1
|
Zhang Y, He J, Zeng H, Xu D, Li W, Wang Y. Advances in prebiotic carbohydrate-based targeted delivery: Overcoming gastrointestinal challenges for bioactive ingredients. Food Chem 2025; 466:142210. [PMID: 39615354 DOI: 10.1016/j.foodchem.2024.142210] [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: 07/24/2024] [Revised: 11/05/2024] [Accepted: 11/20/2024] [Indexed: 12/14/2024]
Abstract
Natural bioactive ingredients face challenges in extensive application owing to low oral bioavailability. This can be improved by overcoming gastrointestinal barriers and facilitating targeted release through delivery strategies. This study provides a comprehensive review of targeted delivery systems using prebiotic carbohydrate matrices, focusing on structures, release mechanisms and applications. The bioactive ingredients can be encapsulated into nanohydrogels, nanoparticles, nanoemulsions, micro/nanocapsules and nanofibres to achieve controlled/targeted delivery to predetermined locations via interactions with pH, mucus, microbiome, enzymes and other factors in the colon. In particular, the prebiotic function of carbohydrates is generated by colonic microbiota degradation and fermentation, producing beneficial postbiotics through multiple metabolic pathways. This study provides certain insights into the in-depth development and application of prebiotic carbohydrate-based targeted delivery systems in the fields of food and health.
Collapse
Affiliation(s)
- Yunzhen Zhang
- School of Food and Health, Beijing Technology and Business University, Haidian, 100048, Beijing, PR China
| | - Jian He
- National Center of Technology Innovation for Dairy, 8 West Guochuang Road, Hohhot 010110, Inner Mongolia, PR China
| | - Hong Zeng
- School of Food and Health, Beijing Technology and Business University, Haidian, 100048, Beijing, PR China
| | - Duoxia Xu
- School of Food and Health, Beijing Technology and Business University, Haidian, 100048, Beijing, PR China
| | - Wenlu Li
- School of Food and Health, Beijing Technology and Business University, Haidian, 100048, Beijing, PR China.
| | - Yanbo Wang
- School of Food and Health, Beijing Technology and Business University, Haidian, 100048, Beijing, PR China.
| |
Collapse
|
2
|
Zhang Y, Amin K, Zhang Q, Yu Z, Jing W, Wang Z, Lyu B, Yu H. The application of dietary fibre as microcapsule wall material in food processing. Food Chem 2025; 463:141195. [PMID: 39276558 DOI: 10.1016/j.foodchem.2024.141195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 08/11/2024] [Accepted: 09/06/2024] [Indexed: 09/17/2024]
Abstract
In the food industry, functional ingredients derived from active substances of natural sources and microbiological resources are gaining acceptance and demand due to their beneficial health properties. However, the inherent instability of these constituents poses a challenge in utilizing their functional properties. Microencapsulation with dietary fibre as wall material technology offers a promising solution, providing convenient manipulability and effective safeguarding of encapsulated substances. This paper presents a comprehensive overview of the current state of research on dietary fibre-based microcapsules in food processing. It examines their functional attributes, the preparation technology, and their applications within the food industry. Furthermore, the constraints associated with industrial production are discussed, as well as potential future developments. This article offers researchers a reference point and a theoretical basis for the selection of innovative food ingredients, the high-value utilisation of dietary fibre, and the design of conservation strategies for functional substances in food production.
Collapse
Affiliation(s)
- Ying Zhang
- College of Food Science and Engineering, Jilin Agricultural University, Changchun 130118, China; Division of Soybean Processing, Soybean Research & Development Center, Chinese Agricultural Research System, Changchun 130118, China
| | - Khalid Amin
- College of Food Science and Engineering, Jilin Agricultural University, Changchun 130118, China; Division of Soybean Processing, Soybean Research & Development Center, Chinese Agricultural Research System, Changchun 130118, China
| | - Qiang Zhang
- College of Food Science and Engineering, Jilin Agricultural University, Changchun 130118, China; Division of Soybean Processing, Soybean Research & Development Center, Chinese Agricultural Research System, Changchun 130118, China
| | - Ziyue Yu
- College of Food Science and Engineering, Jilin Agricultural University, Changchun 130118, China; Division of Soybean Processing, Soybean Research & Development Center, Chinese Agricultural Research System, Changchun 130118, China
| | - Wendan Jing
- College of Food Science and Engineering, Jilin Agricultural University, Changchun 130118, China; Division of Soybean Processing, Soybean Research & Development Center, Chinese Agricultural Research System, Changchun 130118, China
| | - Zhaohui Wang
- College of Food Science and Engineering, Jilin Agricultural University, Changchun 130118, China; Division of Soybean Processing, Soybean Research & Development Center, Chinese Agricultural Research System, Changchun 130118, China
| | - Bo Lyu
- College of Food Science and Engineering, Jilin Agricultural University, Changchun 130118, China; Division of Soybean Processing, Soybean Research & Development Center, Chinese Agricultural Research System, Changchun 130118, China.
