosage, or outcome parameters measured. However, a contribution of the methods used to produce the live bioactive ingredients should not be neglected as a possible additional factor in the observed clinical outcome variations. It is well established that manufacturing conditions play a role in determining the survival and viability of probiotics, but much less is known about their influence on the probiotic molecular composition and functionality. In this review, we briefly summarize the evidence obtained for Lacticaseibacillus rhamnosus GG and Lactiplantibacillus plantarum WCFS1, highlighting that expression and presence of probiotic niche factor (NF) and/or effector molecules (EM) may be altered during production of those two well-characterized lactobacillaceae probiotic strains. Subsequently, we summarize in more depth what is the present state of knowledge about bifidobacterial probiotic NF and EM; how their expression may be modified by manufacturing related environmental factors and how that may affect their biological activity in the host. This review highlights the importance of gathering knowledge on probiotic NF and EM, to validate them as surrogate markers of probiotic functionality. We further propose that monitoring of validated NF and/or EM during production and/or in the final preparation could complement viable count assessments that are currently applied in industry. Overall, we suggest that implementation of molecular level quality controls (i.e., based on validated NF and EM), could provide mode of action based in vitro tests contributing to better control the health-promoting reliability of probiotic products.
Collapse
Affiliation(s)
- Stéphane Duboux
- Nestlé Research, Lausanne, Switzerland.,Host-Microbe Interactomics Group, Wageningen University and Research, Wageningen, Netherlands
| | - Myrthe Van Wijchen
- Nestlé Research, Lausanne, Switzerland.,Host-Microbe Interactomics Group, Wageningen University and Research, Wageningen, Netherlands
| | - Michiel Kleerebezem
- Host-Microbe Interactomics Group, Wageningen University and Research, Wageningen, Netherlands
| |
Collapse
|
17
|
Requena T, Pérez Martínez G. Probiotics, Prebiotics, Synbiotics, Postbiotics and Other Biotics. What's Next? COMPREHENSIVE GUT MICROBIOTA 2022:197-210. [DOI: 10.1016/b978-0-12-819265-8.00094-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
18
|
Jastrząb R, Graczyk D, Siedlecki P. Molecular and Cellular Mechanisms Influenced by Postbiotics. Int J Mol Sci 2021; 22:ijms222413475. [PMID: 34948270 PMCID: PMC8707144 DOI: 10.3390/ijms222413475] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 11/29/2021] [Accepted: 12/07/2021] [Indexed: 12/12/2022] Open
Abstract
In recent years, commensal bacteria colonizing the human body have been recognized as important determinants of health and multiple pathologic conditions. Among the most extensively studied commensal bacteria are the gut microbiota, which perform a plethora of functions, including the synthesis of bioactive products, metabolism of dietary compounds, and immunomodulation, both through attenuation and immunostimulation. An imbalance in the microbiota population, i.e., dysbiosis, has been linked to many human pathologies, including various cancer types and neurodegenerative diseases. Targeting gut microbiota and microbiome-host interactions resulting from probiotics, prebiotics, and postbiotics is a growing opportunity for the effective treatment of various diseases. As more research is being conducted, the microbiome field is shifting from simple descriptive analysis of commensal compositions to more molecular, cellular, and functional studies. Insight into these mechanisms is of paramount importance for understanding and modulating the effects that microbiota, probiotics, and their derivatives exert on host health.
Collapse
|
19
|
Kumar H, Schütz F, Bhardwaj K, Sharma R, Nepovimova E, Dhanjal DS, Verma R, Kumar D, Kuča K, Cruz-Martins N. Recent advances in the concept of paraprobiotics: Nutraceutical/functional properties for promoting children health. Crit Rev Food Sci Nutr 2021:1-16. [PMID: 34748444 DOI: 10.1080/10408398.2021.1996327] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Probiotics consumption has been associated with various health promoting benefits, including disease prevention and even treatment by modulating gut microbiota. Contrary to this, probiotics may also overstimulate the immune system, trigger systemic infections, harmful metabolic activities, and promote gene transfer. In children, the fragile immune system and impaired intestinal barrier may boost the occurrence of adverse effects following probiotics' consumption. To overcome these health challenges, the key focus has been shifted toward non-viable probiotics, also called paraprobiotics. Cell wall polysaccharides, peptidoglycans, surface proteins and teichoic acid present on cell's surface are involved in the interaction of paraprobiotics with the host, ultimately providing health benefits. Among other benefits, paraprobiotics possess the ability to regulate innate and adaptive immunity, exert anti-adhesion, anti-biofilm, anti-hypertensive, anti-inflammatory, antioxidant, anti-proliferative, and antagonistic effects against pathogens, while also enhance clinical impact and general safety when administered in children in comparison to probiotics. Clinical evidence have underlined the paraprobiotics impact in children and young infants against atopic dermatitis, respiratory and gastrointestinal infections, in addition to be useful for immunocompromised individuals. Therefore, this review focuses on probiotics-related issues in children's health and also discusses the Lactobacillus and Bifidobacterium spp. qualities for qualifying as paraprobiotics and their role in promoting the children's health.
Collapse
Affiliation(s)
- Harsh Kumar
- School of Bioengineering & Food Technology, Shoolini University of Biotechnology and Management Sciences, Solan, India
| | - Francine Schütz
- Department of Medicine/Biomedicine, Faculty of Medicine, University of Porto, Porto, Portugal
| | - Kanchan Bhardwaj
- School of Biological and Environmental Sciences, Shoolini University of Biotechnology and Management Sciences, Solan, India
| | - Ruchi Sharma
- School of Bioengineering & Food Technology, Shoolini University of Biotechnology and Management Sciences, Solan, India
| | - Eugenie Nepovimova
- Department of Chemistry, Faculty of Science, University of Hradec Kralove, Hradec Kralove, Czech Republic
| | - Daljeet Singh Dhanjal
- School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, Punjab, India
| | - Rachna Verma
- School of Biological and Environmental Sciences, Shoolini University of Biotechnology and Management Sciences, Solan, India
| | - Dinesh Kumar
- School of Bioengineering & Food Technology, Shoolini University of Biotechnology and Management Sciences, Solan, India
| | - Kamil Kuča
- Department of Chemistry, Faculty of Science, University of Hradec Kralove, Hradec Kralove, Czech Republic
| | - Natália Cruz-Martins
- Department of Medicine/Biomedicine, Faculty of Medicine, University of Porto, Porto, Portugal.,Institute for Research and Innovation in Health (i3S), University of Porto, Porto, Portugal.,Institute of Research and Advanced Training in Health Sciences and Technologies (CESPU), Rua Central de Gandra, Gandra, PRD, Portugal
| |
Collapse
|
20
|
Mkaouar H, Mariaule V, Rhimi S, Hernandez J, Kriaa A, Jablaoui A, Akermi N, Maguin E, Lesner A, Korkmaz B, Rhimi M. Gut Serpinome: Emerging Evidence in IBD. Int J Mol Sci 2021; 22:ijms22116088. [PMID: 34200095 PMCID: PMC8201313 DOI: 10.3390/ijms22116088] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 06/01/2021] [Accepted: 06/03/2021] [Indexed: 12/11/2022] Open
Abstract
Inflammatory bowel diseases (IBD) are incurable disorders whose prevalence and global socioeconomic impact are increasing. While the role of host genetics and immunity is well documented, that of gut microbiota dysbiosis is increasingly being studied. However, the molecular basis of the dialogue between the gut microbiota and the host remains poorly understood. Increased activity of serine proteases is demonstrated in IBD patients and may contribute to the onset and the maintenance of the disease. The intestinal proteolytic balance is the result of an equilibrium between the proteases and their corresponding inhibitors. Interestingly, the serine protease inhibitors (serpins) encoded by the host are well reported; in contrast, those from the gut microbiota remain poorly studied. In this review, we provide a concise analysis of the roles of serine protease in IBD physiopathology and we focus on the serpins from the gut microbiota (gut serpinome) and their relevance as a promising therapeutic approach.
Collapse
Affiliation(s)
- Héla Mkaouar
- Microbiota Interaction with Human and Animal Team (MIHA), Micalis Institute, AgroParisTech, Université Paris-Saclay, INRAE, 78350 Jouy-en-Josas, France; (H.M.); (V.M.); (S.R.); (A.K.); (A.J.); (N.A.); (E.M.)
| | - Vincent Mariaule
- Microbiota Interaction with Human and Animal Team (MIHA), Micalis Institute, AgroParisTech, Université Paris-Saclay, INRAE, 78350 Jouy-en-Josas, France; (H.M.); (V.M.); (S.R.); (A.K.); (A.J.); (N.A.); (E.M.)
| | - Soufien Rhimi
- Microbiota Interaction with Human and Animal Team (MIHA), Micalis Institute, AgroParisTech, Université Paris-Saclay, INRAE, 78350 Jouy-en-Josas, France; (H.M.); (V.M.); (S.R.); (A.K.); (A.J.); (N.A.); (E.M.)
| | - Juan Hernandez
- Department of Clinical Sciences, Nantes-Atlantic College of Veterinary Medicine and Food Sciences (Oniris), University of Nantes, 101 Route de Gachet, 44300 Nantes, France;
| | - Aicha Kriaa
- Microbiota Interaction with Human and Animal Team (MIHA), Micalis Institute, AgroParisTech, Université Paris-Saclay, INRAE, 78350 Jouy-en-Josas, France; (H.M.); (V.M.); (S.R.); (A.K.); (A.J.); (N.A.); (E.M.)
| | - Amin Jablaoui
- Microbiota Interaction with Human and Animal Team (MIHA), Micalis Institute, AgroParisTech, Université Paris-Saclay, INRAE, 78350 Jouy-en-Josas, France; (H.M.); (V.M.); (S.R.); (A.K.); (A.J.); (N.A.); (E.M.)
| | - Nizar Akermi
- Microbiota Interaction with Human and Animal Team (MIHA), Micalis Institute, AgroParisTech, Université Paris-Saclay, INRAE, 78350 Jouy-en-Josas, France; (H.M.); (V.M.); (S.R.); (A.K.); (A.J.); (N.A.); (E.M.)
| | - Emmanuelle Maguin
- Microbiota Interaction with Human and Animal Team (MIHA), Micalis Institute, AgroParisTech, Université Paris-Saclay, INRAE, 78350 Jouy-en-Josas, France; (H.M.); (V.M.); (S.R.); (A.K.); (A.J.); (N.A.); (E.M.)
| | - Adam Lesner
- Faculty of Chemistry, University of Gdansk, Uniwersytet Gdanski, Chemistry, Wita Stwosza 63, PL80-308 Gdansk, Poland;
| | - Brice Korkmaz
- INSERM UMR-1100, “Research Center for Respiratory Diseases” and University of Tours, 37032 Tours, France;
| | - Moez Rhimi
- Microbiota Interaction with Human and Animal Team (MIHA), Micalis Institute, AgroParisTech, Université Paris-Saclay, INRAE, 78350 Jouy-en-Josas, France; (H.M.); (V.M.); (S.R.); (A.K.); (A.J.); (N.A.); (E.M.)
