1
|
Lee Y, Koh HG, Kim KH, Jin YS, Sung BH, Kim J. Enhancing the persistence of engineered biotherapeutics in the gut: Adhesion, glycan metabolism, and environmental resistance. Adv Drug Deliv Rev 2025; 221:115591. [PMID: 40250567 DOI: 10.1016/j.addr.2025.115591] [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: 12/30/2024] [Revised: 03/20/2025] [Accepted: 04/14/2025] [Indexed: 04/20/2025]
Abstract
Engineered live biotherapeutic products (eLBPs) are receiving increasing attention as next-generation therapeutics to treat a variety of diseases with high specificity and effectiveness. Despite their potential, eLBPs face challenges, such as limited colonization, competition with native microbiota, nutrient depletion, and susceptibility to gastrointestinal stresses, which ultimately reduce their persistence in the gut and hinder their therapeutic efficacy. This review examines the key strategies to enhance the persistence and activity of eLBPs in the gut environment. First, methods to strengthen the adhesion capacity of eLBPs are discussed, including genetic engineering to express adhesins and chemical surface modifications to improve their binding to mucus and epithelial cells. Second, strategies to improve the ability of eLBPs to efficiently use mucin-derived sugars, which are continuously secreted by intestinal epithelial cells, were highlighted. These strategies involve the introduction and optimization of glycan-degrading enzymes and metabolic pathways for key mucin sugars, such as N-acetylglucosamine, galactose, and sialic acid, to support sustained energy production and enhance gut colonization. Third, strategies to improve the resistance of eLBPs against environmental stress are discussed, including genetic modifications to stabilize cell membranes, enhancement of ion pump activity, overexpression of stress-response proteins, and encapsulation techniques to provide protection. The implementation of these strategies can address challenges related to gut colonization by eLBPs, thereby enhancing their metabolic activity and enabling sustained and efficient secretion of therapeutic molecules. This review offers a comprehensive framework for developing and optimizing eLBPs, paving the way for their successful clinical application with enhanced effectiveness in treating gastrointestinal and systemic diseases.
Collapse
Affiliation(s)
- Yujin Lee
- Institute of Food Industrialization, Institutes of Green Bioscience and Technology, Seoul National University, Gangwon-do 25354, Republic of Korea; Graduate School of International Agricultural Technology, Seoul National University, Pyeongchang-gun 25354 Gangwon-do, Republic of Korea
| | - Hyun Gi Koh
- Department of Biological and Chemical Engineering, Hongik University, Sejong 30016, Republic of Korea
| | - Kyoung Heon Kim
- Department of Biotechnology, Graduate School, Korea University, Seoul 02841, Republic of Korea
| | - Yong-Su Jin
- Department of Food Science and Human Nutrition, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA; Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Bong Hyun Sung
- Synthetic Biology and Bioengineering Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Republic of Korea; Department of Biosystems and Bioengineering, KRIBB School of Biotechnology, Korea University of Science and Technology (UST), Daejeon 34113, Republic of Korea
| | - Jungyeon Kim
- Institute of Food Industrialization, Institutes of Green Bioscience and Technology, Seoul National University, Gangwon-do 25354, Republic of Korea; Graduate School of International Agricultural Technology, Seoul National University, Pyeongchang-gun 25354 Gangwon-do, Republic of Korea.
| |
Collapse
|
2
|
Taşkoparan Ş, Altınay C, Barbaros Özer H. Recent updates of probiotic dairy-based beverages. Food Funct 2025; 16:1656-1669. [PMID: 39962909 DOI: 10.1039/d4fo06322h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/22/2025]
Abstract
There is a rapid paradigm shift in the food consumption habits of consumers globally. The interest in healthier, safer, minimally processed and nature-identical foods is the driving force of this paradigm shift. Although the roots of this consumer trend go back further, especially the Covid-19 pandemic has contributed to the acceleration of this process. The effects of probiotics on human health have been known for many years. The commercial success of some probiotic microorganism strains, supported by clinical studies, is also evident. Probiotic microorganisms can be found in commercial products in a wide range of forms including powder, tablets or incorporated into liquid or solid food matrices. Milk and dairy products are suitable vehicles for the delivery of probiotics into the human body. Apart from well-established dairy-based probiotic foods including yogurt and yogurt-type beverages, in recent years some dairy products supplemented or enhanced with postbiotics and paraprobiotics are gaining popularity. The incorporation of next-generation probiotics in probiotic beverage formulations has also attracted the attention of researchers. The current state-of-the art for the utilization of next-generation probiotics, postbiotics and paraprobiotics in dairy-based probiotic beverages is the main focus of this review. Conventional milk-, whey- and buttermilk-based probiotic beverages are also covered.
Collapse
Affiliation(s)
- Şevval Taşkoparan
- Ankara University Faculty of Agriculture Department of Dairy Technology, Diskapi, Ankara, Turkey.
| | - Canan Altınay
- Ankara University Faculty of Agriculture Department of Dairy Technology, Diskapi, Ankara, Turkey.
| | - H Barbaros Özer
- Ankara University Faculty of Agriculture Department of Dairy Technology, Diskapi, Ankara, Turkey.
| |
Collapse
|
3
|
Saleem W, Aslam A, Tariq M, Nauwynck H. Intestinal mucus: the unsung hero in the battle against viral gastroenteritis. Gut Pathog 2025; 17:11. [PMID: 39972475 PMCID: PMC11841282 DOI: 10.1186/s13099-025-00684-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Accepted: 02/04/2025] [Indexed: 02/21/2025] Open
Abstract
Intestinal mucus plays a crucial role in defending against enteric infections by protecting the vulnerable intestinal epithelial cells both physically and through its various constituents. Despite this, numerous gastroenteritis-causing viruses, such as rotavirus, coronavirus, adenovirus, astrovirus, calicivirus, and enterovirus, continue to pose significant threats to humans and animals. While several studies have examined the interactions between these viruses and intestinal mucus, significant gaps remain in understanding the full protective potential of intestinal mucus against these pathogens. This review aims to elucidate the protective role of intestinal mucus in viral gastroenteritis. It begins with a comprehensive literature overview of (i) intestinal mucus, (ii) enteric viruses of medical and veterinary importance, and (iii) the known interactions between various enteric viruses and intestinal mucus. Following this, a case study is presented to highlight the age-dependent blocking effect of porcine intestinal mucus against transmissible gastroenteritis virus, a porcine coronavirus. Finally, the review discusses future investigation directions to further explore the potential of intestinal mucus as a defense mechanism against viral gastroenteritis to stimulate further research in this dynamic and critical area.
Collapse
Affiliation(s)
- Waqar Saleem
- Laboratory of Virology, Department of Translational Physiology, Infectiology and Public Health, Faculty of Veterinary Medicine, Merelbeke, 9820, Belgium.
| | - Ateeqa Aslam
- Laboratory of Virology, Department of Translational Physiology, Infectiology and Public Health, Faculty of Veterinary Medicine, Merelbeke, 9820, Belgium
| | - Mehlayl Tariq
- Laboratory of Biomedical Chemistry, Hirszfeld Institute of Immunology and Experimental Therapy, Wroclaw, 53-114, Poland
| | - Hans Nauwynck
- Laboratory of Virology, Department of Translational Physiology, Infectiology and Public Health, Faculty of Veterinary Medicine, Merelbeke, 9820, Belgium
| |
Collapse
|
4
|
Hossainpour H, Khazaei S, Mahmoudi H. The Effect of Probiotics and Microbiota on Nervous System and Mental Illnesses. Cent Nerv Syst Agents Med Chem 2025; 25:109-122. [PMID: 39313893 DOI: 10.2174/0118715249315760240905043415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 07/08/2024] [Accepted: 07/29/2024] [Indexed: 09/25/2024]
Abstract
The microbiota that inhabits the gastrointestinal tract has been linked to various gastrointestinal and non-gastrointestinal disorders. Scientists have been studying how the bacteria in our intestines have an effect on our brain and nervous system. This connection is called the "microbiota-gut-brain axis". Given the capacity of probiotics, which are live non-pathogenic microorganisms, to reinstate the normal microbial population within the host and confer advantages, their potential impact has been subjected to scrutiny with regard to neurological and mental conditions. Material sourced for this review included peer-reviewed literature annotated in the PubMed, Web of Science, Scopus, and Google Scholar databases. The result has indicated the integration of probiotics into a child's diet to enhance the neuro-behavioral symptoms. Notwithstanding this, the current data set has been found to be insufficient and inconclusive. The potential utility of probiotics for the prevention or treatment of neurologic and mental disorders has become a subject of substantial interest.