| | - Hansong Yu
- College of Food Science and Engineering, Jilin Agricultural University, Changchun 130118, China; Division of Soybean Processing, Soybean Research & Development Center, Chinese Agricultural Research System, Changchun 130118, China.
| |
Collapse
|
3
|
Mamun AA, Geng P, Wang S, Shao C, Xiao J. IUPHAR review: Targeted therapies of signaling pathways based on the gut microbiome in autism spectrum disorders: Mechanistic and therapeutic applications. Pharmacol Res 2024; 211:107559. [PMID: 39733842 DOI: 10.1016/j.phrs.2024.107559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 12/22/2024] [Accepted: 12/23/2024] [Indexed: 12/31/2024]
Abstract
Autism spectrum disorders (ASD) are complex neurodevelopmental disorders characterized by impairments in social interaction, communication and repetitive activities. Gut microbiota significantly influences behavior and neurodevelopment by regulating the gut-brain axis. This review explores gut microbiota-influenced treatments for ASD, focusing on their therapeutic applications and mechanistic insights. In addition, this review discusses the interactions between gut microbiota and the immune, metabolic and neuroendocrine systems, focusing on crucial microbial metabolites including short-chain fatty acids (SCFAs) and several neurotransmitters. Furthermore, the review explores various therapy methods including fecal microbiota transplantation, dietary modifications, probiotics and prebiotics and evaluates their safety and efficacy in reducing ASD symptoms. The discussion shows the potential of customized microbiome-based therapeutics and the integration of multi-omics methods to understand the underlying mechanisms. Moreover, the review explores the intricate relationship between gut microbiota and ASD, aiming to develop innovative therapies that utilize the gut microbiome to improve the clinical outcomes of ASD patients. Microbial metabolites such as neurotransmitter precursors, tryptophan metabolites and SCFAs affect brain development and behavior. Symptoms of ASD are linked to changes in these metabolites. Dysbiosis in the gut microbiome may impact neuroinflammatory processes linked to autism, negatively affecting immune signaling pathways. Research indicates that probiotics and prebiotics can improve gut microbiota and alleviate symptoms in ASD patients. Fecal microbiota transplantation may also improve behavioral symptoms and restore gut microbiota balance. The review emphasizes the need for further research on gut microbiota modification as a potential therapeutic approach for ASD, highlighting its potential in clinical settings.
Collapse
Affiliation(s)
- Abdullah Al Mamun
- Central Laboratory of The Lishui Hospital of Wenzhou Medical University, The First Affiliated Hospital of Lishui University, Lishui People's Hospital, Lishui, Zhejiang 323000, China; Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Peiwu Geng
- Central Laboratory of The Lishui Hospital of Wenzhou Medical University, The First Affiliated Hospital of Lishui University, Lishui People's Hospital, Lishui, Zhejiang 323000, China
| | - Shuanghu Wang
- Central Laboratory of The Lishui Hospital of Wenzhou Medical University, The First Affiliated Hospital of Lishui University, Lishui People's Hospital, Lishui, Zhejiang 323000, China
| | - Chuxiao Shao
- Central Laboratory of The Lishui Hospital of Wenzhou Medical University, The First Affiliated Hospital of Lishui University, Lishui People's Hospital, Lishui, Zhejiang 323000, China.
| | - Jian Xiao
- Central Laboratory of The Lishui Hospital of Wenzhou Medical University, The First Affiliated Hospital of Lishui University, Lishui People's Hospital, Lishui, Zhejiang 323000, China; Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China; Department of Wound Healing, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China.