- Correspondence:
| |
Collapse
|
21
|
CESA-LUNA CATHERINE, ALATORRE-CRUZ JULIAMARÍA, CARREÑO-LÓPEZ RICARDO, QUINTERO-HERNÁNDEZ VERÓNICA, BAEZ ANTONINO. Emerging Applications of Bacteriocins as Antimicrobials, Anticancer Drugs, and Modulators of The Gastrointestinal Microbiota. Pol J Microbiol 2021; 70:143-159. [PMID: 34349808 PMCID: PMC8326989 DOI: 10.33073/pjm-2021-020] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 04/06/2021] [Accepted: 04/25/2021] [Indexed: 02/06/2023] Open
Abstract
The use of bacteriocins holds great promise in different areas such as health, food, nutrition, veterinary, nanotechnology, among others. Many research groups worldwide continue to advance the knowledge to unravel a novel range of therapeutic agents and food preservatives. This review addresses the advances of bacteriocins and their producer organisms as biocontrol agents for applications in the medical industry and agriculture. Furthermore, the bacteriocin mechanism of action and structural characteristics will be reviewed. Finally, the potential role of bacteriocins to modulate the signaling in host-associated microbial communities will be discussed.
Collapse
Affiliation(s)
- CATHERINE CESA-LUNA
- Centro de Investigaciones en Ciencias Microbiológicas (CICM), Instituto de Ciencias (IC), Benemérita Universidad Autónoma de Puebla (BUAP), Puebla, México
| | | | - RICARDO CARREÑO-LÓPEZ
- Centro de Investigaciones en Ciencias Microbiológicas (CICM), Instituto de Ciencias (IC), Benemérita Universidad Autónoma de Puebla (BUAP), Puebla, México
| | | | - ANTONINO BAEZ
- Centro de Investigaciones en Ciencias Microbiológicas (CICM), Instituto de Ciencias (IC), Benemérita Universidad Autónoma de Puebla (BUAP), Puebla, México
| |
Collapse
|
22
|
Hou JJ, Wang X, Li Y, Su S, Wang YM, Wang BM. The relationship between gut microbiota and proteolytic activity in irritable bowel syndrome. Microb Pathog 2021; 157:104995. [PMID: 34048892 DOI: 10.1016/j.micpath.2021.104995] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 05/05/2021] [Accepted: 05/05/2021] [Indexed: 02/08/2023]
Abstract
Irritable bowel syndrome (IBS) is a common functional gastrointestinal disease that affects 3.8-9.2% of the world population. It affects the physiology and psychology of patients and increases the burden on families, the healthcare system, society, and economic development. Presently, a large number of studies have shown that compared to healthy individuals, the composition and diversity of gut microbiota in IBS patients have changed, and the proteolytic activity (PA) in fecal supernatant and colonic mucosa of IBS patients has also increased. These findings indicate that the imbalance of intestinal microecology and intestinal protein hydrolysis is closely related to IBS. Furthermore, the intestinal flora is a key substance that regulates the PA and is associated with IBS. The current review described the intestinal microecology and intestinal proteolytic activity of patients with IBS and also discussed the effect of intestinal flora on PA. In summary, this study proposed a pivotal role of gut microbiota and PA in IBS, respectively, and provided an in-depth insight into the diagnosis and treatment targets of IBS as well as the formulation of new treatment strategies for other digestive diseases and protease-related diseases.
Collapse
Affiliation(s)
- Jun-Jie Hou
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin, PR China.
| | - Xin Wang
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin, PR China.
| | - Ying Li
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin, PR China.
| | - Shuai Su
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin, PR China.
| | - Yu-Ming Wang
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin, PR China.
| | - Bang-Mao Wang
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin, PR China.
| |
Collapse
|
23
|
Galipeau HJ, Caminero A, Turpin W, Bermudez-Brito M, Santiago A, Libertucci J, Constante M, Raygoza Garay JA, Rueda G, Armstrong S, Clarizio A, Smith MI, Surette MG, Bercik P, Croitoru K, Verdu EF. Novel Fecal Biomarkers That Precede Clinical Diagnosis of Ulcerative Colitis. Gastroenterology 2021; 160:1532-1545. [PMID: 33310084 DOI: 10.1053/j.gastro.2020.12.004] [Citation(s) in RCA: 123] [Impact Index Per Article: 30.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 12/02/2020] [Accepted: 12/02/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND & AIMS Altered gut microbiota composition and function have been associated with inflammatory bowel diseases, including ulcerative colitis (UC), but the causality and mechanisms remain unknown. METHODS We applied 16S ribosomal RNA gene sequencing, shotgun metagenomic sequencing, in vitro functional assays, and gnotobiotic colonizations to define the microbial composition and function in fecal samples obtained from a cohort of healthy individuals at risk for inflammatory bowel diseases (pre-UC) who later developed UC (post-UC) and matched healthy control individuals (HCs). RESULTS Microbiota composition of post-UC samples was different from HC and pre-UC samples; however, functional analysis showed increased fecal proteolytic and elastase activity before UC onset. Metagenomics identified more than 22,000 gene families that were significantly different between HC, pre-UC, and post-UC samples. Of these, 237 related to proteases and peptidases, suggesting a bacterial component to the pre-UC proteolytic signature. Elastase activity inversely correlated with the relative abundance of Adlercreutzia and other potentially beneficial taxa and directly correlated with known proteolytic taxa, such as Bacteroides vulgatus. High elastase activity was confirmed in Bacteroides isolates from fecal samples. The bacterial contribution and functional significance of the proteolytic signature were investigated in germ-free adult mice and in dams colonized with HC, pre-UC, or post-UC microbiota. Mice colonized with or born from pre-UC-colonized dams developed higher fecal proteolytic activity and an inflammatory immune tone compared with HC-colonized mice. CONCLUSIONS We have identified increased fecal proteolytic activity that precedes the clinical diagnosis of UC and associates with gut microbiota changes. This proteolytic signature may constitute a noninvasive biomarker of inflammation to monitor at-risk populations that can be targeted therapeutically with antiproteases.
Collapse
Affiliation(s)
- Heather J Galipeau
- Farncombe Family Digestive Health Research Institute, Department of Medicine, McMaster University, Hamilton, Canada
| | - Alberto Caminero
- Farncombe Family Digestive Health Research Institute, Department of Medicine, McMaster University, Hamilton, Canada
| | - Williams Turpin
- Zane Cohen Centre for Digestive Diseases, Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada; Division of Gastroenterology, Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Miriam Bermudez-Brito
- Farncombe Family Digestive Health Research Institute, Department of Medicine, McMaster University, Hamilton, Canada
| | - Alba Santiago
- Farncombe Family Digestive Health Research Institute, Department of Medicine, McMaster University, Hamilton, Canada
| | - Josie Libertucci
- Farncombe Family Digestive Health Research Institute, Department of Medicine, McMaster University, Hamilton, Canada
| | - Marco Constante
- Farncombe Family Digestive Health Research Institute, Department of Medicine, McMaster University, Hamilton, Canada
| | - Juan Antonio Raygoza Garay
- Zane Cohen Centre for Digestive Diseases, Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada; Division of Gastroenterology, Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Gaston Rueda
- Farncombe Family Digestive Health Research Institute, Department of Medicine, McMaster University, Hamilton, Canada
| | - Sarah Armstrong
- Farncombe Family Digestive Health Research Institute, Department of Medicine, McMaster University, Hamilton, Canada
| | - Alex Clarizio
- Farncombe Family Digestive Health Research Institute, Department of Medicine, McMaster University, Hamilton, Canada
| | - Michelle I Smith
- Zane Cohen Centre for Digestive Diseases, Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Michael G Surette
- Farncombe Family Digestive Health Research Institute, Department of Medicine, McMaster University, Hamilton, Canada
| | - Premysl Bercik
- Farncombe Family Digestive Health Research Institute, Department of Medicine, McMaster University, Hamilton, Canada
| | - Kenneth Croitoru
- Zane Cohen Centre for Digestive Diseases, Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada; Division of Gastroenterology, Department of Medicine, University of Toronto, Toronto, Ontario, Canada.
| | - Elena F Verdu
- Farncombe Family Digestive Health Research Institute, Department of Medicine, McMaster University, Hamilton, Canada.
| |
Collapse
|
24
|
Duboux S, Golliard M, Muller JA, Bergonzelli G, Bolten CJ, Mercenier A, Kleerebezem M. Carbohydrate-controlled serine protease inhibitor (serpin) production in Bifidobacterium longum subsp. longum. Sci Rep 2021; 11:7236. [PMID: 33790385 PMCID: PMC8012564 DOI: 10.1038/s41598-021-86740-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Accepted: 03/19/2021] [Indexed: 12/12/2022] Open
Abstract
The Serine Protease Inhibitor (serpin) protein has been suggested to play a key role in the interaction of bifidobacteria with the host. By inhibiting intestinal serine proteases, it might allow bifidobacteria to reside in specific gut niches. In inflammatory diseases where serine proteases contribute to the innate defense mechanism of the host, serpin may dampen the damaging effects of inflammation. In view of the beneficial roles of this protein, it is important to understand how its production is regulated. Here we demonstrate that Bifidobacterium longum NCC 2705 serpin production is tightly regulated by carbohydrates. Galactose and fructose increase the production of this protein while glucose prevents it, suggesting the involvement of catabolite repression. We identified that di- and oligosaccharides containing galactose (GOS) and fructose (FOS) moieties, including the human milk oligosaccharide Lacto-N-tetraose (LNT), are able to activate serpin production. Moreover, we show that the carbohydrate mediated regulation is conserved within B. longum subsp. longum strains but not in other bifidobacterial taxons harboring the serpin coding gene, highlighting that the serpin regulation circuits are not only species- but also subspecies- specific. Our work demonstrates that environmental conditions can modulate expression of an important effector molecule of B. longum, having potential important implications for probiotic manufacturing and supporting the postulated role of serpin in the ability of bifidobacteria to colonize the intestinal tract.