Collapse
Affiliation(s)
- Hadi Hossainpour
- Department of Microbiology, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
- Department of Microbiology, School of Medicine, Student Research Committee, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Salman Khazaei
- Department of Epidemiology, School of Public Health, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Hassan Mahmoudi
- Department of Microbiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
- Department of Microbiology, Nahavand School of Allied Medical Sciences, Hamadan University of Medical Sciences, Hamadan, Iran
| |
Collapse
|
5
|
Phùng TTT, Dupont S, Beney L, Moundanga S, Denimal E, Hồ PH, Karbowiak T. Ex-vivo investigation of probiotic bacterial adhesion to the intestinal mucus. Heliyon 2024; 10:e36339. [PMID: 39253206 PMCID: PMC11382035 DOI: 10.1016/j.heliyon.2024.e36339] [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: 02/19/2024] [Revised: 08/09/2024] [Accepted: 08/13/2024] [Indexed: 09/11/2024] Open
Abstract
Recent research has promoted considerable interest in the potential health benefits of the new generation of probiotics. Despite the abundance of probiotic supplements, their adhesion and thereby colonization in the intestinal tract of the host, a determining factor of probiotic efficacy, remains questionable. Indeed, the gastrointestinal tract, a multi-component and complex system, obscures the comprehensive understanding of the probiotic adhesion mechanism. This study aimed to investigate the adhesion capacity of probiotic bacteria using two ex-vivo approaches that were specifically developed to investigate the bacteria-mucus agglomeration and the viable adhesion to intestinal mucus. Five probiotic bacterial strains including Escherichia coli, Lactiplantibacillus plantarum, Faecalibacterium duncaniae, Bifidobacterium longum, and Bifidobacterium longum str. infantis were selected for the investigation. In that context, higher adhesion to mucus was demonstrated by E. coli, L. plantarum, and B. infantis, emphasizing strain-specific differences. While total agglomeration capacity ranged from 8 % to 82 %, actual viable adhesion to mucus remained rather low (0.6 %-2.9 %). SEM images revealed that morphological characteristics, chain and/or cluster forming ability, as well as the presence of surface exopolysaccharides, might have an impact on bacterial adhesion. This study contributes knowledge on probiotic adhesion as well as simple and effective ex-vivo approaches to investigate the bacterial adhesion to the intestinal mucus, which is prerequisite for further colonization in the gut of the host.
Collapse
Affiliation(s)
- Thị-Thanh-Trúc Phùng
- Université Bourgogne Franche-Comté, Institut Agro, Université de Bourgogne, INRAe, UMR PAM 1517, 1 Esplanade Erasme, 21000, Dijon, France
| | - Sébastien Dupont
- Université Bourgogne Franche-Comté, Institut Agro, Université de Bourgogne, INRAe, UMR PAM 1517, 1 Esplanade Erasme, 21000, Dijon, France
| | - Laurent Beney
- Université Bourgogne Franche-Comté, Institut Agro, Université de Bourgogne, INRAe, UMR PAM 1517, 1 Esplanade Erasme, 21000, Dijon, France
| | - Sylvie Moundanga
- Université Bourgogne Franche-Comté, Institut Agro, Université de Bourgogne, INRAe, UMR PAM 1517, 1 Esplanade Erasme, 21000, Dijon, France
| | - Emmanuel Denimal
- Université Bourgogne Franche-Comté, Institut Agro, Université de Bourgogne, INRAe, UMR PAM 1517, 1 Esplanade Erasme, 21000, Dijon, France
| | - Phú-Hà Hồ
- Hanoi University of Science and Technology, School of Chemistry and Life Science, 1 Dai Co Viet Road, Hanoi, Viet Nam
| | - Thomas Karbowiak
- Université Bourgogne Franche-Comté, Institut Agro, Université de Bourgogne, INRAe, UMR PAM 1517, 1 Esplanade Erasme, 21000, Dijon, France
| |
Collapse
|
6
|
Weibel N, Curcio M, Schreiber A, Arriaga G, Mausy M, Mehdy J, Brüllmann L, Meyer A, Roth L, Flury T, Pecina V, Starlinger K, Dernič J, Jungfer K, Ackle F, Earp J, Hausmann M, Jinek M, Rogler G, Antunes Westmann C. Engineering a Novel Probiotic Toolkit in Escherichia coli Nissle 1917 for Sensing and Mitigating Gut Inflammatory Diseases. ACS Synth Biol 2024; 13:2376-2390. [PMID: 39115381 PMCID: PMC11334186 DOI: 10.1021/acssynbio.4c00036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 06/13/2024] [Accepted: 07/25/2024] [Indexed: 08/17/2024]
Abstract
Inflammatory bowel disease (IBD) is characterized by chronic intestinal inflammation with no cure and limited treatment options that often have systemic side effects. In this study, we developed a target-specific system to potentially treat IBD by engineering the probiotic bacterium Escherichia coli Nissle 1917 (EcN). Our modular system comprises three components: a transcription factor-based sensor (NorR) capable of detecting the inflammation biomarker nitric oxide (NO), a type 1 hemolysin secretion system, and a therapeutic cargo consisting of a library of humanized anti-TNFα nanobodies. Despite a reduction in sensitivity, our system demonstrated a concentration-dependent response to NO, successfully secreting functional nanobodies with binding affinities comparable to the commonly used drug Adalimumab, as confirmed by enzyme-linked immunosorbent assay and in vitro assays. This newly validated nanobody library expands EcN therapeutic capabilities. The adopted secretion system, also characterized for the first time in EcN, can be further adapted as a platform for screening and purifying proteins of interest. Additionally, we provided a mathematical framework to assess critical parameters in engineering probiotic systems, including the production and diffusion of relevant molecules, bacterial colonization rates, and particle interactions. This integrated approach expands the synthetic biology toolbox for EcN-based therapies, providing novel parts, circuits, and a model for tunable responses at inflammatory hotspots.
Collapse
Affiliation(s)
- Nathalie Weibel
- University
of Zürich, Campus Irchel Winterthurerstrasse 190, 8057 Zürich, Switzerland
| | - Martina Curcio
- University
of Zürich, Campus Irchel Winterthurerstrasse 190, 8057 Zürich, Switzerland
| | - Atilla Schreiber
- University
of Zürich, Campus Irchel Winterthurerstrasse 190, 8057 Zürich, Switzerland
| | - Gabriel Arriaga
- University
of Zürich, Campus Irchel Winterthurerstrasse 190, 8057 Zürich, Switzerland
| | - Marine Mausy
- University
of Zürich, Campus Irchel Winterthurerstrasse 190, 8057 Zürich, Switzerland
| | - Jana Mehdy
- University
of Zürich, Campus Irchel Winterthurerstrasse 190, 8057 Zürich, Switzerland
| | - Lea Brüllmann
- University
of Zürich, Campus Irchel Winterthurerstrasse 190, 8057 Zürich, Switzerland
| | - Andreas Meyer
- University
of Zürich, Campus Irchel Winterthurerstrasse 190, 8057 Zürich, Switzerland
| | - Len Roth
- University
of Zürich, Campus Irchel Winterthurerstrasse 190, 8057 Zürich, Switzerland
| | - Tamara Flury
- University
of Zürich, Campus Irchel Winterthurerstrasse 190, 8057 Zürich, Switzerland
| | - Valerie Pecina
- University
of Zürich, Campus Irchel Winterthurerstrasse 190, 8057 Zürich, Switzerland
| | - Kim Starlinger
- University
of Zürich, Campus Irchel Winterthurerstrasse 190, 8057 Zürich, Switzerland
| | - Jan Dernič
- Institute
of Pharmacology and Toxicology, University
of Zürich, Winterthurerstrasse
190, CH-8057 Zürich, Switzerland
| | - Kenny Jungfer
- Department
of Biochemistry, University of Zürich, Winterthurerstrasse 190, CH-8057 Zürich, Switzerland
| | - Fabian Ackle
- Institute
of Medical Microbiology, University of Zürich, Gloriastrasse 28/30, CH-8006 Zürich, Switzerland
| | - Jennifer Earp
- Institute
of Medical Microbiology, University of Zürich, Gloriastrasse 28/30, CH-8006 Zürich, Switzerland
| | - Martin Hausmann
- Department
of Gastroenterology and Hepatology, University
Hospital Zürich and Zürich University, Rämistrasse 100, 8091 Zurich, Switzerland
| | - Martin Jinek
- Department
of Biochemistry, University of Zürich, Winterthurerstrasse 190, CH-8057 Zürich, Switzerland
| | - Gerhard Rogler
- Department
of Gastroenterology and Hepatology, University
Hospital Zürich and Zürich University, Rämistrasse 100, 8091 Zurich, Switzerland
| | - Cauã Antunes Westmann
- Department
of Evolutionary Biology and Environmental Studies, University of Zürich, Winterthurerstrasse 190, CH-8057 Zürich, Switzerland
- Swiss
Institute of Bioinformatics, Quartier Sorge-Batiment Genopode, 1015 Lausanne, Switzerland
| |
Collapse
|
7
|