| |
Collapse
|
4
|
Yoo Y, Kim S, Lee W, Kim J, Son B, Lee KJ, Shin H. The prebiotic potential of dietary onion extracts: shaping gut microbial structures and promoting beneficial metabolites. mSystems 2024:e0118924. [PMID: 39714164 DOI: 10.1128/msystems.01189-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Accepted: 11/23/2024] [Indexed: 12/24/2024] Open
Abstract
Onions are well-known vegetables that offer various health benefits. This study explores the impact of onion extracts on gut microbiome using an in vitro fecal incubation model and metabolome analysis. Fecal samples were collected from 19 healthy donors and incubated in the presence or absence of onion extracts for 24 h. To reduce inter-individual variability in the gut microbiome, we employed enterotyping based on baseline fecal microbiota: 14 subjects with a Bacteroides-dominant type (enterotype B) and 5 subjects with Prevotella-dominant type (enterotype P). Alpha diversity was significantly reduced in the onion-treated group compared to the non-treated control group in both Bacteroides- and Prevotella-dominant types. However, significant structural differences in bacterial communities were observed based on weighted UniFrac distance. Notably, short-chain fatty acid (SCFA)-producing bacteria, such as Bifidobacterium_388775, Feacalibacterium, and Fusicatenibacter, were overrepresented in response to onion extracts in enterotype B. Furthermore, genes related to butyrate production were significantly overrepresented in the onion-treated group within enterotype B. Consistent with the enriched taxa and the predicted metabolic pathways, SCFAs and their related metabolites were significantly enriched in the onion-treated group. Additionally, tryptophan metabolism-derived metabolites, including indolelactate (ILA) and indolepropionate (IPA), were elevated by 4- and 32-fold, respectively, in the onion-treated group compared to the control group. In vitro growth assays showed an increase in lactobacilli strains in the presence of onion extracts. These results provide evidence that onion extracts could serve as promising prebiotics by altering gut microbial structure and promoting the production of beneficiary metabolites, including SCFAs and indole derivatives, and enhancing the growth of probiotics.IMPORTANCEThis study is significant as it provides compelling evidence that onion extracts have the potential to serve as effective prebiotics. Utilizing an in vitro fecal incubation model and enterotyping to reduce inter-individual variability, the research demonstrates how onion extracts can alter gut microbial structure and promote the production of beneficial metabolites, including SCFAs and indole derivatives like ILA and IPA. Additionally, onion extract treatment enhances the growth of beneficial probiotics. The findings underscore the potential of onion extracts to improve gut health by enriching specific beneficial bacteria and metabolic pathways, thereby supporting the development of functional foods aimed at improving gut microbiota composition and metabolic health.
Collapse
Affiliation(s)
- Yebeen Yoo
- Department of Food Science and Biotechnology, College of Life Science, Sejong University, Seoul, South Korea
| | - Seongok Kim
- Department of Food Science and Biotechnology, College of Life Science, Sejong University, Seoul, South Korea
- Carbohydrate Bioproduct Research Center, Sejong University, Seoul, South Korea
| | - WonJune Lee
- Carbohydrate Bioproduct Research Center, Sejong University, Seoul, South Korea
| | - Jinwoo Kim
- Department of Food Science and Biotechnology, College of Life Science, Sejong University, Seoul, South Korea
- Carbohydrate Bioproduct Research Center, Sejong University, Seoul, South Korea
| | - Bokyung Son
- Department of Food Biotechnology, Dong-A University, Busan, Republic of Korea
| | - Kwang Jun Lee
- Division of Zoonotic and Vector Borne Diseases Research, Center for Infectious Diseases Research, National Institute of Health, Cheongju, South Korea
| | - Hakdong Shin
- Department of Food Science and Biotechnology, College of Life Science, Sejong University, Seoul, South Korea
- Carbohydrate Bioproduct Research Center, Sejong University, Seoul, South Korea
| |
Collapse
|
5
|
Xue H, Tang Y, Zha M, Xie K, Tan J. The structure-function relationships and interaction between polysaccharides and intestinal microbiota: A review. Int J Biol Macromol 2024; 291:139063. [PMID: 39710020 DOI: 10.1016/j.ijbiomac.2024.139063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Revised: 12/12/2024] [Accepted: 12/19/2024] [Indexed: 12/24/2024]
Abstract
The gut microbiota, as a complex ecosystem, can affect many physiological aspects of the host's diet, disease development, drug metabolism, and immune system regulation. Polysaccharides have various biological activities including antioxidant, anti-tumor, and regulating gut microbiota, etc. Polysaccharides cannot be degraded by human digestive enzymes. However, the interaction between gut microbiota and polysaccharides can lead to the degradation and utilization of polysaccharides. Disordered intestinal flora leads to diseases such as diabetes, hyperlipidemia, tumors, and diarrhea. Notably, polysaccharides can regulate the gut microbiota, promote the proliferation of probiotics and the SCFAs production, and thus improve the related-diseases and maintain body health. The relationship between polysaccharides and gut microbiota is gradually becoming clear. Nevertheless, the structure-function relationships between polysaccharides and gut microbiota still need further exploration. Hence, this paper systematically reviews the structure-function relationships between polysaccharides and gut microbiota from four aspects including molecular weight, glycosidic bonds, monosaccharide composition, and advanced structure. Moreover, this review outlines the effect of polysaccharides on gut microbiota metabolism and improves diseases by regulating gut microbiota. Furthermore, this article introduces the impact of gut microbiota on polysaccharide metabolism. The findings can provide the scientific basis for in-depth research on body health and reasonable diet.