Collapse
Affiliation(s)
- S Duboux
- Nestlé Research, Lausanne, Switzerland. .,Host-Microbe Interactomics Group, Wageningen University and Research, De Elst 1, 6708 WD, Wageningen, The Netherlands.
| | | | | | | | | | - A Mercenier
- Host-Microbe Interactomics Group, Wageningen University and Research, De Elst 1, 6708 WD, Wageningen, The Netherlands
| | - M Kleerebezem
- Host-Microbe Interactomics Group, Wageningen University and Research, De Elst 1, 6708 WD, Wageningen, The Netherlands.
| |
Collapse
|
25
|
Spence MA, Mortimer MD, Buckle AM, Minh BQ, Jackson CJ. A Comprehensive Phylogenetic Analysis of the Serpin Superfamily. Mol Biol Evol 2021; 38:2915-2929. [PMID: 33744972 PMCID: PMC8233489 DOI: 10.1093/molbev/msab081] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Serine protease inhibitors (serpins) are found in all kingdoms of life and play essential roles in multiple physiological processes. Owing to the diversity of the superfamily, phylogenetic analysis is challenging and prokaryotic serpins have been speculated to have been acquired from Metazoa through horizontal gene transfer due to their unexpectedly high homology. Here, we have leveraged a structural alignment of diverse serpins to generate a comprehensive 6,000-sequence phylogeny that encompasses serpins from all kingdoms of life. We show that in addition to a central “hub” of highly conserved serpins, there has been extensive diversification of the superfamily into many novel functional clades. Our analysis indicates that the hub proteins are ancient and are similar because of convergent evolution, rather than the alternative hypothesis of horizontal gene transfer. This work clarifies longstanding questions in the evolution of serpins and provides new directions for research in the field of serpin biology.
Collapse
Affiliation(s)
- Matthew A Spence
- Research School of Chemistry, Australian National University, Canberra, ACT, Australia
| | - Matthew D Mortimer
- Research School of Chemistry, Australian National University, Canberra, ACT, Australia
| | - Ashley M Buckle
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Melbourne, VIC, Australia
| | - Bui Quang Minh
- Research School of Computing and Research School of Biology, Australian National University, Canberra, ACT, Australia
| | - Colin J Jackson
- Research School of Chemistry, Australian National University, Canberra, ACT, Australia.,Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, Research School of Chemistry, Australian National University, Canberra, ACT, Australia.,Australian Research Council Centre of Excellence in Synthetic Biology, Research School of Chemistry, Australian National University, Canberra, ACT, Australia
| |
Collapse
|
26
|
Fu R, Zhang P, Jin G, Wang L, Qi S, Cao Y, Martin C, Zhang Y. Versatility in acyltransferase activity completes chicoric acid biosynthesis in purple coneflower. Nat Commun 2021; 12:1563. [PMID: 33692355 PMCID: PMC7946891 DOI: 10.1038/s41467-021-21853-6] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Accepted: 02/11/2021] [Indexed: 02/05/2023] Open
Abstract
Purple coneflower (Echinacea purpurea (L.) Moench) is a popular native North American herbal plant. Its major bioactive compound, chicoric acid, is reported to have various potential physiological functions, but little is known about its biosynthesis. Here, taking an activity-guided approach, we identify two cytosolic BAHD acyltransferases that form two intermediates, caftaric acid and chlorogenic acid. Surprisingly, a unique serine carboxypeptidase-like acyltransferase uses chlorogenic acid as its acyl donor and caftaric acid as its acyl acceptor to produce chicoric acid in vacuoles, which has evolved its acyl donor specificity from the better-known 1-O-β-D-glucose esters typical for this specific type of acyltransferase to chlorogenic acid. This unusual pathway seems unique to Echinacea species suggesting convergent evolution of chicoric acid biosynthesis. Using these identified acyltransferases, we have reconstituted chicoric acid biosynthesis in tobacco. Our results emphasize the flexibility of acyltransferases and their roles in the evolution of specialized metabolism in plants.
Collapse
Affiliation(s)
- Rao Fu
- Key Laboratory of Bio-resource and Eco-environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, 610064, China
| | - Pingyu Zhang
- Key Laboratory of Bio-resource and Eco-environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, 610064, China
| | - Ge Jin
- Key Laboratory of Bio-resource and Eco-environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, 610064, China
| | - Lianglei Wang
- Key Laboratory of Bio-resource and Eco-environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, 610064, China
| | - Shiqian Qi
- Department of Urology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Yang Cao
- Center of Growth, Metabolism and Aging, College of Life Sciences, Sichuan University, Chengdu, 610064, China
| | - Cathie Martin
- Department of Metabolic Biology and Biological Chemistry, John Innes Centre, Norwich, NR4 7UH, UK
| | - Yang Zhang
- Key Laboratory of Bio-resource and Eco-environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, 610064, China.
| |
Collapse
|
27
|
Mariaule V, Kriaa A, Soussou S, Rhimi S, Boudaya H, Hernandez J, Maguin E, Lesner A, Rhimi M. Digestive Inflammation: Role of Proteolytic Dysregulation. Int J Mol Sci 2021; 22:ijms22062817. [PMID: 33802197 PMCID: PMC7999743 DOI: 10.3390/ijms22062817] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 03/04/2021] [Accepted: 03/05/2021] [Indexed: 12/11/2022] Open
Abstract
Dysregulation of the proteolytic balance is often associated with diseases. Serine proteases and matrix metalloproteases are involved in a multitude of biological processes and notably in the inflammatory response. Within the framework of digestive inflammation, several studies have stressed the role of serine proteases and matrix metalloproteases (MMPs) as key actors in its pathogenesis and pointed to the unbalance between these proteases and their respective inhibitors. Substantial efforts have been made in developing new inhibitors, some of which have reached clinical trial phases, notwithstanding that unwanted side effects remain a major issue. However, studies on the proteolytic imbalance and inhibitors conception are directed toward host serine/MMPs proteases revealing a hitherto overlooked factor, the potential contribution of their bacterial counterpart. In this review, we highlight the role of proteolytic imbalance in human digestive inflammation focusing on serine proteases and MMPs and their respective inhibitors considering both host and bacterial origin.
Collapse
Affiliation(s)
- Vincent Mariaule
- Microbiota Interaction with Human and Animal Team (MIHA), Micalis Institute, AgroParisTech, Université Paris-Saclay, INRAE, F-78350 Jouy-en-Josas, France; (V.M.); (A.K.); (S.S.); (S.R.); (H.B.); (E.M.)
| | - Aicha Kriaa
- Microbiota Interaction with Human and Animal Team (MIHA), Micalis Institute, AgroParisTech, Université Paris-Saclay, INRAE, F-78350 Jouy-en-Josas, France; (V.M.); (A.K.); (S.S.); (S.R.); (H.B.); (E.M.)
| | - Souha Soussou
- Microbiota Interaction with Human and Animal Team (MIHA), Micalis Institute, AgroParisTech, Université Paris-Saclay, INRAE, F-78350 Jouy-en-Josas, France; (V.M.); (A.K.); (S.S.); (S.R.); (H.B.); (E.M.)
| | - Soufien Rhimi
- Microbiota Interaction with Human and Animal Team (MIHA), Micalis Institute, AgroParisTech, Université Paris-Saclay, INRAE, F-78350 Jouy-en-Josas, France; (V.M.); (A.K.); (S.S.); (S.R.); (H.B.); (E.M.)
| | - Houda Boudaya
- Microbiota Interaction with Human and Animal Team (MIHA), Micalis Institute, AgroParisTech, Université Paris-Saclay, INRAE, F-78350 Jouy-en-Josas, France; (V.M.); (A.K.); (S.S.); (S.R.); (H.B.); (E.M.)
| | - Juan Hernandez
- Department of Clinical Sciences, Nantes-Atlantic College of Veterinary Medicine and Food Sciences (Oniris), University of Nantes, 101 Route de Gachet, 44300 Nantes, France;
| | - Emmanuelle Maguin
- Microbiota Interaction with Human and Animal Team (MIHA), Micalis Institute, AgroParisTech, Université Paris-Saclay, INRAE, F-78350 Jouy-en-Josas, France; (V.M.); (A.K.); (S.S.); (S.R.); (H.B.); (E.M.)
| | - Adam Lesner
- Faculty of Chemistry, University of Gdansk, Wita Stwosza 63, PL80-308 Gdansk, Poland;
| | - Moez Rhimi
- Microbiota Interaction with Human and Animal Team (MIHA), Micalis Institute, AgroParisTech, Université Paris-Saclay, INRAE, F-78350 Jouy-en-Josas, France; (V.M.); (A.K.); (S.S.); (S.R.); (H.B.); (E.M.)