Ermann Lundberg L, Pallabi Mishra P, Liu P, Forsberg MM, Sverremark-Ekström E, Grompone G, Håkansson S, Linninge C, Roos S. Bifidobacterium longum subsp. longum BG-L47 boosts growth and activity of Limosilactobacillus reuteri DSM 17938 and its extracellular membrane vesicles. Appl Environ Microbiol 2024; 90:e0024724. [PMID: 38888338 PMCID: PMC11267924 DOI: 10.1128/aem.00247-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Accepted: 05/14/2024] [Indexed: 06/20/2024] Open
Abstract
The aim of this study was to identify a Bifidobacterium strain that improves the performance of Limosilactobacillus reuteri DSM 17938. Initial tests showed that Bifidobacterium longum subsp. longum strains boosted the growth of DSM 17938 during in vivo-like conditions. Further characterization revealed that one of the strains, BG-L47, had better bile and acid tolerance compared to BG-L48, as well as mucus adhesion compared to both BG-L48 and the control strain BB536. BG-L47 also had the capacity to metabolize a broad range of carbohydrates and sugar alcohols. Mapping of glycoside hydrolase (GH) genes of BG-L47 and BB536 revealed many GHs associated with plant-fiber utilization. However, BG-L47 had a broader phenotypic fiber utilization capacity. In addition, B. longum subsp. longum cells boosted the bioactivity of extracellular membrane vesicles (MV) produced by L. reuteri DSM 17938 during co-cultivation. Secreted 5'-nucleotidase (5'NT), an enzyme that converts AMP into the signal molecule adenosine, was increased in MV boosted by BG-L47. The MV exerted an improved antagonistic effect on the pain receptor transient receptor potential vanilloid 1 (TRPV1) and increased the expression of the immune development markers IL-6 and IL-1ß in a peripheral blood mononuclear cell (PBMC) model. Finally, the safety of BG-L47 was evaluated both by genome safety assessment and in a human safety study. Microbiota analysis showed that the treatment did not induce significant changes in the composition. In conclusion, B. longum subsp. longum BG-L47 has favorable physiological properties, can boost the in vitro activity of L. reuteri DSM 17938, and is safe for consumption, making it a candidate for further evaluation in probiotic studies. IMPORTANCE By using probiotics that contain a combination of strains with synergistic properties, the likelihood of achieving beneficial interactions with the host can increase. In this study, we first performed a broad screening of Bifidobacterium longum subsp. longum strains in terms of synergistic potential and physiological properties. We identified a superior strain, BG-L47, with favorable characteristics and potential to boost the activity of the known probiotic strain Limosilactobacillus reuteri DSM 17938. Furthermore, we demonstrated that BG-L47 is safe for consumption in a human randomized clinical study and by performing a genome safety assessment. This work illustrates that bacteria-bacteria interactions differ at the strain level and further provides a strategy for finding and selecting companion strains of probiotics.
Collapse
Affiliation(s)
- Ludwig Ermann Lundberg
- Department of Molecular Sciences, Uppsala BioCenter, Swedish University of Agricultural Sciences, Uppsala, Sweden
- BioGaia, Stockholm, Sweden
| | - Punya Pallabi Mishra
- Department of Molecular Sciences, Uppsala BioCenter, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | | | - Manuel Mata Forsberg
- The Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - Eva Sverremark-Ekström
- The Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | | | - Sebastian Håkansson
- BioGaia, Stockholm, Sweden
- Division of Applied Microbiology, Department of Chemistry, Lund University, Lund, Sweden
| | - Caroline Linninge
- BioGaia, Stockholm, Sweden
- Department of Food Technology, Engineering and Nutrition, Lund University, Lund, Sweden
| | - Stefan Roos
- Department of Molecular Sciences, Uppsala BioCenter, Swedish University of Agricultural Sciences, Uppsala, Sweden
- BioGaia, Stockholm, Sweden
| |
Collapse
|
8
|
Cho YS, Han K, Xu J, Moon JJ. Novel strategies for modulating the gut microbiome for cancer therapy. Adv Drug Deliv Rev 2024; 210:115332. [PMID: 38759702 PMCID: PMC11268941 DOI: 10.1016/j.addr.2024.115332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 05/08/2024] [Accepted: 05/13/2024] [Indexed: 05/19/2024]
Abstract
Recent advancements in genomics, transcriptomics, and metabolomics have significantly advanced our understanding of the human gut microbiome and its impact on the efficacy and toxicity of anti-cancer therapeutics, including chemotherapy, immunotherapy, and radiotherapy. In particular, prebiotics, probiotics, and postbiotics are recognized for their unique properties in modulating the gut microbiota, maintaining the intestinal barrier, and regulating immune cells, thus emerging as new cancer treatment modalities. However, clinical translation of microbiome-based therapy is still in its early stages, facing challenges to overcome physicochemical and biological barriers of the gastrointestinal tract, enhance target-specific delivery, and improve drug bioavailability. This review aims to highlight the impact of prebiotics, probiotics, and postbiotics on the gut microbiome and their efficacy as cancer treatment modalities. Additionally, we summarize recent innovative engineering strategies designed to overcome challenges associated with oral administration of anti-cancer treatments. Moreover, we will explore the potential benefits of engineered gut microbiome-modulating approaches in ameliorating the side effects of immunotherapy and chemotherapy.
Collapse
Affiliation(s)
- Young Seok Cho
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, USA; Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Kai Han
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 21009, China; Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 21009, China
| | - Jin Xu
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, USA; Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - James J Moon
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, USA; Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA; Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA; Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109, USA.
| |
Collapse
|
9
|
Oviedo-León JF, Cornejo-Mazón M, Ortiz-Hernández R, Torres-Ramírez N, Hernández-Sánchez H, Castro-Rodríguez DC. Exploration adhesion properties of Liquorilactobacillus and Lentilactobacillus isolated from two different sources of tepache kefir grains. PLoS One 2024; 19:e0297900. [PMID: 38324577 PMCID: PMC10849267 DOI: 10.1371/journal.pone.0297900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 01/15/2024] [Indexed: 02/09/2024] Open
Abstract
Due to the distinctive characteristics of probiotics, it is essential to pinpoint strains originating from diverse sources that prove efficacious in addressing a range of pathologies linked to dysfunction of the intestinal barrier. Nine strains of lactic acid bacteria were isolated from two different sources of tepache kefir grains (KAS2, KAS3, KAS4, KAS7, KAL4, KBS2, KBS3, KBL1 and KBL3), and were categorized to the genus Lacticaseibacillus, Liquorilactobacillus, and Lentilactobacillus by 16S rRNA gene. Kinetic behaviors of these strains were evaluated in MRS medium, and their probiotic potential was performed: resistance to low pH, tolerance to pepsin, pancreatin, bile salts, antibiotic resistance, hemolytic activity, and adhesion ability. KAS7 strain presented a higher growth rate (0.50 h-1) compared with KAS2 strain, who presented a lower growth rate (0.29 h-1). KBS2 strain was the only strain that survived the in vitro stomach simulation conditions (29.3%). Strain KBL1 demonstrated significantly higher viability (90.6%) in the in vitro intestine simulation conditions. Strain KAS2 demonstrated strong hydrophilic character with chloroform (85.6%) and xylol (57.6%) and a higher percentage of mucin adhesion (87.1%). However, strains KBS2 (84.8%) and KBL3 (89.5%) showed the highest autoaggregation values. In terms of adhesion to the intestinal epithelium in rats, strains KAS2, KAS3 and KAS4 showed values above 80%. The growth of the strains KAS2, KAS3, KAS4, KBS2, and KBL3 was inhibited by cefuroxime, cefotaxime, tetracycline, ampicillin, erythromycin, and cephalothin. Strains KBS2 (41.9% and 33.5%) and KBL3 (42.5% and 32.8%) had the highest co-aggregation values with S. aureus and E. coli. The results obtained in this study indicate that lactic acid bacteria isolated from tepache can be considered as candidates for potentially probiotic bacteria, laying the foundations to evaluate their probiotic functionality in vivo and thus to be used in the formulation of functional foods.