Collapse
Affiliation(s)
- Hongkun Xue
- College of Traditional Chinese Medicine, Hebei University, No. 342 Yuhua East Road, Lianchi District, Baoding 071002, China
| | - Yingqi Tang
- College of Traditional Chinese Medicine, Hebei University, No. 342 Yuhua East Road, Lianchi District, Baoding 071002, China
| | - Min Zha
- College of Traditional Chinese Medicine, Hebei University, No. 342 Yuhua East Road, Lianchi District, Baoding 071002, China
| | - Kaifang Xie
- College of Textile and Fashion, Hunan Institute of Engineering, NO. 88 East Fuxing Road, Yuetang District, Xiangtan 411100, China
| | - Jiaqi Tan
- Medical Comprehensive Experimental Center, Hebei University, No. 342 Yuhua East Road, Lianchi District, Baoding 071002, China.
| |
Collapse
|
6
|
Prattico C, Gonzalez E, Dridi L, Jazestani S, Low KE, Abbott DW, Maurice CF, Castagner B. Identification of novel fructo-oligosaccharide bacterial consumers by pulse metatranscriptomics in a human stool sample. mSphere 2024:e0066824. [PMID: 39699190 DOI: 10.1128/msphere.00668-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Accepted: 11/26/2024] [Indexed: 12/20/2024] Open
Abstract
Dietary fibers influence the composition of the human gut microbiota and directly contribute to its downstream effects on host health. As more research supports the use of glycans as prebiotics for therapeutic applications, the need to identify the gut bacteria that metabolize glycans of interest increases. Fructo-oligosaccharide (FOS) is a common diet-derived glycan that is fermented by the gut microbiota and has been used as a prebiotic. Despite being well studied, we do not yet have a complete picture of all FOS-consuming gut bacterial taxa. To identify new bacterial consumers, we used a short exposure of microbial communities in a stool sample to FOS or galactomannan as the sole carbon source to induce glycan metabolism genes. We then performed metatranscriptomics, paired with whole metagenomic sequencing, and 16S amplicon sequencing. The short incubation was sufficient to cause induction of genes involved in carbohydrate metabolism, like carbohydrate-active enzymes (CAZymes), including glycoside hydrolase family 32 genes, which hydrolyze fructan polysaccharides like FOS and inulin. Interestingly, FOS metabolism transcripts were notably overexpressed in Blautia species not previously reported to be fructan consumers. We therefore validated the ability of different Blautia species to ferment fructans by monitoring their growth and fermentation in defined media. This pulse metatranscriptomics approach is a useful method to find novel consumers of prebiotics and increase our understanding of prebiotic metabolism by CAZymes in the gut microbiota. IMPORTANCE Complex carbohydrates are key contributors to the composition of the human gut microbiota and play an essential role in the microbiota's effects on host health. Understanding which bacteria consume complex carbohydrates, or glycans, provides a mechanistic link between dietary prebiotics and their beneficial health effects, an essential step for their therapeutic application. Here, we used a pulse metatranscriptomics pipeline to identify bacterial consumers based on glycan metabolism induction in a human stool sample. We identified novel consumers of fructo-oligosaccharide among Blautia species, expanding our understanding of this well-known glycan. Our approach can be applied to identify consumers of understudied glycans and expand our prebiotic repertoire. It can also be used to study prebiotic glycans directly in stool samples in distinct patient populations to help delineate the prebiotic mechanism.