- Correspondence:
| |
Collapse
|
28
|
Haber M, Burgsdorf I, Handley KM, Rubin-Blum M, Steindler L. Genomic Insights Into the Lifestyles of Thaumarchaeota Inside Sponges. Front Microbiol 2021; 11:622824. [PMID: 33537022 PMCID: PMC7848895 DOI: 10.3389/fmicb.2020.622824] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Accepted: 12/14/2020] [Indexed: 11/28/2022] Open
Abstract
Sponges are among the oldest metazoans and their success is partly due to their abundant and diverse microbial symbionts. They are one of the few animals that have Thaumarchaeota symbionts. Here we compare genomes of 11 Thaumarchaeota sponge symbionts, including three new genomes, to free-living ones. Like their free-living counterparts, sponge-associated Thaumarchaeota can oxidize ammonia, fix carbon, and produce several vitamins. Adaptions to life inside the sponge host include enrichment in transposases, toxin-antitoxin systems and restriction modifications systems, enrichments previously reported also from bacterial sponge symbionts. Most thaumarchaeal sponge symbionts lost the ability to synthesize rhamnose, which likely alters their cell surface and allows them to evade digestion by the host. All but one archaeal sponge symbiont encoded a high-affinity, branched-chain amino acid transporter system that was absent from the analyzed free-living thaumarchaeota suggesting a mixotrophic lifestyle for the sponge symbionts. Most of the other unique features found in sponge-associated Thaumarchaeota, were limited to only a few specific symbionts. These features included the presence of exopolyphosphatases and a glycine cleavage system found in the novel genomes. Thaumarchaeota have thus likely highly specific interactions with their sponge host, which is supported by the limited number of host sponge species to which each of these symbionts is restricted.
Collapse
Affiliation(s)
- Markus Haber
- Department of Marine Biology, Leon H. Charney School of Marine Sciences, University of Haifa, Haifa, Israel
- Department of Aquatic Microbial Ecology, Institute of Hydrobiology, Biology Centre CAS, České Budějovice, Czechia
| | - Ilia Burgsdorf
- Department of Marine Biology, Leon H. Charney School of Marine Sciences, University of Haifa, Haifa, Israel
| | - Kim M. Handley
- School of Biological Sciences, The University of Auckland, Auckland, New Zealand
| | - Maxim Rubin-Blum
- Israel Oceanographic and Limnological Research Institute, Haifa, Israel
| | - Laura Steindler
- Department of Marine Biology, Leon H. Charney School of Marine Sciences, University of Haifa, Haifa, Israel
| |
Collapse
|
29
|
Exploring the Ecology of Bifidobacteria and Their Genetic Adaptation to the Mammalian Gut. Microorganisms 2020; 9:microorganisms9010008. [PMID: 33375064 PMCID: PMC7822027 DOI: 10.3390/microorganisms9010008] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 12/16/2020] [Accepted: 12/18/2020] [Indexed: 12/14/2022] Open
Abstract
The mammalian gut is densely inhabited by microorganisms that have coevolved with their host. Amongst these latter microorganisms, bifidobacteria represent a key model to study host–microbe interaction within the mammalian gut. Remarkably, bifidobacteria naturally occur in a range of ecological niches that are either directly or indirectly connected to the animal gastrointestinal tract. They constitute one of the dominant bacterial members of the intestinal microbiota and are among the first colonizers of the mammalian gut. Notably, the presence of bifidobacteria in the gut has been associated with several health-promoting activities. In this review, we aim to provide an overview of current knowledge on the genetic diversity and ecology of bifidobacteria. Furthermore, we will discuss how this important group of gut bacteria is able to colonize and survive in the mammalian gut, so as to facilitate host interactions.
Collapse
|
30
|
Redweik GAJ, Jochum J, Mellata M. Live Bacterial Prophylactics in Modern Poultry. Front Vet Sci 2020; 7:592312. [PMID: 33195630 PMCID: PMC7655978 DOI: 10.3389/fvets.2020.592312] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 09/21/2020] [Indexed: 11/25/2022] Open
Abstract
Commercial poultry farms frequently use live bacterial prophylactics like vaccines and probiotics to prevent bacterial infections. Due to the emergence of antibiotic-resistant bacteria in poultry animals, a closer examination into the health benefits and limitations of commercial, live prophylactics as an alternative to antibiotics is urgently needed. In this review, we summarize the peer-reviewed literature of several commercial live bacterial vaccines and probiotics. Per our estimation, there is a paucity of peer-reviewed published research regarding these products, making repeatability, product-comparison, and understanding biological mechanisms difficult. Furthermore, we briefly-outline significant issues such as probiotic-label accuracy, lack of commercially available live bacterial vaccines for major poultry-related bacteria such as Campylobacter and Clostridium perfringens, as well research gaps (i.e., probiotic-mediated vaccine adjuvancy, gut-brain-microbiota axis). Increased emphasis on these areas would open several avenues for research, ranging from improving protection against bacterial pathogens to using these prophylactics to modulate animal behavior.
Collapse
Affiliation(s)
- Graham A. J. Redweik
- Department of Food Science and Human Nutrition, Iowa State University, Ames, IA, United States
- Interdepartmental Microbiology Graduate Program, Iowa State University, Ames, IA, United States
| | - Jared Jochum
- Department of Food Science and Human Nutrition, Iowa State University, Ames, IA, United States
- Interdepartmental Microbiology Graduate Program, Iowa State University, Ames, IA, United States
| | - Melha Mellata
- Department of Food Science and Human Nutrition, Iowa State University, Ames, IA, United States
- Interdepartmental Microbiology Graduate Program, Iowa State University, Ames, IA, United States
| |
Collapse
|
31
|
|
32
|
Ruan W, Engevik MA, Spinler JK, Versalovic J. Healthy Human Gastrointestinal Microbiome: Composition and Function After a Decade of Exploration. Dig Dis Sci 2020; 65:695-705. [PMID: 32067143 DOI: 10.1007/s10620-020-06118-4] [Citation(s) in RCA: 135] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The human gastrointestinal (GI) tract contains communities of microbes (bacteria, fungi, viruses) that vary by anatomic location and impact human health. Microbial communities differ in composition based on age, diet, and location in the gastrointestinal tract. Differences in microbial composition have been associated with chronic disease states. In terms of function, microbial metabolites provide key signals that help maintain healthy human physiology. Alterations of the healthy gastrointestinal microbiome have been linked to the development of various disease states including inflammatory bowel disease, diabetes, and colorectal cancer. While the definition of a healthy GI microbiome cannot be precisely identified, features of a healthy gut microbiome include relatively greater biodiversity and relative abundances of specific phyla and genera. Microbes with desirable functional profiles for the human host have been identified, in addition to specific metabolic features of the microbiome. This article reviews the composition and function of the healthy human GI microbiome, including the relative abundances of different bacterial taxa and the specific metabolic pathways and classes of microbial metabolites contributing to human health and disease prevention.
Collapse
Affiliation(s)
- Wenly Ruan
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA.,Section of Gastroenterology, Hepatology, and Nutrition, Texas Children's Hospital, Houston, TX, USA
| | - Melinda A Engevik
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX, USA.,Department of Pathology, Texas Children's Hospital, 1102 Bates St., Feigin Tower Suite 830, Houston, TX, 77030, USA
| | - Jennifer K Spinler
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX, USA.,Department of Pathology, Texas Children's Hospital, 1102 Bates St., Feigin Tower Suite 830, Houston, TX, 77030, USA
| | - James Versalovic
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX, USA. .,Department of Pathology, Texas Children's Hospital, 1102 Bates St., Feigin Tower Suite 830, Houston, TX, 77030, USA.
| |
Collapse
|
33
|
Yong SJ, Tong T, Chew J, Lim WL. Antidepressive Mechanisms of Probiotics and Their Therapeutic Potential. Front Neurosci 2020; 13:1361. [PMID: 32009871 PMCID: PMC6971226 DOI: 10.3389/fnins.2019.01361] [Citation(s) in RCA: 105] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2019] [Accepted: 12/02/2019] [Indexed: 12/16/2022] Open
Abstract
The accumulating knowledge of the host-microbiota interplay gives rise to the microbiota-gut-brain (MGB) axis. The MGB axis depicts the interkingdom communication between the gut microbiota and the brain. This communication process involves the endocrine, immune and neurotransmitters systems. Dysfunction of these systems, along with the presence of gut dysbiosis, have been detected among clinically depressed patients. This implicates the involvement of a maladaptive MGB axis in the pathophysiology of depression. Depression refers to symptoms that characterize major depressive disorder (MDD), a mood disorder with a disease burden that rivals that of heart diseases. The use of probiotics to treat depression has gained attention in recent years, as evidenced by increasing numbers of animal and human studies that have supported the antidepressive efficacy of probiotics. Physiological changes observed in these studies allow for the elucidation of probiotics antidepressive mechanisms, which ultimately aim to restore proper functioning of the MGB axis. However, the understanding of mechanisms does not yet complete the endeavor in applying probiotics to treat MDD. Other challenges remain which include the heterogeneous nature of both the gut microbiota composition and depressive symptoms in the clinical setting. Nevertheless, probiotics offer some advantages over standard pharmaceutical antidepressants, in terms of residual symptoms, side effects and stigma involved. This review outlines antidepressive mechanisms of probiotics based on the currently available literature and discusses therapeutic potentials of probiotics for depression.
Collapse
Affiliation(s)
- Shin Jie Yong
- Department of Biological Sciences, School of Science and Technology, Sunway University, Bandar Sunway, Malaysia
| | - Tommy Tong
- Department of Biological Sciences, School of Science and Technology, Sunway University, Bandar Sunway, Malaysia
| | - Jactty Chew
- Department of Biological Sciences, School of Science and Technology, Sunway University, Bandar Sunway, Malaysia
| | - Wei Ling Lim
- Department of Biological Sciences, School of Science and Technology, Sunway University, Bandar Sunway, Malaysia
| |
Collapse
|
34
|
Pyclik M, Srutkova D, Schwarzer M, Górska S. Bifidobacteria cell wall-derived exo-polysaccharides, lipoteichoic acids, peptidoglycans, polar lipids and proteins - their chemical structure and biological attributes. Int J Biol Macromol 2019; 147:333-349. [PMID: 31899242 DOI: 10.1016/j.ijbiomac.2019.12.227] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 12/13/2019] [Accepted: 12/24/2019] [Indexed: 02/07/2023]
Abstract
A variety of health benefits has been documented to be associated with the consumption of probiotic bacteria, namely bifidobacteria and lactobacilli. Thanks to the scientific advances in recent years we are beginning to understand the molecular mechanisms by which bacteria in general and probiotic bacteria in particular act as host physiology and immune system modulators. More recently, the focus has shifted from live bacteria towards bacteria-derived defined molecules, so called postbiotics. These molecules may represent safer alternative compared to the live bacteria while retaining the desired effects on the host. The excellent source of effector macromolecules is the bacterial envelope. It contains compounds that are pivotal in the adhesion phenomenon, provide direct bacteria-to-host signaling capacity and the associated physiological impact and immunomodulatory properties of bacteria. Here we comprehensively review the structure and biological role of Bifidobacterium surface and cell wall molecules: exopolysaccharides, cell wall polysaccharides, lipoteichoic acids, polar lipids, peptidoglycans and proteins. We discuss their involvement in direct signaling to the host cells and their described immunomodulatory effects.