Collapse
Affiliation(s)
- Julián Fernando Oviedo-León
- Departamento de Ingeniería Bioquímica, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Unidad Profesional Adolfo López Mateos, Mexico City, Mexico
| | - Maribel Cornejo-Mazón
- Departamento Biofísica, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Carpio y Plan de Ayala, Mexico City, Mexico
| | - Rosario Ortiz-Hernández
- Departamento de Biología Celular, Facultad de Ciencias, Universidad Nacional Autónoma de Mexico (UNAM), Mexico City, Mexico
| | - Nayeli Torres-Ramírez
- Departamento de Biología Celular, Facultad de Ciencias, Universidad Nacional Autónoma de Mexico (UNAM), Mexico City, Mexico
| | - Humberto Hernández-Sánchez
- Departamento de Ingeniería Bioquímica, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Unidad Profesional Adolfo López Mateos, Mexico City, Mexico
| | - Diana C. Castro-Rodríguez
- Investigadores CONAHCyT, Departamento de Biología de la Reproducción, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| |
Collapse
|
10
|
Guo J, Zhou B, Niu Y, Liu L, Yang L. Engineered probiotics introduced to improve intestinal microecology for the treatment of chronic diseases: present state and perspectives. J Diabetes Metab Disord 2023; 22:1029-1038. [PMID: 37975092 PMCID: PMC10638336 DOI: 10.1007/s40200-023-01279-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 08/05/2023] [Indexed: 11/19/2023]
Abstract
Purpose Correcting intestinal microecological imbalance has become one of the core strategies to treat chronic diseases. Some traditional microecology-based therapies targeting intestine, such as prebiotic therapy, probiotic therapy and fecal microbiota transplantation therapy, have been used in the prevention and treatment of clinical chronic diseases, which still facing low safety and poor controllability problems. The development of synthetic biology technology has promoted the development of intestinal microecology-based therapeutics for chronic diseases, which exhibiting higher robustness and controllability, and become an important part of the next generation of microecological therapy. The purpose of this review is to summarize the application of synthetic biology in intestinal microecology-based therapeutics for chronic diseases. Methods The available literatures were searched to find out experimental studies and relevant review articles on the application of synthetic biology in intestinal microecology-based therapeutics for chronic diseases from year 1990 to 2023. Results Evidence proposed that synthetic biology has been applied in the intestinal microecology-based therapeutics for chronic diseases, covering metabolic diseases (e.g. diabetes, obesity, nonalcoholic fatty liver disease and phenylketonuria), digestive diseases (e.g. inflammatory bowel disease and colorectal cancer), and neurodegenerative diseases (e.g. Alzheimer's disease and Parkinson's disease). Conclusion This review summarizes the application of synthetic biology in intestinal microecology-based therapeutics for major chronic diseases and discusses the opportunities and challenges in the above process, providing clinical possibilities of synthetic biology technology applied in microecological therapies.
Collapse
Affiliation(s)
- Jianquan Guo
- Key Laboratory of Coal Environmental Pathogenicity and Prevention, (Shanxi Medical University), Ministry of Education, Taiyuan, PR China
- School of Public Health, Shanxi Medical University, Taiyuan, 030001 Shanxi PR China
| | - Bangyuan Zhou
- School of Public Health, Shanxi Medical University, Taiyuan, 030001 Shanxi PR China
| | - Yali Niu
- School of Public Health, Shanxi Medical University, Taiyuan, 030001 Shanxi PR China
| | - Liangpo Liu
- School of Public Health, Shanxi Medical University, Taiyuan, 030001 Shanxi PR China
| | - Liyang Yang
- School of Basic Medical Sciences, Shanxi University of Chinese Medicine, 030619 Jinzhong, PR China
| |
Collapse
|
11
|
Wang CM, Fernez MT, Woolston BM, Carrier RL. Native gastrointestinal mucus: Critical features and techniques for studying interactions with drugs, drug carriers, and bacteria. Adv Drug Deliv Rev 2023; 200:114966. [PMID: 37329985 PMCID: PMC11184232 DOI: 10.1016/j.addr.2023.114966] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 06/09/2023] [Accepted: 06/12/2023] [Indexed: 06/19/2023]
Abstract
Gastrointestinal mucus plays essential roles in modulating interactions between intestinal lumen contents, including orally delivered drug carriers and the gut microbiome, and underlying epithelial and immune tissues and cells. This review is focused on the properties of and methods for studying native gastrointestinal mucus and its interactions with intestinal lumen contents, including drug delivery systems, drugs, and bacteria. The properties of gastrointestinal mucus important to consider in its analysis are first presented, followed by a discussion of different experimental setups used to study gastrointestinal mucus. Applications of native intestinal mucus are then described, including experimental methods used to study mucus as a barrier to drug delivery and interactions with intestinal lumen contents that impact barrier properties. Given the significance of the microbiota in health and disease, its impact on drug delivery and drug metabolism, and the use of probiotics and microbe-based delivery systems, analysis of interactions of bacteria with native intestinal mucus is then reviewed. Specifically, bacteria adhesion to, motility within, and degradation of mucus is discussed. Literature noted is focused largely on applications of native intestinal mucus models as opposed to isolated mucins or reconstituted mucin gels.
Collapse
Affiliation(s)
- Chia-Ming Wang
- Department of Bioengineering, Northeastern University, Boston, MA, USA
| | - Matthew T Fernez
- Department of Chemical Engineering, Northeastern University, Boston, MA, USA
| | - Benjamin M Woolston
- Department of Chemical Engineering, Northeastern University, Boston, MA, USA
| | - Rebecca L Carrier
- Department of Bioengineering, Northeastern University, Boston, MA, USA; Department of Chemical Engineering, Northeastern University, Boston, MA, USA; Department of Biology, Northeastern University, Boston, MA, USA.
| |
Collapse
|
12
|
Kiepś J, Juzwa W, Olejnik A, Sip A, Tomaszewska-Gras J, Dembczyński R. The Effects of Cellular Membrane Damage on the Long-Term Storage and Adhesion of Probiotic Bacteria in Caco-2 Cell Line. Nutrients 2023; 15:3484. [PMID: 37571422 PMCID: PMC10421378 DOI: 10.3390/nu15153484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 07/31/2023] [Accepted: 08/05/2023] [Indexed: 08/13/2023] Open
Abstract
Adhesion is one of the main factors responsible for the probiotic properties of bacteria in the human gut. Membrane proteins affected by cellular damage are one of the key aspects determining adhesion. Fluid-bed-dried preparations containing probiotic bacteria were analyzed in terms of their stability (temperature of glass transition) and shelf life in different conditions (modified atmosphere, refrigeration). Imaging flow cytometry was utilized to determine four subpopulations of cells based on their physiological and morphological properties. Lastly, adhesion was measured in bacteria cultured in optimal conditions and treated with heat shock. The results show that the subpopulations with no or low levels of cell membrane damage exhibit the ability to adhere to Caco-2 cells. The temperature of protein denaturation in bacteria was recorded as being between 65 °C and 70 °C. The highest glass transition temperature (Tg) value for hydroxypropyl methylcellulose (used as a coating substance) was measured at 152.6 °C. Drying and coating can be utilized as a sufficient treatment, allowing a long shelf-life (up to 12 months). It is, however, worth noting that technological processing, especially with high temperatures, may decrease the probiotic value of the preparation by damaging the bacterial cells.
Collapse
Affiliation(s)
- Jakub Kiepś
- Department of Biotechnology and Food Microbiology, Poznan University of Life Sciences, 60-627 Poznan, Poland; (W.J.); (A.O.); (A.S.)
| | - Wojciech Juzwa
- Department of Biotechnology and Food Microbiology, Poznan University of Life Sciences, 60-627 Poznan, Poland; (W.J.); (A.O.); (A.S.)
| | - Anna Olejnik
- Department of Biotechnology and Food Microbiology, Poznan University of Life Sciences, 60-627 Poznan, Poland; (W.J.); (A.O.); (A.S.)
| | - Anna Sip
- Department of Biotechnology and Food Microbiology, Poznan University of Life Sciences, 60-627 Poznan, Poland; (W.J.); (A.O.); (A.S.)
| | - Jolanta Tomaszewska-Gras
- Department of Food Safety and Quality Management, Poznan University of Life Sciences, 60-624 Poznan, Poland;
| | - Radosław Dembczyński
- Department of Biotechnology and Food Microbiology, Poznan University of Life Sciences, 60-627 Poznan, Poland; (W.J.); (A.O.); (A.S.)
| |
Collapse
|
13
|
Fan Q, Zeng X, Wu Z, Guo Y, Du Q, Tu M, Pan D. Nanocoating of lactic acid bacteria: properties, protection mechanisms, and future trends. Crit Rev Food Sci Nutr 2023; 64:10148-10163. [PMID: 37318213 DOI: 10.1080/10408398.2023.2220803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Lactic acid bacteria (LAB) is a type of probiotic that may benefit intestinal health. Recent advances in nanoencapsulation provide an effective strategy to protect them from harsh conditions via surface functionalization coating techniques. Herein, the categories and features of applicable encapsulation methods are compared to highlight the significant role of nanoencapsulation. Commonly used food-grade biopolymers (polysaccharides and protein) and nanomaterials (nanocellulose and starch nanoparticles) are summarized along with their characteristics and advances to demonstrate enhanced combination effects in LAB co-encapsulation. Nanocoating for LAB provides an integrity dense or smooth layer attributed to the cross-linking and assembly of the protectant. The synergism of multiple chemical forces allows for the formation of subtle coatings, including electrostatic attractions, hydrophobic interactions, π-π, and metallic bonds. Multilayer shells have stable physical transition properties that could increase the space between the probiotic cells and the outer environment, thus delaying the microcapsules burst time in the gut. Probiotic delivery stability can be promoted by enhancing the thickness of the encapsulated layer and nanoparticle binding. Maintenance of benefits and minimization of nanotoxicity are desirable, and green synthesized nanoparticles are emerging. Future trends include optimized formulation, especially using biocompatible materials, protein or plant-based materials, and material modification.