Collapse
Affiliation(s)
- Catherine Prattico
- Department of Microbiology & Immunology, Faculty of Medicine and Health Sciences, McGill University, Montréal, Québec, Canada
| | - Emmanuel Gonzalez
- Canadian Centre for Computational Genomics, McGill Genome Centre, McGill University, Montréal, Québec, Canada
- Department of Human Genetics, McGill University, Montréal, Québec, Canada
- Gerald Bronfman Department of Oncology, McGill University, Montréal, Québec, Canada
| | - Lharbi Dridi
- Department of Pharmacology & Therapeutics, Faculty of Medicine and Health Sciences, McGill University, Montréal, Québec, Canada
| | - Shiva Jazestani
- Department of Pharmacology & Therapeutics, Faculty of Medicine and Health Sciences, McGill University, Montréal, Québec, Canada
| | - Kristin E Low
- Agriculture and Agri-Food Canada, Lethbridge Research and Development Centre, Lethbridge, Alberta, Canada
| | - D Wade Abbott
- Agriculture and Agri-Food Canada, Lethbridge Research and Development Centre, Lethbridge, Alberta, Canada
| | - Corinne F Maurice
- Department of Microbiology & Immunology, Faculty of Medicine and Health Sciences, McGill University, Montréal, Québec, Canada
- McGill Centre for Microbiome Research, McGill University, Montréal, Québec, Canada
| | - Bastien Castagner
- Department of Pharmacology & Therapeutics, Faculty of Medicine and Health Sciences, McGill University, Montréal, Québec, Canada
- McGill Centre for Microbiome Research, McGill University, Montréal, Québec, Canada
| |
Collapse
|
7
|
Kaur S, Patel BCK, Collen A, Malhotra R. The microbiome and the eye: a new era in ophthalmology. Eye (Lond) 2024:10.1038/s41433-024-03517-z. [PMID: 39702789 DOI: 10.1038/s41433-024-03517-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 11/10/2024] [Accepted: 11/26/2024] [Indexed: 12/21/2024] Open
Abstract
The human microbiome has progressively been recognised for its role in various disease processes. In ophthalmology, complex interactions between the gut and distinct ocular microbiota within each structure and microenvironment of the eye has advanced our knowledge on the multi-directional relationships of these ecosystems. Increasingly, studies have shown that modulation of the microbiome can be achieved through faecal microbiota transplantation and synbiotics producing favourable outcomes for ophthalmic diseases. As ophthalmologists, we are obliged to educate our patients on measures to cultivate a healthy gut microbiome through a range of holistic measures. Further integrative studies combining microbial metagenomics, metatranscriptomics and metabolomics are necessary to fully characterise the human microbiome and enable targeted therapeutic interventions.
Collapse
Affiliation(s)
- Simerdip Kaur
- Department of Ophthalmology, University Hospitals Sussex NHS Foundation Trust, Sussex Eye Hospital, Eastern Road, Brighton, BN2 5BF, UK.
- Corneoplastic Unit, Queen Victoria Hospital, East Grinstead, RH19 3DZ, UK.
| | - Bhupendra C K Patel
- Department of Ophthalmology and Visual Sciences, John A. Moran Eye Center, University of Utah, Salt Lake City, Utah, 84132, USA
| | - Alanna Collen
- Unaffiliated officially. Independent author, London, UK
| | - Raman Malhotra
- Corneoplastic Unit, Queen Victoria Hospital, East Grinstead, RH19 3DZ, UK
| |
Collapse
|
8
|
Zhang C, Hao L, Zhu Y, Zhang X, Zhao H, Zhang B. In vitro fermentation characteristics and modulation effects of polysaccharide fractions from Schisandra sphenanthera on intestinal microflora. Int J Biol Macromol 2024; 289:138771. [PMID: 39701254 DOI: 10.1016/j.ijbiomac.2024.138771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Revised: 12/10/2024] [Accepted: 12/11/2024] [Indexed: 12/21/2024]
Abstract
Schisandra Sphenanthera polysaccharides fractions (SSPs), namely SSP40, SSP60, and SSP80, were obtained by gradient precipitation with 40 %, 60 %, and 80 % (v/v) ethanol, respectively. It was found that gradient ethanol precipitation (GEP) significantly affected the physicochemical and structural characteristics of SSPs, including molecular weight, monosaccharide composition, and surface morphology. Compared to fractions SSP40 and SSP60, SSP80 was observed to have a lower molecular weight (22.58 kDa) and certain specific monosaccharide composition, such as lower glucose content and higher galactose, arabinose, rhamnose, and galacturonic acid content. Furthermore, the apparent porosity of the SSPs increased with increasing ethanol concentration in GEP. After fermentation at 37 °C for 48 h, fraction SSP80 prominently promoted the production of more short-chain fatty acids (SCFAs), increasing from an initial 1.39 ± 0.08 to 26.75 ± 0.54 mmol/L. The SSP fraction types extracted by GEP greatly affected the modulation of the intestinal microflora at different levels. The SSP80 fraction with excellent structure demonstrated the best ability to modulate the intestinal microflora by increasing the relative abundance of Bacteroides, Faecalibacterium and Dialister and decreasing the relative abundance of Escherichia-Shigella. The remarkable differences in modulating the intestinal microflora confirmed the importance of carefully selecting GEP to fraction SSPs that promote health.