Collapse
Affiliation(s)
- Marcelina Pyclik
- Laboratory of Microbiome Immunobiology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Weigla 12, 53-114 Wroclaw, Poland
| | - Dagmar Srutkova
- Laboratory of Gnotobiology, Institute of Microbiology of the Czech Academy of Sciences, Novy Hradek, Czech Republic
| | - Martin Schwarzer
- Laboratory of Gnotobiology, Institute of Microbiology of the Czech Academy of Sciences, Novy Hradek, Czech Republic.
| | - Sabina Górska
- Laboratory of Microbiome Immunobiology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Weigla 12, 53-114 Wroclaw, Poland.
| |
Collapse
|
35
|
Alessandri G, Ossiprandi MC, MacSharry J, van Sinderen D, Ventura M. Bifidobacterial Dialogue With Its Human Host and Consequent Modulation of the Immune System. Front Immunol 2019; 10:2348. [PMID: 31632412 PMCID: PMC6779802 DOI: 10.3389/fimmu.2019.02348] [Citation(s) in RCA: 87] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Accepted: 09/17/2019] [Indexed: 12/12/2022] Open
Abstract
Since bifidobacteria are among the pioneering colonizers of the human infant gut, their interaction with their host is believed to start soon following birth. Several members of the Bifidobacterium genus are purported to exert various health-promoting effects at local and systemic levels, e.g., limiting pathogen colonization/invasion, influencing gut homeostasis, and influencing the immune system through changes in innate and/or adaptive immune responses. This has promoted extensive research efforts to shed light on the precise mechanisms by which bifidobacteria are able to stimulate and interact with the host immune system. These studies uncovered a variety of secreted or surface-associated molecules that act as essential mediators for the establishment of a bifidobacteria-host immune system dialogue, and that allow interactions with mucosa-associated immune cells. Additionally, the by-products generated from bifidobacterial carbohydrate metabolism act as vectors that directly and indirectly trigger the host immune response, the latter by stimulating growth of other commensal microorganisms such as propionate- or butyrate-producing bacteria. This review is aimed to provide a comprehensive overview on the wide variety of strategies employed by bifidobacteria to engage with the host immune system.
Collapse
Affiliation(s)
- Giulia Alessandri
- Department of Veterinary Medical Science, University of Parma, Parma, Italy
| | - Maria Cristina Ossiprandi
- Department of Veterinary Medical Science, University of Parma, Parma, Italy
- Microbiome Research Hub, University of Parma, Parma, Italy
| | - John MacSharry
- APC Microbiome Institute and School of Microbiology, Bioscience Institute, National University of Ireland, Cork, Ireland
| | - Douwe van Sinderen
- APC Microbiome Institute and School of Microbiology, Bioscience Institute, National University of Ireland, Cork, Ireland
| | - Marco Ventura
- Microbiome Research Hub, University of Parma, Parma, Italy
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences, and Environmental Sustainability, University of Parma, Parma, Italy
| |
Collapse
|
36
|
Mkaouar H, Akermi N, Kriaa A, Abraham AL, Jablaoui A, Soussou S, Mokdad-Gargouri R, Maguin E, Rhimi M. Serine protease inhibitors and human wellbeing interplay: new insights for old friends. PeerJ 2019; 7:e7224. [PMID: 31531264 PMCID: PMC6718151 DOI: 10.7717/peerj.7224] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Accepted: 05/31/2019] [Indexed: 12/14/2022] Open
Abstract
Serine Protease Inhibitors (Serpins) control tightly regulated physiological processes and their dysfunction is associated to various diseases. Thus, increasing interest is given to these proteins as new therapeutic targets. Several studies provided functional and structural data about human serpins. By comparison, only little knowledge regarding bacterial serpins exists. Through the emergence of metagenomic studies, many bacterial serpins were identified from numerous ecological niches including the human gut microbiota. The origin, distribution and function of these proteins remain to be established. In this report, we shed light on the key role of human and bacterial serpins in health and disease. Moreover, we analyze their function, phylogeny and ecological distribution. This review highlights the potential use of bacterial serpins to set out new therapeutic approaches.
Collapse
Affiliation(s)
- Héla Mkaouar
- INRA, UMR1319 MICALIS, Jouy-en-Josas, France, AgroParisTech, UMR MICALIS, Jouy-en-Josas, France
| | - Nizar Akermi
- INRA, UMR1319 MICALIS, Jouy-en-Josas, France, AgroParisTech, UMR MICALIS, Jouy-en-Josas, France
| | - Aicha Kriaa
- INRA, UMR1319 MICALIS, Jouy-en-Josas, France, AgroParisTech, UMR MICALIS, Jouy-en-Josas, France
| | | | - Amin Jablaoui
- INRA, UMR1319 MICALIS, Jouy-en-Josas, France, AgroParisTech, UMR MICALIS, Jouy-en-Josas, France
| | - Souha Soussou
- INRA, UMR1319 MICALIS, Jouy-en-Josas, France, AgroParisTech, UMR MICALIS, Jouy-en-Josas, France
| | - Raja Mokdad-Gargouri
- Laboratory of Molecular Biology of Eukaryotes, Center of Biotechnology of Sfax, University of Sfax, Sfax, Tunisia
| | - Emmanuelle Maguin
- INRA, UMR1319 MICALIS, Jouy-en-Josas, France, AgroParisTech, UMR MICALIS, Jouy-en-Josas, France
| | - Moez Rhimi
- INRA, UMR1319 MICALIS, Jouy-en-Josas, France, AgroParisTech, UMR MICALIS, Jouy-en-Josas, France
| |
Collapse
|
37
|
Zakharevich NV, Nezametdinova VZ, Averina OV, Chekalina MS, Alekseeva MG, Danilenko VN. Complete Genome Sequence of Bifidobacterium angulatum GT102: Potential Genes and Systems of Communication with Host. RUSS J GENET+ 2019. [DOI: 10.1134/s1022795419070160] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
38
|
Akermi N, Mkaouar H, Kriaa A, Jablaoui A, Soussou S, Gargouri A, Coleman AW, Perret F, Maguin E, Rhimi M. para-Sulphonato-calix[n]arene capped silver nanoparticles challenge the catalytic efficiency and the stability of a novel human gut serine protease inhibitor. Chem Commun (Camb) 2019; 55:8935-8938. [PMID: 31286126 DOI: 10.1039/c9cc03183a] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
The Eubacterium saburreum serine protease inhibitor from the human gut microbiota inhibits the eukaryotic pancreatic elastase associated with acute pancreatitis. Interestingly, the inhibition efficiency and stability are markedly increased by the para-sulphonato-calix[8]arene capped silver nanoparticles. Moreover, this enzyme is distinguishable by its high inhibitory effect at broad pH range between 2-10 and temperatures from 10 to 40 °C, in the presence of para-sulphonato-calix[8]arene capped silver nanoparticles the enzyme remains active even at 70 °C.
Collapse
Affiliation(s)
- Nizar Akermi
- Micalis Institute, INRA, AgroParisTech, Université Paris-Saclay, F-78350 Jouy-en-Josas, France.
| | - Hela Mkaouar
- Micalis Institute, INRA, AgroParisTech, Université Paris-Saclay, F-78350 Jouy-en-Josas, France.
| | - Aicha Kriaa
- Micalis Institute, INRA, AgroParisTech, Université Paris-Saclay, F-78350 Jouy-en-Josas, France.
| | - Amin Jablaoui
- Micalis Institute, INRA, AgroParisTech, Université Paris-Saclay, F-78350 Jouy-en-Josas, France.
| | - Souha Soussou
- Micalis Institute, INRA, AgroParisTech, Université Paris-Saclay, F-78350 Jouy-en-Josas, France.
| | - Ali Gargouri
- Laboratory of Molecular Biology of Eukaryotes, Center of Biotechnology of Sfax, University of Sfax, 3038, Sfax, Tunisia
| | | | - Florent Perret
- Univ Lyon, Université Lyon 1, Institut de Chimie et Biochimie Moléculaires et Supramoléculaires, UMR 5246 CNRS, F-69622, Villeurbanne, France
| | - Emmanuelle Maguin
- Micalis Institute, INRA, AgroParisTech, Université Paris-Saclay, F-78350 Jouy-en-Josas, France.
| | - Moez Rhimi
- Micalis Institute, INRA, AgroParisTech, Université Paris-Saclay, F-78350 Jouy-en-Josas, France.
| |
Collapse
|
39
|
Pasquevich KA, Carabajal MV, Guaimas FF, Bruno L, Roset MS, Coria LM, Rey Serrantes DA, Comerci DJ, Cassataro J. Omp19 Enables Brucella abortus to Evade the Antimicrobial Activity From Host's Proteolytic Defense System. Front Immunol 2019; 10:1436. [PMID: 31297115 PMCID: PMC6607954 DOI: 10.3389/fimmu.2019.01436] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Accepted: 06/07/2019] [Indexed: 01/18/2023] Open
Abstract
Pathogenic microorganisms confront several proteolytic events in the molecular interplay with their host, highlighting that proteolysis and its regulation play an important role during infection. Microbial inhibitors, along with their target endogenous/exogenous enzymes, may directly affect the host's defense mechanisms and promote infection. Omp19 is a Brucella spp. conserved lipoprotein anchored by the lipid portion in the Brucella outer membrane. Previous work demonstrated that purified unlipidated Omp19 (U-Omp19) has protease inhibitor activity against gastrointestinal and lysosomal proteases. In this work, we found that a Brucella omp19 deletion mutant is highly attenuated in mice when infecting by the oral route. This attenuation can be explained by bacterial increased susceptibility to host proteases met by the bacteria during establishment of infection. Omp19 deletion mutant has a cell division defect when exposed to pancreatic proteases that is linked to cell-cycle arrest in G1-phase, Omp25 degradation on the cell envelope and CtrA accumulation. Moreover, Omp19 deletion mutant is more susceptible to killing by macrophage derived microsomes than wt strain. Preincubation with gastrointestinal proteases led to an increased susceptibility of Omp19 deletion mutant to macrophage intracellular killing. Thus, in this work, we describe for the first time a physiological function of B. abortus Omp19. This activity enables Brucella to better thrive in the harsh gastrointestinal tract, where protection from proteolytic degradation can be a matter of life or death, and afterwards invade the host and bypass intracellular proteases to establish the chronic infection.