Collapse
Affiliation(s)
- Qing Fan
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, China
- Key Laboratory of Animal Protein Food Processing Technology of Zhejiang Province, College of Food and Pharmaceutical Sciences, Ningbo University, Ningbo, China
- Zhejiang-Malaysia Joint Research Laboratory for Agricultural Product Processing and Nutrition, Ningbo University, Ningbo, China
| | - Xiaoqun Zeng
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, China
- Key Laboratory of Animal Protein Food Processing Technology of Zhejiang Province, College of Food and Pharmaceutical Sciences, Ningbo University, Ningbo, China
- Zhejiang-Malaysia Joint Research Laboratory for Agricultural Product Processing and Nutrition, Ningbo University, Ningbo, China
| | - Zhen Wu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, China
- Key Laboratory of Animal Protein Food Processing Technology of Zhejiang Province, College of Food and Pharmaceutical Sciences, Ningbo University, Ningbo, China
- Zhejiang-Malaysia Joint Research Laboratory for Agricultural Product Processing and Nutrition, Ningbo University, Ningbo, China
| | - Yuxing Guo
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, China
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, China
| | - Qiwei Du
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, China
- Key Laboratory of Animal Protein Food Processing Technology of Zhejiang Province, College of Food and Pharmaceutical Sciences, Ningbo University, Ningbo, China
- Zhejiang-Malaysia Joint Research Laboratory for Agricultural Product Processing and Nutrition, Ningbo University, Ningbo, China
| | - Maolin Tu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, China
- Key Laboratory of Animal Protein Food Processing Technology of Zhejiang Province, College of Food and Pharmaceutical Sciences, Ningbo University, Ningbo, China
- Zhejiang-Malaysia Joint Research Laboratory for Agricultural Product Processing and Nutrition, Ningbo University, Ningbo, China
| | - Daodong Pan
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, China
- Key Laboratory of Animal Protein Food Processing Technology of Zhejiang Province, College of Food and Pharmaceutical Sciences, Ningbo University, Ningbo, China
- Zhejiang-Malaysia Joint Research Laboratory for Agricultural Product Processing and Nutrition, Ningbo University, Ningbo, China
| |
Collapse
|
14
|
Abstract
Live biotherapeutic products (LBPs), including symbiotic and genetically engineered bacteria, are a promising class of emerging therapeutics that are widely investigated both preclinically and clinically for their oral delivery to the gastrointestinal (GI) tract. One emergent delivery strategy involves the direct functionalization of LBP surfaces through noncovalent or covalent modifications to control LBP interactions with the GI microenvironment, thereby improving their viability, attachment, or therapeutic effect. However, unlike other therapeutic modalities, LBPs are living organisms which present two unique challenges for surface modifications: (1) this approach can directly interfere with key LBP biological processes (e.g., colonization, metabolite secretion) and (2) modification can be variable due to the dynamic nature of LBP surfaces. Collectively, these factors remain uncharacterized as they relate to the oral delivery of LBPs. Herein, we leverage our previously reported surface modification platform, which enables LBP surface-presentation of targeting ligands, to broadly evaluate and characterize surface modifications on LBPs. Specifically, we evaluate how LBP growth affects the dilution of surface-presented targeting ligands and the subsequent loss of specific target attachment over time. Next, we describe key surface modification parameters (e.g., concentration, residence time) that can be optimized to facilitate LBP target attachment. We then characterize how bioconjugation influences the suitability of LBPs for oral delivery by evaluating their growth, viability, storage, toxicity against mammalian cells, and in vivo colonization. Broadly, we describe key parameters that influence the performance of surface modified LBPs and subsequently outline an experimental pipeline for characterizing and evaluating their suitability for oral delivery.
Collapse
Affiliation(s)
- Ava M. Vargason
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Aaron C. Anselmo
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| |
Collapse
|
15
|
Hidalgo VM, Babot JD, Fernández MM, Perez Chaia A, Audisio C, Apella MC. Characterization of lactic acid bacteria isolated from the poultry intestinal environment with anti-Salmonella activity in vitro. Braz J Microbiol 2023; 54:435-447. [PMID: 36333643 PMCID: PMC9944612 DOI: 10.1007/s42770-022-00860-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 10/22/2022] [Indexed: 11/07/2022] Open
Abstract
The purpose of this research was the genotypic identification of lactic acid bacteria (LAB), isolated from the gastrointestinal tract (GIT) of healthy adult birds, and the study of their safety regarding antibiotic resistance, physiological and functional properties involved in the colonization of the GIT of poultry, and Salmonella exclusion, as members of a potential mixed probiotic supplement for poultry. The nucleotidic sequence from Lactobacillus crispatus P1, L. animalis L3, and Enterococcus faecium CRL 1385 (ex-J96) showed 100, 99.8, and 99.3% identity with L. crispatus DSM 20584 T, Ligilactobacillus salivarius ATCC 11741 T, and E. faecium ATCC 19434 T, respectively. These strains showed no resistance to relevant antibiotics usually administered to animals proposed by the European Food Safety Authority. They could endure the detrimental conditions of the gastrointestinal tract (pH 2.6 and oxgall 0.1 and 0.4% w/v). In an ex vivo assay, the LAB showed high adherence to the three sections of the GIT, reaching values higher than 70%. The adhesion to mucus was strain-dependent: L. crispatus CRL 1453 evidenced the highest adhesion (> 19%) while Lig. salivarius subsp. salivarius CRL 1417 and E. faecium CRL 1385 adhered to a lower extent (> 9 and 2%, respectively). Moreover, the LAB elicited remarkable anti-Salmonella activity, taking into account that they could inhibit elevated counts of different Salmonella serovars, especially the host-specific serovars S. Gallinarum and S. Pullorum (up to 8 log CFU/mL decrease in Salmonella counts).
Collapse
Affiliation(s)
- Victor Maximiliano Hidalgo
- Estación Experimental Agroindustrial Obispo Colombres (EEAOC-Tucumán), San Miguel de Tucumán, Av. William Cross 3150, (T4101XAC), Tucumán, Argentina
| | - Jaime Daniel Babot
- Centro de Referencia para Lactobacilos (CERELA-CCT CONICET NOA Sur), San Miguel de Tucumán, Chacabuco 145, (T4000ILC), Tucumán, Argentina.
| | - María Magdalena Fernández
- Centro de Referencia para Lactobacilos (CERELA-CCT CONICET NOA Sur), San Miguel de Tucumán, Chacabuco 145, (T4000ILC), Tucumán, Argentina
| | - Adriana Perez Chaia
- Centro de Referencia para Lactobacilos (CERELA-CCT CONICET NOA Sur), San Miguel de Tucumán, Chacabuco 145, (T4000ILC), Tucumán, Argentina
- Universidad Nacional de Tucumán, Ayacucho 491, (T4000INI) San Miguel de Tucumán, Tucumán, Argentina
| | - Carina Audisio
- Instituto de Investigaciones para la Industria Química (INIQUI-CCT CONICET Salta-Jujuy), Buenos Aires 177, A4402FDC, Salta, Salta, Argentina
| | - María Cristina Apella
- Centro de Referencia para Lactobacilos (CERELA-CCT CONICET NOA Sur), San Miguel de Tucumán, Chacabuco 145, (T4000ILC), Tucumán, Argentina.