Collapse
Affiliation(s)
- Chen Zhang
- Beijing Key Laboratory of Forest Food Processing and Safety, College of Biological Science & Biotechnology, Beijing Forestry University, Beijing 100083, China
| | - Lei Hao
- Beijing Key Laboratory of Forest Food Processing and Safety, College of Biological Science & Biotechnology, Beijing Forestry University, Beijing 100083, China
| | - Yadong Zhu
- Beijing Key Laboratory of Forest Food Processing and Safety, College of Biological Science & Biotechnology, Beijing Forestry University, Beijing 100083, China
| | - Xiaojia Zhang
- Beijing Key Laboratory of Forest Food Processing and Safety, College of Biological Science & Biotechnology, Beijing Forestry University, Beijing 100083, China
| | - Hongfei Zhao
- Beijing Key Laboratory of Forest Food Processing and Safety, College of Biological Science & Biotechnology, Beijing Forestry University, Beijing 100083, China.
| | - Bolin Zhang
- Beijing Key Laboratory of Forest Food Processing and Safety, College of Biological Science & Biotechnology, Beijing Forestry University, Beijing 100083, China.
| |
Collapse
|
9
|
Al-Beltagi M. Nutritional management and autism spectrum disorder: A systematic review. World J Clin Pediatr 2024; 13:99649. [PMID: 39654662 PMCID: PMC11572612 DOI: 10.5409/wjcp.v13.i4.99649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 09/21/2024] [Accepted: 10/15/2024] [Indexed: 10/30/2024] Open
Abstract
BACKGROUND Autism spectrum disorder (ASD) presents unique challenges related to feeding and nutritional management. Children with ASD often experience feeding difficulties, including food selectivity, refusal, and gastrointestinal issues. Various interventions have been explored to address these challenges, including dietary modifications, vitamin supplementation, feeding therapy, and behavioral interventions. AIM To provide a comprehensive overview of the current evidence on nutritional management in ASD. We examine the effectiveness of dietary interventions, vitamin supplements, feeding therapy, behavioral interventions, and mealtime practices in addressing the feeding challenges and nutritional needs of children with ASD. METHODS We systematically searched relevant literature up to June 2024, using databases such as PubMed, PsycINFO, and Scopus. Studies were included if they investigated dietary interventions, nutritional supplements, or behavioral strategies to improve feeding behaviors in children with ASD. We assessed the quality of the studies and synthesized findings on the impact of various interventions on feeding difficulties and nutritional outcomes. Data extraction focused on intervention types, study designs, participant characteristics, outcomes measured, and intervention effectiveness. RESULTS The review identified 316 studies that met the inclusion criteria. The evidence indicates that while dietary interventions and nutritional supplements may offer benefits in managing specific symptoms or deficiencies, the effectiveness of these approaches varies. Feeding therapy and behavioral interventions, including gradual exposure and positive reinforcement, promise to improve food acceptance and mealtime behaviors. The findings also highlight the importance of creating supportive mealtime environments tailored to the sensory and behavioral needs of children with ASD. CONCLUSION Nutritional management for children with ASD requires a multifaceted approach that includes dietary modifications, supplementation, feeding therapy, and behavioral strategies. The review underscores the need for personalized interventions and further research to refine treatment protocols and improve outcomes. Collaborative efforts among healthcare providers, educators, and families are essential to optimize this population's nutritional health and feeding practices. Enhancing our understanding of intervention sustainability and long-term outcomes is essential for optimizing care and improving the quality of life for children with ASD and their families.