Collapse
Affiliation(s)
- Karina A Pasquevich
- Consejo Nacional de Investigaciones Científicas y Técnicas (UNSAM-CONICET), Instituto de Investigaciones Biotecnológicas Dr. Rodolfo A. Ugalde, Universidad Nacional de San Martín, Buenos Aires, Argentina
| | - Marianela V Carabajal
- Consejo Nacional de Investigaciones Científicas y Técnicas (UNSAM-CONICET), Instituto de Investigaciones Biotecnológicas Dr. Rodolfo A. Ugalde, Universidad Nacional de San Martín, Buenos Aires, Argentina
| | - Francisco F Guaimas
- Consejo Nacional de Investigaciones Científicas y Técnicas (UNSAM-CONICET), Instituto de Investigaciones Biotecnológicas Dr. Rodolfo A. Ugalde, Universidad Nacional de San Martín, Buenos Aires, Argentina
| | - Laura Bruno
- Consejo Nacional de Investigaciones Científicas y Técnicas (UNSAM-CONICET), Instituto de Investigaciones Biotecnológicas Dr. Rodolfo A. Ugalde, Universidad Nacional de San Martín, Buenos Aires, Argentina
| | - Mara S Roset
- Consejo Nacional de Investigaciones Científicas y Técnicas (UNSAM-CONICET), Instituto de Investigaciones Biotecnológicas Dr. Rodolfo A. Ugalde, Universidad Nacional de San Martín, Buenos Aires, Argentina
| | - Lorena M Coria
- Consejo Nacional de Investigaciones Científicas y Técnicas (UNSAM-CONICET), Instituto de Investigaciones Biotecnológicas Dr. Rodolfo A. Ugalde, Universidad Nacional de San Martín, Buenos Aires, Argentina
| | - Diego A Rey Serrantes
- Consejo Nacional de Investigaciones Científicas y Técnicas (UNSAM-CONICET), Instituto de Investigaciones Biotecnológicas Dr. Rodolfo A. Ugalde, Universidad Nacional de San Martín, Buenos Aires, Argentina
| | - Diego J Comerci
- Consejo Nacional de Investigaciones Científicas y Técnicas (UNSAM-CONICET), Instituto de Investigaciones Biotecnológicas Dr. Rodolfo A. Ugalde, Universidad Nacional de San Martín, Buenos Aires, Argentina
| | - Juliana Cassataro
- Consejo Nacional de Investigaciones Científicas y Técnicas (UNSAM-CONICET), Instituto de Investigaciones Biotecnológicas Dr. Rodolfo A. Ugalde, Universidad Nacional de San Martín, Buenos Aires, Argentina
| |
Collapse
|
40
|
Abstract
Aim:to review available data confirming the pathogenetic role of the intestinal microbiota in the formation of irritable bowel syndrome (IBS).Key findings.Changes in the intestinal biotope cause the development of visceral hypersensitivity and impaired intestinal motor activity, as well as neuroimmune transmission. This article discusses the main aspects of the biological properties of probiotic bacteria in terms of their action within the “brain — intestine — microbiota” chain. The results of experimental and clinical studies elucidating the mechanisms of action of probiotic cultures have been generalized. The understanding of these mechanisms allows practitioners to make informed decisions in prescribing probiotics to IBS patients. Key concepts concerning fecal microbiota transplantation, as well as the prospects and difficulties of implementing this approach are considered.Conclusions.The term “microbiota — intestine — brain” clearly demonstrates the correlation between the main functional components of IBS. Meta-analyses and systematic reviews confirm the efficacy of probiotics in IBS. However, further research into probiotic therapy options is needed to identify specific bacterial strains with proven clinical efficacy. The fecal microbiota transplantation method also requires further research, since many issues associated with this approach remain unclear.
Collapse
|
41
|
Clemente M, Corigliano MG, Pariani SA, Sánchez-López EF, Sander VA, Ramos-Duarte VA. Plant Serine Protease Inhibitors: Biotechnology Application in Agriculture and Molecular Farming. Int J Mol Sci 2019; 20:E1345. [PMID: 30884891 PMCID: PMC6471620 DOI: 10.3390/ijms20061345] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2019] [Revised: 02/14/2019] [Accepted: 02/18/2019] [Indexed: 11/12/2022] Open
Abstract
The serine protease inhibitors (SPIs) are widely distributed in living organisms like bacteria, fungi, plants, and humans. The main function of SPIs as protease enzymes is to regulate the proteolytic activity. In plants, most of the studies of SPIs have been focused on their physiological role. The initial studies carried out in plants showed that SPIs participate in the regulation of endogenous proteolytic processes, as the regulation of proteases in seeds. Besides, it was observed that SPIs also participate in the regulation of cell death during plant development and senescence. On the other hand, plant SPIs have an important role in plant defense against pests and phytopathogenic microorganisms. In the last 20 years, several transgenic plants over-expressing SPIs have been produced and tested in order to achieve the increase of the resistance against pathogenic insects. Finally, in molecular farming, SPIs have been employed to minimize the proteolysis of recombinant proteins expressed in plants. The present review discusses the potential biotechnological applications of plant SPIs in the agriculture field.
Collapse
Affiliation(s)
- Marina Clemente
- Instituto Tecnológico Chascomús (INTECH), UNSAM-CONICET, Chascomús, Provincia de Buenos Aires B7130, Argentina.
| | - Mariana G Corigliano
- Instituto Tecnológico Chascomús (INTECH), UNSAM-CONICET, Chascomús, Provincia de Buenos Aires B7130, Argentina.
| | - Sebastián A Pariani
- Instituto Tecnológico Chascomús (INTECH), UNSAM-CONICET, Chascomús, Provincia de Buenos Aires B7130, Argentina.
| | - Edwin F Sánchez-López
- Instituto Tecnológico Chascomús (INTECH), UNSAM-CONICET, Chascomús, Provincia de Buenos Aires B7130, Argentina.
| | - Valeria A Sander
- Instituto Tecnológico Chascomús (INTECH), UNSAM-CONICET, Chascomús, Provincia de Buenos Aires B7130, Argentina.
| | - Víctor A Ramos-Duarte
- Instituto Tecnológico Chascomús (INTECH), UNSAM-CONICET, Chascomús, Provincia de Buenos Aires B7130, Argentina.
| |
Collapse
|
42
|
|
43
|
Dicks LMT, Dreyer L, Smith C, van Staden AD. A Review: The Fate of Bacteriocins in the Human Gastro-Intestinal Tract: Do They Cross the Gut-Blood Barrier? Front Microbiol 2018; 9:2297. [PMID: 30323796 PMCID: PMC6173059 DOI: 10.3389/fmicb.2018.02297] [Citation(s) in RCA: 103] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Accepted: 09/07/2018] [Indexed: 12/20/2022] Open
Abstract
The intestinal barrier, consisting of the vascular endothelium, epithelial cell lining, and mucus layer, covers a surface of about 400 m2. The integrity of the gut wall is sustained by transcellular proteins forming tight junctions between the epithelial cells. Protected by three layers of mucin, the gut wall forms a non-permeable barrier, keeping digestive enzymes and microorganisms within the luminal space, separate from the blood stream. Microorganisms colonizing the gut may produce bacteriocins in an attempt to outcompete pathogens. Production of bacteriocins in a harsh and complex environment such as the gastro-intestinal tract (GIT) may be below minimal inhibitory concentration (MIC) levels. At such low levels, the stability of bacteriocins may be compromised. Despite this, most bacteria in the gut have the ability to produce bacteriocins, distributed throughout the GIT. With most antimicrobial studies being performed in vitro, we know little about the migration of bacteriocins across epithelial barriers. The behavior of bacteriocins in the GIT is studied ex vivo, using models, flow cells, or membranes resembling the gut wall. Furthermore, little is known about the effect bacteriocins have on the immune system. It is generally believed that the peptides will be destroyed by macrophages once they cross the gut wall. Studies done on the survival of neurotherapeutic peptides and their crossing of the brain-blood barrier, along with other studies on small peptides intravenously injected, may provide some answers. In this review, the stability of bacteriocins in the GIT, their effect on gut epithelial cells, and their ability to cross epithelial cells are discussed. These are important questions to address in the light of recent papers advocating the use of bacteriocins as possible alternatives to, or used in combination with, antibiotics.