- Universidad Nacional de Tucumán, Ayacucho 491, (T4000INI) San Miguel de Tucumán, Tucumán, Argentina.
| |
Collapse
|
16
|
Huang Y, Lin X, Yu S, Chen R, Chen W. Intestinal Engineered Probiotics as Living Therapeutics: Chassis Selection, Colonization Enhancement, Gene Circuit Design, and Biocontainment. ACS Synth Biol 2022; 11:3134-3153. [PMID: 36094344 DOI: 10.1021/acssynbio.2c00314] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Intestinal probiotics are often used for the in situ treatment of diseases, such as metabolic disorders, tumors, and chronic inflammatory infections. Recently, there has been an increased emphasis on intelligent, customized treatments with a focus on long-term efficacy; however, traditional probiotic therapy has not kept up with this trend. The use of synthetic biology to construct gut-engineered probiotics as live therapeutics is a promising avenue in the treatment of specific diseases, such as phenylketonuria and inflammatory bowel disease. These studies generally involve a series of fundamental design issues: choosing an engineered chassis, improving the colonization ability of engineered probiotics, designing functional gene circuits, and ensuring the safety of engineered probiotics. In this review, we summarize the relevant past research, the progress of current research, and discuss the key issues that restrict the widespread application of intestinal engineered probiotic living therapeutics.
Collapse
Affiliation(s)
- Yan Huang
- Team SZU-China at iGEM 2021, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen 518060, China
| | - Xiaojun Lin
- Team SZU-China at iGEM 2021, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen 518060, China
| | - Siyang Yu
- Team SZU-China at iGEM 2021, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen 518060, China
| | - Ruiyue Chen
- Team SZU-China at iGEM 2021, Institute for Advanced Study, Shenzhen University, Shenzhen 518060, China
| | - Weizhao Chen
- Team SZU-China at iGEM 2021, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen 518060, China.,Shenzhen Key Laboratory for Microbial Gene Engineering, Shenzhen University, Shenzhen 518060, China
| |
Collapse
|
17
|
Jiang K, Wen X, Pettersson T, Crouzier T. Engineering Surfaces with Immune Modulating Properties of Mucin Hydrogels. ACS APPLIED MATERIALS & INTERFACES 2022; 14:39727-39735. [PMID: 36000701 PMCID: PMC9460428 DOI: 10.1021/acsami.1c19250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Accepted: 08/05/2022] [Indexed: 06/15/2023]
Abstract
Hydrogels of cross-linked mucin glycoproteins (Muc-gel) have shown strong immune-modulating properties toward macrophages in vitro, which are translated in vivo by the dampening of the foreign body response to implantation in mice. Beyond mucin hydrogels, other biomaterials such as sensors, electrodes, and other long-term implants would also benefit from such immune-modulating properties. In this work, we aimed to transfer the bioactivity observed for three-dimensional Muc-gels to the surface of two model materials by immobilizing mucin into thin films (Muc-film) using covalent layer-by-layer assembly. We tested how the surface immobilization of mucins affects macrophage responses compared to Muc-gels. We showed that Muc-films on soft polyacrylamide gels mimic Muc-gel in their modulation of macrophage responses with activated gene expression of inflammatory cytokines on day 1 and then dampening them on day 3. Also, the markers of polarized macrophages, M1 and M2, were expressed at the same level for macrophages on Muc-film-coated soft polyacrylamide gels and Muc-gel. In contrast, Muc-film-coated hard polystyrene led to a different macrophage response compared to Muc-gel, having no activated expression of inflammatory cytokines and a different M1 marker expression. This suggested that the substrate mechanical properties and mucin molecular configuration determined by substrate-mucin interactions affect mucin immune-modulating properties. We conclude that mucin immune-modulating properties can be transferred to materials by mucin surface immobilization but will be dependent on the substrate chemical and mechanical properties.
Collapse
Affiliation(s)
- Kun Jiang
- Division
of Glycoscience, Department of Chemistry, School of Engineering Sciences
in Chemistry, Biotechnology and Health, KTH, Royal Institute of Technology, AlbaNova University Center, Stockholm 106 91, Sweden
- AIMES
- Center for the Advancement of Integrated Medical and Engineering
Sciences at Karolinska Institutet and KTH
Royal Institute of Technology, Stockholm SE-100 44, Sweden
- Department
of Neuroscience, Karolinska Institutet, Stockholm SE-171 77, Sweden
| | - Xueyu Wen
- Division
of Glycoscience, Department of Chemistry, School of Engineering Sciences
in Chemistry, Biotechnology and Health, KTH, Royal Institute of Technology, AlbaNova University Center, Stockholm 106 91, Sweden
| | - Torbjörn Pettersson
- Division
of Fibre Technology, Department of Fibre and Polymer Technology, School
of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, Stockholm SE-100 44, Sweden
| | - Thomas Crouzier
- Division
of Glycoscience, Department of Chemistry, School of Engineering Sciences
in Chemistry, Biotechnology and Health, KTH, Royal Institute of Technology, AlbaNova University Center, Stockholm 106 91, Sweden
- AIMES
- Center for the Advancement of Integrated Medical and Engineering
Sciences at Karolinska Institutet and KTH
Royal Institute of Technology, Stockholm SE-100 44, Sweden
- Department
of Neuroscience, Karolinska Institutet, Stockholm SE-171 77, Sweden
| |
Collapse
|
18
|
Heavey MK, Durmusoglu D, Crook N, Anselmo AC. Discovery and delivery strategies for engineered live biotherapeutic products. Trends Biotechnol 2022; 40:354-369. [PMID: 34481657 PMCID: PMC8831446 DOI: 10.1016/j.tibtech.2021.08.002] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 08/02/2021] [Accepted: 08/03/2021] [Indexed: 10/20/2022]
Abstract
Genetically engineered microbes that secrete therapeutics, sense and respond to external environments, and/or target specific sites in the gut fall under an emergent class of therapeutics, called live biotherapeutic products (LBPs). As live organisms that require symbiotic host interactions, LBPs offer unique therapeutic opportunities, but also face distinct challenges in the gut microenvironment. In this review, we describe recent approaches (often demonstrated using traditional probiotic microorganisms) to discover LBP chassis and genetic parts utilizing omics-based methods and highlight LBP delivery strategies, with a focus on addressing physiological challenges that LBPs encounter after oral administration. Finally, we share our perspective on the opportunity to apply an integrated approach, wherein discovery and delivery strategies are utilized synergistically, towards tailoring and optimizing LBP efficacy.
Collapse
Affiliation(s)
- Mairead K. Heavey
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Deniz Durmusoglu
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, North Carolina, USA
| | - Nathan Crook
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, NC, USA.
| | - Aaron C. Anselmo
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA,Correspondence: (A.C. Anselmo), (N. Crook)
| |
Collapse
|
19
|
Native and Engineered Probiotics: Promising Agents against Related Systemic and Intestinal Diseases. Int J Mol Sci 2022; 23:ijms23020594. [PMID: 35054790 PMCID: PMC8775704 DOI: 10.3390/ijms23020594] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 12/29/2021] [Accepted: 01/03/2022] [Indexed: 12/12/2022] Open
Abstract
Intestinal homeostasis is a dynamic balance involving the interaction between the host intestinal mucosa, immune barrier, intestinal microecology, nutrients, and metabolites. Once homeostasis is out of balance, it will increase the risk of intestinal diseases and is also closely associated with some systemic diseases. Probiotics (Escherichia coli Nissle 1917, Akkermansia muciniphila, Clostridium butyricum, lactic acid bacteria and Bifidobacterium spp.), maintaining the gut homeostasis through direct interaction with the intestine, can also exist as a specific agent to prevent, alleviate, or cure intestinal-related diseases. With genetic engineering technology advancing, probiotics can also show targeted therapeutic properties. The aims of this review are to summarize the roles of potential native and engineered probiotics in oncology, inflammatory bowel disease, and obesity, discussing the therapeutic applications of these probiotics.
Collapse
|
20
|
Javanshir N, Hosseini GNG, Sadeghi M, Esmaeili R, Satarikia F, Ahmadian G, Allahyari N. Evaluation of the Function of Probiotics, Emphasizing the Role of their Binding to the Intestinal Epithelium in the Stability and their Effects on the Immune System. Biol Proced Online 2021; 23:23. [PMID: 34847891 PMCID: PMC8903605 DOI: 10.1186/s12575-021-00160-w] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 11/04/2021] [Indexed: 12/11/2022] Open
Abstract
Due to the importance of using cost-effective methods for therapeutic purposes, the function of probiotics as safe microorganisms and the study of their relevant functional mechanisms have recently been in the spotlight. Finding the mechanisms of attachment and stability and their beneficial effects on the immune system can be useful in identifying and increasing the therapeutic effects of probiotics. In this review, the functional mechanisms of probiotics were comprehensively investigated. Relevant articles were searched in scientific sources, documents, and databases, including PubMed, NCBI, Bactibace, OptiBac, and Bagel4. The most important functional mechanisms of probiotics and their effects on strengthening the epithelial barrier, competitive inhibition of pathogenic microorganisms, production of antimicrobials, binding and interaction with the host, and regulatory effects on the immune system were discussed. In this regard, the attachment of probiotics to the epithelium is very important because the prerequisite for their proper functioning is to establish a proper connection to the epithelium. Therefore, more attention should be paid to the binding effect of probiotics, including sortase A, a significant factor involved in the expression of sortase-dependent proteins (SDP), on their surface as mediators of intestinal epithelial cell binding. In general, by investigating the functional mechanisms of probiotics, it was concluded that the mechanism by which probiotics regulate the immune system and adhesion capacity can directly and indirectly have preventive and therapeutic effects on a wide range of diseases. However, further study of these mechanisms requires extensive research on various aspects.