Collapse
Affiliation(s)
- Mohammed Al-Beltagi
- Department of Pediatric, Faculty of Medicine, Tanta University, Tanta 31511, Alghrabia, Egypt
- Department of Pediatric, University Medical Center, King Abdulla Medical City, Arabian Gulf University, Manama 26671, Bahrain
| |
Collapse
|
10
|
Eskandari A, Leow TC, Rahman MBA, Oslan SN. Recent insight into the advances and prospects of microbial lipases and their potential applications in industry. Int Microbiol 2024; 27:1597-1631. [PMID: 38489100 DOI: 10.1007/s10123-024-00498-7] [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: 01/29/2024] [Revised: 03/04/2024] [Accepted: 03/07/2024] [Indexed: 03/17/2024]
Abstract
Enzymes play a crucial role in various industrial sectors. These biocatalysts not only ensure sustainability and safety but also enhance process efficiency through their unique specificity. Lipases possess versatility as biocatalysts and find utilization in diverse bioconversion reactions. Presently, microbial lipases are gaining significant focus owing to the rapid progress in enzyme technology and their widespread implementation in multiple industrial procedures. This updated review presents new knowledge about various origins of microbial lipases, such as fungi, bacteria, and yeast. It highlights both the traditional and modern purification methods, including precipitation and chromatographic separation, the immunopurification technique, the reversed micellar system, the aqueous two-phase system (ATPS), and aqueous two-phase flotation (ATPF), moreover, delves into the diverse applications of microbial lipases across several industries, such as food, vitamin esters, textile, detergent, biodiesel, and bioremediation. Furthermore, the present research unveils the obstacles encountered in employing lipase, the patterns observed in lipase engineering, and the application of CRISPR/Cas genome editing technology for altering the genes responsible for lipase production. Additionally, the immobilization of microorganisms' lipases onto various carriers also contributes to enhancing the effectiveness and efficiencies of lipases in terms of their catalytic activities. This is achieved by boosting their resilience to heat and ionic conditions (such as inorganic solvents, high-level pH, and temperature). The process also facilitates the ease of recycling them and enables a more concentrated deposition of the enzyme onto the supporting material. Consequently, these characteristics have demonstrated their suitability for application as biocatalysts in diverse industries.
Collapse
Affiliation(s)
- Azadeh Eskandari
- Enzyme and Microbial Technology Research Centre, Universiti Putra Malaysia, UPM, 43400, Serdang, Selangor, Malaysia
- Department of Biochemistry, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, UPM, 43400, Serdang, Selangor, Malaysia
| | - Thean Chor Leow
- Enzyme and Microbial Technology Research Centre, Universiti Putra Malaysia, UPM, 43400, Serdang, Selangor, Malaysia
- Department of Cell and Molecular Biology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, UPM, 43400, Serdang, Selangor, Malaysia
- Enzyme Technology and X-ray Crystallography Laboratory, VacBio 5, Institute of Bioscience, Universiti Putra Malaysia, UPM, 43400, Serdang, Selangor, Malaysia
| | | | - Siti Nurbaya Oslan
- Enzyme and Microbial Technology Research Centre, Universiti Putra Malaysia, UPM, 43400, Serdang, Selangor, Malaysia.
- Department of Biochemistry, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, UPM, 43400, Serdang, Selangor, Malaysia.
- Enzyme Technology and X-ray Crystallography Laboratory, VacBio 5, Institute of Bioscience, Universiti Putra Malaysia, UPM, 43400, Serdang, Selangor, Malaysia.
| |
Collapse
|
11
|
Dahiya P, Kumari S, Behl M, Kashyap A, Kumari D, Thakur K, Devi M, Kumari N, Kaushik N, Walia A, Bhatt AK, Bhatia RK. Guardians of the Gut: Harnessing the Power of Probiotic Microbiota and Their Exopolysaccharides to Mitigate Heavy Metal Toxicity in Human for Better Health. Probiotics Antimicrob Proteins 2024; 16:1937-1953. [PMID: 38733461 DOI: 10.1007/s12602-024-10281-9] [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] [Accepted: 04/26/2024] [Indexed: 05/13/2024]
Abstract
Heavy metal pollution is a significant global health concern, posing risks to both the environment and human health. Exposure to heavy metals happens through various channels like contaminated water, food, air, and workplaces, resulting in severe health implications. Heavy metals also disrupt the gut's microbial balance, leading to dysbiosis characterized by a decrease in beneficial microorganisms and proliferation in harmful ones, ultimately exacerbating health problems. Probiotic microorganisms have demonstrated their ability to adsorb and sequester heavy metals, while their exopolysaccharides (EPS) exhibit chelating properties, aiding in mitigating heavy metal toxicity. These beneficial microorganisms aid in restoring gut integrity through processes like biosorption, bioaccumulation, and biotransformation of heavy metals. Incorporating probiotic strains with high affinity for heavy metals into functional foods and supplements presents a practical approach to mitigating heavy metal toxicity while enhancing gut health. Utilizing probiotic microbiota and their exopolysaccharides to address heavy metal toxicity offers a novel method for improving human health through modulation of the gut microbiome. By combining probiotics and exopolysaccharides, a distinctive strategy emerges for mitigating heavy metal toxicity, highlighting promising avenues for therapeutic interventions and health improvements. Further exploration in this domain could lead to groundbreaking therapies and preventive measures, underscoring probiotic microbiota and exopolysaccharides as natural and environmentally friendly solutions to heavy metal toxicity. This, in turn, could enhance public health by safeguarding the gut from environmental contaminants.