Collapse
Affiliation(s)
- Leon M. T. Dicks
- Department of Microbiology, Stellenbosch University, Stellenbosch, South Africa
| | - Leané Dreyer
- Department of Microbiology, Stellenbosch University, Stellenbosch, South Africa
| | - Carine Smith
- Department of Physiological Sciences, Stellenbosch University, Stellenbosch, South Africa
| | - Anton D. van Staden
- Department of Microbiology, Stellenbosch University, Stellenbosch, South Africa
- Department of Physiological Sciences, Stellenbosch University, Stellenbosch, South Africa
| |
Collapse
|
44
|
Vinusha KS, Deepika K, Johnson TS, Agrawal GK, Rakwal R. Proteomic studies on lactic acid bacteria: A review. Biochem Biophys Rep 2018; 14:140-148. [PMID: 29872746 PMCID: PMC5986552 DOI: 10.1016/j.bbrep.2018.04.009] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2018] [Revised: 04/02/2018] [Accepted: 04/17/2018] [Indexed: 02/07/2023] Open
Abstract
Probiotics are amongst the most common microbes in the gastro-intestinal tract of humans and other animals. Prominent among probiotics are Lactobacillus and Bifidobacterium. They offer wide-ranging health promoting benefits to the host which include reduction in pathological alterations, stimulation of mucosal immunity and interaction with mediators of inflammation among others. Proteomics plays a vital role in understanding biological functions of a cell. Proteomics is also slowly and steadily adding to the existing knowledge on role of probiotics. In this paper, the proteomics of probiotics, with special reference to lactic acid bacteria is reviewed with a view to understand i) proteome map, ii) mechanism of adaptation to harsh gut environment such as low pH and bile acid, iii) role of cell surface proteins in adhering to intestinal epithelial cells, and iv) as a tool to answer basic cell functions. We have also reviewed various analytical methods used to carry out proteome analysis, in which 2D-MS and LC-MS/MS approaches were found to be versatile methods to perform high-throughput sample analyses even for a complex gut samples. Further, we present future road map of understanding gut microbes combining meta-proteomics, meta-genomics, meta-transcriptomics and -metabolomics.
Collapse
Affiliation(s)
- K Sri Vinusha
- Department of Biotechnology, K. L. E. F. deemed University, Guntur District, Vaddeswaram, Andhra Pradesh 522502, India
| | - K Deepika
- Department of Biotechnology, K. L. E. F. deemed University, Guntur District, Vaddeswaram, Andhra Pradesh 522502, India
| | - T Sudhakar Johnson
- Department of Biotechnology, K. L. E. F. deemed University, Guntur District, Vaddeswaram, Andhra Pradesh 522502, India
| | - Ganesh K Agrawal
- Research Laboratory for Biotechnology and Biochemistry (RLABB), GPO Box 13265, Kathmandu, Nepal.,GRADE Academy Private Limited, Adarsh Nagar-13, Birgunj, Nepal
| | - Randeep Rakwal
- Research Laboratory for Biotechnology and Biochemistry (RLABB), GPO Box 13265, Kathmandu, Nepal.,GRADE Academy Private Limited, Adarsh Nagar-13, Birgunj, Nepal.,Faculty of Health and Sport Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8574, Japan.,Global Research Center for Innovative Life Science, Peptide Drug Innovation, School of Pharmacy and Pharmaceutical Sciences, Hoshi University, 4-41 Ebara 2-chome, Shinagawa, Tokyo 142-8501, Japan
| |
Collapse
|
45
|
Buhner S, Hahne H, Hartwig K, Li Q, Vignali S, Ostertag D, Meng C, Hörmannsperger G, Braak B, Pehl C, Frieling T, Barbara G, De Giorgio R, Demir IE, Ceyhan GO, Zeller F, Boeckxstaens G, Haller D, Kuster B, Schemann M. Protease signaling through protease activated receptor 1 mediate nerve activation by mucosal supernatants from irritable bowel syndrome but not from ulcerative colitis patients. PLoS One 2018. [PMID: 29529042 PMCID: PMC5846775 DOI: 10.1371/journal.pone.0193943] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Background & aims The causes of gastrointestinal complaints in irritable bowel syndrome (IBS) remain poorly understood. Altered nerve function has emerged as an important pathogenic factor as IBS mucosal biopsy supernatants consistently activate enteric and sensory neurons. We investigated the neurally active molecular components of such supernatants from patients with IBS and quiescent ulcerative colitis (UC). Method Effects of supernatants from 7 healthy controls (HC), 20 IBS and 12 UC patients on human and guinea pig submucous neurons were studied with neuroimaging techniques. We identify differentially expressed proteins with proteome analysis. Results Nerve activation by IBS supernatants was prevented by the protease activated receptor 1 (PAR1) antagonist SCHE79797. UC supernatants also activated enteric neurons through protease dependent mechanisms but without PAR1 involvement. Proteome analysis of the supernatants identified 204 proteins, among them 17 proteases as differentially expressed between IBS, UC and HC. Of those the four proteases elastase 3a, chymotrypsin C, proteasome subunit type beta-2 and an unspecified isoform of complement C3 were significantly more abundant in IBS compared to HC and UC supernatants. Of eight proteases, which were upregulated in IBS, the combination of elastase 3a, cathepsin L and proteasome alpha subunit-4 showed the highest prediction accuracy of 98% to discriminate between IBS and HC groups. Elastase synergistically potentiated the effects of histamine and serotonin–the two other main neuroactive substances in the IBS supernatants. A serine protease inhibitor isolated from the probiotic Bifidobacterium longum NCC2705 (SERPINBL), known to inhibit elastase-like proteases, prevented nerve activation by IBS supernatants. Conclusion Proteases in IBS and UC supernatants were responsible for nerve activation. Our data demonstrate that proteases, particularly those signalling through neuronal PAR1, are biomarker candidates for IBS, and protease profiling may be used to characterise IBS.
Collapse
Affiliation(s)
- Sabine Buhner
- Human Biology, Technische Universität München, Freising, Germany
| | - Hannes Hahne
- Proteomics and Bioanalytics, Technische Universität München, Freising, Germany
| | - Kerstin Hartwig
- Human Biology, Technische Universität München, Freising, Germany
| | - Qin Li
- Human Biology, Technische Universität München, Freising, Germany
- Department of Physiology, Shangdong University, Shangdong, China
| | - Sheila Vignali
- Human Biology, Technische Universität München, Freising, Germany
| | - Daniela Ostertag
- Human Biology, Technische Universität München, Freising, Germany
| | - Chen Meng
- Proteomics and Bioanalytics, Technische Universität München, Freising, Germany
| | | | - Breg Braak
- Department of Gastroenterology and Hepatology, Academic Medical Center, Amsterdam, The Netherlands
| | | | | | - Giovanni Barbara
- Department of Medical and Surgical Sciences, St. Orsola Hospital, Bologna, Italy
| | - Roberto De Giorgio
- Department of Clinical Sciences, Nuovo Arcispedale S. Anna, University of Ferrara, Ferrara, Italy
| | - Ihsan Ekin Demir
- Department of General Surgery, University Hospital Rechts der Isar, Technische Universität München, Germany
| | - Güralp Onur Ceyhan
- Department of General Surgery, University Hospital Rechts der Isar, Technische Universität München, Germany
| | | | - Guy Boeckxstaens
- Translational Research Centre for Gastrointestinal Disorders, University Hospital Gasthuisberg, Catholic University of Leuven, Leuven, Belgium
| | - Dirk Haller
- Nutrition and Immunology, Technische Universität München, Freising, Germany
| | - Bernhard Kuster
- Proteomics and Bioanalytics, Technische Universität München, Freising, Germany
| | - Michael Schemann
- Human Biology, Technische Universität München, Freising, Germany
- * E-mail:
| |
Collapse
|
46
|
Omics of bifidobacteria: research and insights into their health-promoting activities. Biochem J 2017; 474:4137-4152. [PMID: 29212851 DOI: 10.1042/bcj20160756] [Citation(s) in RCA: 89] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Revised: 10/10/2017] [Accepted: 11/06/2017] [Indexed: 12/17/2022]
Abstract
Members of the genus Bifidobacterium include gut commensals that are particularly abundant among the microbial communities residing in the gut of healthy breast-fed infants, where their presence has been linked to many beneficial host effects. Next-generation DNA sequencing and comparative and functional genome methodologies have been shown to be particularly useful in exploring the diversity of this genus. These combined approaches have allowed the identification of genetic features related to bifidobacterial establishment in the gut, involving host-microbe as well as microbe-microbe interactions. Among these, proteinaceous structures, which protrude from the bacterial surface, i.e. pili or fimbriae, and exopolysaccharidic cell surface layers or capsules represent crucial features that assist in their colonization and persistence in the gut. As bifidobacteria are colonizers of the large intestine, they have to be able to cope with various sources of osmotic, oxidative, bile and acid stress during their transit across the gastric barrier and the small intestine. Bifidobacterial genomes thus encode various survival mechanisms, such as molecular chaperones and efflux pumps, to overcome such challenges. Bifidobacteria represent part of an anaerobic gut community, and feed on nondigestible carbohydrates through a specialized fermentative metabolic pathway, which in turn produces growth substrates for other members of the gut community. Conversely, bifidobacteria may also be dependent on other (bifido)bacteria to access host- and diet-derived glycans, and these complex co-operative interactions, based on resource sharing and cross-feeding strategies, represent powerful driving forces that shape gut microbiota composition.
Collapse
|
47
|
Ruiz L, Delgado S, Ruas-Madiedo P, Sánchez B, Margolles A. Bifidobacteria and Their Molecular Communication with the Immune System. Front Microbiol 2017; 8:2345. [PMID: 29255450 PMCID: PMC5722804 DOI: 10.3389/fmicb.2017.02345] [Citation(s) in RCA: 215] [Impact Index Per Article: 26.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Accepted: 11/15/2017] [Indexed: 12/16/2022] Open
Abstract
Bifidobacterium represents a genus within the phylum Actinobacteria which is one of the major phyla in the healthy intestinal tract of humans. Bifidobacterium is one of the most abundant genera in adults, but its predominance is even more pronounced in infants, especially during lactation, when they can constitute the majority of the total bacterial population. They are one of the pioneering colonizers of the early gut microbiota, and they are known to play important roles in the metabolism of dietary components, otherwise indigestible in the upper parts of the intestine, and in the maturation of the immune system. Bifidobacteria have been shown to interact with human immune cells and to modulate specific pathways, involving innate and adaptive immune processes. In this mini-review, we provide an overview of the current knowledge on the immunomodulatory properties of bifidobacteria and the mechanisms and molecular players underlying these processes, focusing on the corresponding implications for human health. We deal with in vitro models suitable for studying strain-specific immunomodulatory activities. These include peripheral blood mononuclear cells and T cell-mediated immune responses, both effector and regulatory cell responses, as well as the modulation of the phenotype of dendritic cells, among others. Furthermore, preclinical studies, mainly germ-free, gnotobiotic, and conventional murine models, and human clinical trials, are also discussed. Finally, we highlight evidence supporting the immunomodulatory effects of bifidobacterial molecules (proteins and peptides, exopolysaccharides, metabolites, and DNA), as well as the role of bifidobacterial metabolism in maintaining immune homeostasis through cross-feeding mechanisms.