Collapse
Affiliation(s)
- Nahid Javanshir
- Department of Industrial and Environmental Biotechnology, National Institute of Genetic Engineering and Biotechnology. (NIGEB), P.O. Box: 14155-6343, Tehran, Iran
| | | | - Mahdieh Sadeghi
- Department of Science, Islamic Azad University - Parand Branch, Parand, Iran
| | | | - Fateme Satarikia
- Department of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Gholamreza Ahmadian
- Department of Industrial and Environmental Biotechnology, National Institute of Genetic Engineering and Biotechnology. (NIGEB), P.O. Box: 14155-6343, Tehran, Iran.
| | - Najaf Allahyari
- Department of Industrial and Environmental Biotechnology, National Institute of Genetic Engineering and Biotechnology. (NIGEB), P.O. Box: 14155-6343, Tehran, Iran.
| |
Collapse
|
21
|
Design and in situ biosynthesis of precision therapies against gastrointestinal pathogens. CURRENT OPINION IN PHYSIOLOGY 2021. [DOI: 10.1016/j.cophys.2021.06.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
22
|
Mejía-Pitta A, Broset E, de la Fuente-Nunez C. Probiotic engineering strategies for the heterologous production of antimicrobial peptides. Adv Drug Deliv Rev 2021; 176:113863. [PMID: 34273423 PMCID: PMC8440409 DOI: 10.1016/j.addr.2021.113863] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 06/10/2021] [Accepted: 07/07/2021] [Indexed: 12/12/2022]
Abstract
Engineered probiotic bacteria represent an innovative approach for treating and detecting a wide range of diseases including those caused by infectious agents. Antimicrobial peptides (AMPs) are promising alternatives to conventional antibiotics for combating antibiotic-resistant infections. These molecules can be delivered orally to the gut by using engineered probiotics, which confer protection against AMP degradation, thus enabling numerous applications including treating drug-resistant enteric pathogens and remodeling the microbiota in real time. Here, we provide an update on the current state of the art on AMP-producing probiotics, discuss methods to enhance gut colonization, and end by outlining future perspectives.
Collapse
Affiliation(s)
- Adriana Mejía-Pitta
- Machine Biology Group, Departments of Psychiatry and Microbiology, Institute for Biomedical Informatics, Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States of America; Departments of Bioengineering and Chemical and Biomolecular Engineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, United States of America; Penn Institute for Computational Science, University of Pennsylvania, Philadelphia, PA, United States of America
| | - Esther Broset
- Machine Biology Group, Departments of Psychiatry and Microbiology, Institute for Biomedical Informatics, Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States of America; Departments of Bioengineering and Chemical and Biomolecular Engineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, United States of America; Penn Institute for Computational Science, University of Pennsylvania, Philadelphia, PA, United States of America
| | - Cesar de la Fuente-Nunez
- Machine Biology Group, Departments of Psychiatry and Microbiology, Institute for Biomedical Informatics, Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States of America; Departments of Bioengineering and Chemical and Biomolecular Engineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, United States of America; Penn Institute for Computational Science, University of Pennsylvania, Philadelphia, PA, United States of America.
| |
Collapse
|
23
|
Caballero AM, Villagrán VAS, Serna AJ, Farrés A. Challenges in the production and use of probiotics as therapeuticals in cancer treatment or prevention. J Ind Microbiol Biotechnol 2021; 48:6356962. [PMID: 34427674 DOI: 10.1093/jimb/kuab052] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 08/03/2021] [Indexed: 01/22/2023]
Abstract
Probiotics were defined as microbial strains that confer health benefits to their consumers. The concept has evolved during the last twenty years, and today metabolites produced by the strains, known as postbiotics, and even dead cells, known as paraprobiotics are closely associated to them. The isolation of commensal strains from human microbiome has led to the development of next generation probiotics. This review aims to present an overview of the developments in the area of cancer prevention and treatment, intimately related to advances in the knowledge of the microbiome role in its genesis and therapy. Strain identification and characterization, production processes, delivery strategies and clinical evaluation are crucial to translate results into the market with solid scientific support. Examples of recent tools in isolation, strain typification, quality control and development of new probiotic strains are described. Probiotics market and regulation were originally developed in the food sector, but these new strategies will impact the pharmaceutical and health sectors, requiring new considerations in regulatory frameworks.
Collapse
Affiliation(s)
- Alejandra Mejía Caballero
- Departamento de Ingeniería Celular y Biocatálisis, Insituto de Biotecnología, Universidad Nacional Autónoma de México, 62210, Cuernavaca, Morelos, México
| | - Vianey Anahi Salas Villagrán
- Departamento de Alimentos y Biotecnología, Facultad de Química, Universidad Nacional Autónoma de México, 04510 Ciudad de México, México
| | - Alaide Jiménez Serna
- Centro de Investigación y Capacitación en Gastronomía, Universidad del Claustro de Sor Juana, 06080 Ciudad de México, México
| | - Amelia Farrés
- Departamento de Alimentos y Biotecnología, Facultad de Química, Universidad Nacional Autónoma de México, 04510 Ciudad de México, México
| |
Collapse
|
24
|
Li X, Tian S, Wang Y, Liu J, Wang J, Lu Y. Broccoli microgreens juice reduces body weight by enhancing insulin sensitivity and modulating gut microbiota in high-fat diet-induced C57BL/6J obese mice. Eur J Nutr 2021; 60:3829-3839. [PMID: 33866422 DOI: 10.1007/s00394-021-02553-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 04/01/2021] [Indexed: 02/06/2023]
Abstract
PURPOSE This study aimed to explore the protective effect of broccoli microgreens juice (BMJ) during C57BL/6J mice obesity development. METHODS The obese model mice, induced by feeding high-fat diet (HFD), were treated with BMJ by gavage for 10 weeks. Melbine was gavaged at 300 mg/(kg bw)/d, as a positive control group. RESULTS BMJ supplementation significantly reduced white adipose tissues (WAT) mass, the body weight and adipocyte size, and increased water intake in HFD-fed mice. Moreover, it improved glucose tolerance, reduced insulin level and HOMA-IR value, and alleviated insulin resistance. Compared with the HFD group, BMJ supplementation significantly increased the relative abundance of Bacteroidetes and decreased the ratio of Firmicutes to Bacteroidetes at the phylum level, and enriched Bacteroides_acidifaciens at the species level. These changes in the composition of gut microbiota are associated with the production of short-chain fatty acids (SCFAs), and reduced LPS levels, and had an obvious anti-inflammatory effect. CONCLUSIONS These findings suggested that the protective effects of BMJ on diet-induced obesity may be involved in gut microbiota-SCFAs-LPS-inflammatory axis. In addition, BMJ can enhance liver antioxidant capacity and reduce liver fat accumulation. Consequently, these results sustain BMJ as a novel functional food for obesity, on the basis of its opposing effects on HFD-induced obesity in mice.
Collapse
Affiliation(s)
- Xiangfei Li
- College of Food Science and Engineering, Nanjing University of Finance and Economics, Nanjing, 210023, People's Republic of China.,College of Food Science and Technology, Nanjing Agricultural University, Nanjing, 210095, People's Republic of China
| | - Shuhua Tian
- College of Food Science and Engineering, Nanjing University of Finance and Economics, Nanjing, 210023, People's Republic of China
| | - Yunfan Wang
- College of Food Science and Engineering, Nanjing University of Finance and Economics, Nanjing, 210023, People's Republic of China
| | - Jie Liu
- China-Canada Joint Lab of Food Nutrition and Health (Beijing), Beijing Technology and Business University, Beijing, 100048, China
| | - Jing Wang
- China-Canada Joint Lab of Food Nutrition and Health (Beijing), Beijing Technology and Business University, Beijing, 100048, China
| | - Yingjian Lu
- College of Food Science and Engineering, Nanjing University of Finance and Economics, Nanjing, 210023, People's Republic of China.
| |
Collapse
|
25
|
Nataraj BH, Shivanna SK, Rao P, Nagpal R, Behare PV. Evolutionary concepts in the functional biotics arena: a mini-review. Food Sci Biotechnol 2021; 30:487-496. [PMID: 33936839 PMCID: PMC8050181 DOI: 10.1007/s10068-020-00818-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 08/19/2020] [Accepted: 08/24/2020] [Indexed: 12/15/2022] Open
Abstract
Over the years, the attempts to elucidate the role of beneficial microorganisms in shaping human health are becoming fairly apparent. The functional impact conferred by such microbes is not only transmitted by viable cells or their metabolites but also through non-viable cells. Extensive research to unveil the protective action of such wonder bugs has resulted in categorizing the beneficial microflora and their bioactive metabolites into a variety of functional biotic concepts based on their intended applications in various forms. In the modern era, these are often termed as probiotics, prebiotics, synbiotics, postbiotics, next-generation probiotics, psychobiotics, oncobiotics, pharmabiotics, and metabiotics. Currently, the concept of traditional probiotics is being widened to include microbes beyond lactic acid bacteria. Indeed, this diversification has broadened the functional food portfolio from food to pharmaceuticals. In this context, the present review aims to summarize the existing biotic concepts and their differences thereof.
Collapse
Affiliation(s)
- Basavaprabhu H. Nataraj
- Technofunctional Starters Lab, National Collection of Dairy Cultures (NCDC), Dairy Microbiology Division, ICAR-National Dairy Research Insititute, Karnal, Haryana 132001 India
| | - Sonu K. Shivanna
- Dairy Chemistry Division, ICAR-National Dairy Research Insititute, Karnal, Haryana 132001 India
| | - Prabha Rao
- Dairy Microbiology Department, Dairy Science College, KVAFSU, Hebbal, Bengaluru, Karnataka 560024 India
| | - Ravinder Nagpal
- Center for Diabetes, Obesity and Metabolism, Department of Internal Medicine and Molecular Medicine, Wake Forest School of Medicine, Winston Salem, NC 27101 USA
| | - Pradip V. Behare
- Technofunctional Starters Lab, National Collection of Dairy Cultures (NCDC), Dairy Microbiology Division, ICAR-National Dairy Research Insititute, Karnal, Haryana 132001 India
| |
Collapse
|
26
|
Baliyan N, Dindhoria K, Kumar A, Thakur A, Kumar R. Comprehensive Substrate-Based Exploration of Probiotics From Undistilled Traditional Fermented Alcoholic Beverage ' Lugri'. Front Microbiol 2021; 12:626964. [PMID: 33776957 PMCID: PMC7994326 DOI: 10.3389/fmicb.2021.626964] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2020] [Accepted: 02/17/2021] [Indexed: 01/27/2023] Open
Abstract
Cereal-based traditional fermented beverages (TFBs) are prevalent among India’s ethnic community, and lugri is one such TFB popular among the tribal people of the Lahaul valley in North-Western Himalaya. Previous studies have reported that lugri harbors probiotics and contains amino acids and vitamins but comprehensive substrate-specific exploration of lugri for probiotic attributes is unexplored. The present study selected three substrate-based lugri (wheat, rice, and barley) to study their biochemical properties and explore potential probiotics. This study screened the best probiotic strains for antioxidant studies and the fermentative process. A biochemical analysis determined that rice-based lugri had a higher alcohol content, electric conductivity, crude protein, and lower pH than barley and wheat-based lugri. A total of 134 distinct morphotypes were screened, and 43 strains were selected based on their qualitatively superior acid and bile tolerance. Rice-based undistilled lugri harbored the most probiotics, with 22 out of 43 strains isolated. All 43 bacterial isolates exhibited properties like cell surface hydrophobicity, cell-auto aggregation, β-galactosidase, and exopolysaccharide production, supporting them as possible probiotics. Based on antibiotic susceptibility, hemolytic activity, and biofilm formation, all the bacterial strains were found to be non-pathogenic. Taxonomically, they ranged among eight distinct genera and 10 different species. Statistically, 12 isolates were found to be the most promising probiotic, and eight strains were isolated from rice-based undistilled lugri. Furthermore, the antioxidant activity of the promising isolates was tested, based on free-radical scavenging ability toward 2,2-diphenyl-1-picrylhydrazyl (4.39–16.41%) and 2,2′-azino-bis (3-ethylbenzothiazoline-6-sulfonic acid) (15.29–57.74%). The strain Lacticaseibacillus paracasei LUL:01 showed the best antioxidant activity and probiotic attributes, and hence was used for the production of fermented milk. The strain LUL:01 fermented the sterile milk within 18 h, and the viable count remained above the legal requirement of 6 log10 CFU/ml during 28 days storage at 4°C. The strain represents a suitable candidate for applying probiotic functional food formulation with several health benefits.
Collapse
Affiliation(s)
- Neha Baliyan
- Biotechnology Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur, India.,Academy of Scientific and Innovative Research (AcSIR), CSIR- Human Resource Development Centre, Ghaziabad, India
| | - Kiran Dindhoria
- Biotechnology Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur, India.,Academy of Scientific and Innovative Research (AcSIR), CSIR- Human Resource Development Centre, Ghaziabad, India
| | - Aman Kumar
- Biotechnology Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur, India.,Academy of Scientific and Innovative Research (AcSIR), CSIR- Human Resource Development Centre, Ghaziabad, India
| | - Aman Thakur
- Biotechnology Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur, India.,Academy of Scientific and Innovative Research (AcSIR), CSIR- Human Resource Development Centre, Ghaziabad, India
| | - Rakshak Kumar
- Biotechnology Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur, India.,Academy of Scientific and Innovative Research (AcSIR), CSIR- Human Resource Development Centre, Ghaziabad, India
| |
Collapse
|
27
|
Mays ZJS, Nair NU. A quantitative model for metabolic intervention using gut microbes. Biotechnol Prog 2021; 37:e3125. [PMID: 33426814 DOI: 10.1002/btpr.3125] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 01/04/2021] [Accepted: 01/05/2021] [Indexed: 11/08/2022]
Abstract
As medicine shifts toward precision-based and personalized therapeutics, utilizing more complex biomolecules to treat increasingly difficult and rare conditions, microorganisms provide an avenue for realizing the production and processing necessary for novel drug pipelines. More so, probiotic microbes can be co-opted to deliver therapeutics by oral administration as living drugs, able to survive and safely transit the digestive tract. As living therapeutics are in their nascency, traditional pharmacokinetic-pharmacodynamic (PK-PD) models for evaluating drug candidates are not appropriate for this novel platform. Using a living therapeutic in late-stage clinical development for phenylketonuria (PKU) as a case study, we adapt traditional oral drug delivery models to properly evaluate and inform the engineering of living therapeutics. We develop the adapted for living therapeutics compartmental absorption and transit (ALT-CAT) model to provide metrics for drug efficacy across nine age groups of PKU patients and evaluate model parameters that are influenced by patient physiology, microbe selection and therapeutic production, and dosing formulations. In particular, the ALT-CAT model describes the mathematical framework to model the behavior of orally delivered engineered bacteria that act as living therapeutics by adapting similar methods that have been developed and widely-used for small molecular drug delivery and absorption.
Collapse
Affiliation(s)
- Zachary J S Mays
- Department of Chemical & Biological Engineering, Tufts University, Medford, Massachusetts, USA
| | - Nikhil U Nair
- Department of Chemical & Biological Engineering, Tufts University, Medford, Massachusetts, USA
| |
Collapse
|
28
|
Asgari S, Pourjavadi A, Licht TR, Boisen A, Ajalloueian F. Polymeric carriers for enhanced delivery of probiotics. Adv Drug Deliv Rev 2020; 161-162:1-21. [PMID: 32702378 DOI: 10.1016/j.addr.2020.07.014] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Revised: 07/15/2020] [Accepted: 07/17/2020] [Indexed: 12/14/2022]
Abstract
Probiotics are live microorganisms (usually bacteria), which are defined by their ability to confer health benefits to the host, if administered adequately. Probiotics are not only used as health supplements but have also been applied in various attempts to prevent and treat gastrointestinal (GI) and non-gastrointestinal diseases such as diarrhea, colon cancer, obesity, diabetes, and inflammation. One of the challenges in the use of probiotics is putative loss of viability by the time of administration. It can be due to procedures that the probiotic products go through during fabrication, storage, or administration. Biocompatible and biodegradable polymers with specific moieties or pH/enzyme sensitivity have shown great potential as carriers of the bacteria for 1) better viability, 2) longer storage times, 3) preservation from the aggressive environment in the stomach and 4) topographically targeted delivery of probiotics. In this review, we focus on polymeric carriers and the procedures applied for encapsulation of the probiotics into them. At the end, some novel methods for specific probiotic delivery, possibilities to improve the targeted delivery of probiotics and some challenges are discussed.
Collapse
|