Collapse
Affiliation(s)
- Lorena Ruiz
- Dairy Research Institute, Spanish National Research Council (Instituto de Productos Lácteos de Asturias - CSIC), Villaviciosa, Spain
| | - Susana Delgado
- Dairy Research Institute, Spanish National Research Council (Instituto de Productos Lácteos de Asturias - CSIC), Villaviciosa, Spain
| | - Patricia Ruas-Madiedo
- Dairy Research Institute, Spanish National Research Council (Instituto de Productos Lácteos de Asturias - CSIC), Villaviciosa, Spain
| | - Borja Sánchez
- Dairy Research Institute, Spanish National Research Council (Instituto de Productos Lácteos de Asturias - CSIC), Villaviciosa, Spain
| | - Abelardo Margolles
- Dairy Research Institute, Spanish National Research Council (Instituto de Productos Lácteos de Asturias - CSIC), Villaviciosa, Spain
| |
Collapse
|
48
|
Milani C, Duranti S, Bottacini F, Casey E, Turroni F, Mahony J, Belzer C, Delgado Palacio S, Arboleya Montes S, Mancabelli L, Lugli GA, Rodriguez JM, Bode L, de Vos W, Gueimonde M, Margolles A, van Sinderen D, Ventura M. The First Microbial Colonizers of the Human Gut: Composition, Activities, and Health Implications of the Infant Gut Microbiota. Microbiol Mol Biol Rev 2017; 81:e00036-17. [PMID: 29118049 PMCID: PMC5706746 DOI: 10.1128/mmbr.00036-17] [Citation(s) in RCA: 1121] [Impact Index Per Article: 140.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The human gut microbiota is engaged in multiple interactions affecting host health during the host's entire life span. Microbes colonize the neonatal gut immediately following birth. The establishment and interactive development of this early gut microbiota are believed to be (at least partially) driven and modulated by specific compounds present in human milk. It has been shown that certain genomes of infant gut commensals, in particular those of bifidobacterial species, are genetically adapted to utilize specific glycans of this human secretory fluid, thus representing a very intriguing example of host-microbe coevolution, where both partners are believed to benefit. In recent years, various metagenomic studies have tried to dissect the composition and functionality of the infant gut microbiome and to explore the distribution across the different ecological niches of the infant gut biogeography of the corresponding microbial consortia, including those corresponding to bacteria and viruses, in healthy and ill subjects. Such analyses have linked certain features of the microbiota/microbiome, such as reduced diversity or aberrant composition, to intestinal illnesses in infants or disease states that are manifested at later stages of life, including asthma, inflammatory bowel disease, and metabolic disorders. Thus, a growing number of studies have reported on how the early human gut microbiota composition/development may affect risk factors related to adult health conditions. This concept has fueled the development of strategies to shape the infant microbiota composition based on various functional food products. In this review, we describe the infant microbiota, the mechanisms that drive its establishment and composition, and how microbial consortia may be molded by natural or artificial interventions. Finally, we discuss the relevance of key microbial players of the infant gut microbiota, in particular bifidobacteria, with respect to their role in health and disease.
Collapse
Affiliation(s)
- Christian Milani
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parma, Italy
| | - Sabrina Duranti
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parma, Italy
| | - Francesca Bottacini
- APC Microbiome Institute and School of Microbiology, National University of Ireland, Cork, Ireland
| | - Eoghan Casey
- APC Microbiome Institute and School of Microbiology, National University of Ireland, Cork, Ireland
| | - Francesca Turroni
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parma, Italy
- Microbiome Research Hub, University of Parma, Parma, Italy
| | - Jennifer Mahony
- APC Microbiome Institute and School of Microbiology, National University of Ireland, Cork, Ireland
| | - Clara Belzer
- Laboratory of Microbiology, Wageningen University, Wageningen, The Netherlands
| | - Susana Delgado Palacio
- Departamento de Microbiologia y Bioquimica de Productos Lacteos, IPLA-CSIC, Villaviciosa, Asturias, Spain
| | - Silvia Arboleya Montes
- Departamento de Microbiologia y Bioquimica de Productos Lacteos, IPLA-CSIC, Villaviciosa, Asturias, Spain
| | - Leonardo Mancabelli
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parma, Italy
| | - Gabriele Andrea Lugli
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parma, Italy
| | - Juan Miguel Rodriguez
- Department of Nutrition, Food Science and Food Technology, Complutense University of Madrid, Madrid, Spain
| | - Lars Bode
- Department of Pediatrics and Larsson-Rosenquist Foundation Mother-Milk-Infant Center of Research Excellence, University of California-San Diego, La Jolla, California, USA
| | - Willem de Vos
- Laboratory of Microbiology, Wageningen University, Wageningen, The Netherlands
- Department of Bacteriology & Immunology, RPU Immunobiology, University of Helsinki, Helsinki, Finland
| | - Miguel Gueimonde
- Departamento de Microbiologia y Bioquimica de Productos Lacteos, IPLA-CSIC, Villaviciosa, Asturias, Spain
| | - Abelardo Margolles
- Departamento de Microbiologia y Bioquimica de Productos Lacteos, IPLA-CSIC, Villaviciosa, Asturias, Spain
| | - Douwe van Sinderen
- APC Microbiome Institute and School of Microbiology, National University of Ireland, Cork, Ireland
| | - Marco Ventura
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parma, Italy
- Microbiome Research Hub, University of Parma, Parma, Italy
| |
Collapse
|
49
|
Lebeer S, Bron PA, Marco ML, Van Pijkeren JP, O'Connell Motherway M, Hill C, Pot B, Roos S, Klaenhammer T. Identification of probiotic effector molecules: present state and future perspectives. Curr Opin Biotechnol 2017; 49:217-223. [PMID: 29153882 DOI: 10.1016/j.copbio.2017.10.007] [Citation(s) in RCA: 174] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Revised: 10/24/2017] [Accepted: 10/31/2017] [Indexed: 01/01/2023]
Abstract
Comprehension of underlying mechanisms of probiotic action will support rationale selection of probiotic strains and targeted clinical study design with a higher likelihood of success. This will consequently contribute to better substantiation of health claims. Here, we aim to provide a perspective from a microbiology point of view that such comprehensive understanding is not straightforward. We show examples of well-documented probiotic effector molecules in Lactobacillus and Bifidobacterium strains, including surface-located molecules such as specific pili, S-layer proteins, exopolysaccharides, muropeptides, as well as more widely produced metabolites such as tryptophan-related and histamine-related metabolites, CpG-rich DNA, and various enzymes such as lactase and bile salt hydrolases. We also present recent advances in genetic tool development, microbiome analyses and model systems, as well as perspectives on how the field could further progress. This opinion is based on a discussion group organized at the annual meeting of the International Scientific Association on Probiotics and Prebiotics (ISAPP) in June 2017.
Collapse
Affiliation(s)
- Sarah Lebeer
- University of Antwerp, Department of Bioscience Engineering, Groenenborgerlaan 171, 2020 Antwerp, Belgium.
| | | | - Maria L Marco
- Department of Food Science & Technology, University of California, Davis, USA
| | | | - Mary O'Connell Motherway
- School of Microbiology and APC Microbiome Institute, National University of Ireland, Western Road, Cork, Ireland
| | - Colin Hill
- School of Microbiology and APC Microbiome Institute, National University of Ireland, Western Road, Cork, Ireland
| | - Bruno Pot
- Yakult R&D, Europe, Almere, The Netherlands; Vrije Universiteit Brussels, Belgium
| | - Stefan Roos
- Swedish University of Agricultural Sciences & BioGaia AB, Sweden
| | - Todd Klaenhammer
- Department of Food, Bioprocessing & Nutrition Sciences, North Carolina State University, Raleigh, USA
| |
Collapse
|
50
|
Engevik MA, Versalovic J. Biochemical Features of Beneficial Microbes: Foundations for Therapeutic Microbiology. Microbiol Spectr 2017; 5:10.1128/microbiolspec.BAD-0012-2016. [PMID: 28984235 PMCID: PMC5873327 DOI: 10.1128/microbiolspec.bad-0012-2016] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Indexed: 12/15/2022] Open
Abstract
Commensal and beneficial microbes secrete myriad products which target the mammalian host and other microbes. These secreted substances aid in bacterial niche development, and select compounds beneficially modulate the host and promote health. Microbes produce unique compounds which can serve as signaling factors to the host, such as biogenic amine neuromodulators, or quorum-sensing molecules to facilitate inter-bacterial communication. Bacterial metabolites can also participate in functional enhancement of host metabolic capabilities, immunoregulation, and improvement of intestinal barrier function. Secreted products such as lactic acid, hydrogen peroxide, bacteriocins, and bacteriocin-like substances can also target the microbiome. Microbes differ greatly in their metabolic potential and subsequent host effects. As a result, knowledge about microbial metabolites will facilitate selection of next-generation probiotics and therapeutic compounds derived from the mammalian microbiome. In this article we describe prominent examples of microbial metabolites and their effects on microbial communities and the mammalian host.
Collapse
Affiliation(s)
- Melinda A Engevik
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX 77030 and Department of Pathology, Texas Children's Hospital, Houston, TX 77030
| | - James Versalovic
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX 77030 and Department of Pathology, Texas Children's Hospital, Houston, TX 77030
| |
Collapse
|