Collapse
Affiliation(s)
- Pushpak Dahiya
- Department of Biotechnology, Himachal Pradesh University, Summer Hill, Shimla, 171005, Himachal Pradesh, India
| | - Sangeeta Kumari
- Department of Biotechnology, Himachal Pradesh University, Summer Hill, Shimla, 171005, Himachal Pradesh, India
| | - Manya Behl
- Department of Biotechnology, Himachal Pradesh University, Summer Hill, Shimla, 171005, Himachal Pradesh, India
| | - Aakash Kashyap
- Department of Biotechnology, Himachal Pradesh University, Summer Hill, Shimla, 171005, Himachal Pradesh, India
| | - Deeksha Kumari
- Department of Biotechnology, Himachal Pradesh University, Summer Hill, Shimla, 171005, Himachal Pradesh, India
| | - Kalpana Thakur
- Department of Biotechnology, Himachal Pradesh University, Summer Hill, Shimla, 171005, Himachal Pradesh, India
| | - Mamta Devi
- Department of Biotechnology, Himachal Pradesh University, Summer Hill, Shimla, 171005, Himachal Pradesh, India
| | - Neelam Kumari
- Department of Biosciences, Himachal Pradesh University, Summer Hill, Shimla, 171005, Himachal Pradesh, India
| | - Neelam Kaushik
- Department of Biotechnology, Himachal Pradesh University, Summer Hill, Shimla, 171005, Himachal Pradesh, India
| | - Abhishek Walia
- Department of Microbiology, College of Basic Sciences, CSK HPKV, Palampur, HP, 176062, India
| | - Arvind Kumar Bhatt
- Department of Biotechnology, Himachal Pradesh University, Summer Hill, Shimla, 171005, Himachal Pradesh, India
| | - Ravi Kant Bhatia
- Department of Biotechnology, Himachal Pradesh University, Summer Hill, Shimla, 171005, Himachal Pradesh, India.
| |
Collapse
|
12
|
Kapoor B, Biswas P, Gulati M, Rani P, Gupta R. Gut microbiome and Alzheimer's disease: What we know and what remains to be explored. Ageing Res Rev 2024; 102:102570. [PMID: 39486524 DOI: 10.1016/j.arr.2024.102570] [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: 06/18/2024] [Revised: 10/26/2024] [Accepted: 10/26/2024] [Indexed: 11/04/2024]
Abstract
With advancement in human microbiome research, an increasing number of scientific evidences have endorsed the key role of gut microbiota in the pathogenesis of Alzheimer disease. Microbiome dysbiosis, characterized by altered diversity and composition, as well as rise of pathobionts influence not only various gut disorder but also central nervous system disorders such as AD. On the basis of accumulated evidences of past few years now it is quite clear that the gut microbiota can control the functions of the central nervous system (CNS) through the gut-brain axis, which provides a new prospective into the interactions between the gut and brain. The main focus of this review is on the molecular mechanism of the crosstalk between the gut microbiota and the brain through the gut-brain axis, and on the onset and development of neurological disorders triggered by the dysbiosis of gut microbiota. Due to microbiota dysbiosis the permeability of the gut and blood brain barrier is increased which may mediate or affect AD. Along with this, bacterial population of the gut microbiota can secrete amyloid proteins and lipopolysaccharides in a large quantity which may create a disturbance in the signaling pathways and the formation of proinflammatory cytokines associated with the pathogenesis of AD. These topics are followed by a critical analysis of potential intervention strategies targeting gut microbiota dysbiosis, including the use of probiotics, prebiotics, metabolites, diets and fecal microbiota transplantation. The main purpose of this review includes the summarization and discussion on the recent finding that may explain the role of the gut microbiota in the development of AD. Understanding of these fundamental mechanisms may provide a new insight into the novel therapeutic strategies for AD.
Collapse
Affiliation(s)
- Bhupinder Kapoor
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, India.
| | - Pratim Biswas
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, India
| | - Monica Gulati
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, India; Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, NSW 2007, Australia
| | - Pooja Rani
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, India
| | - Reena Gupta
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, India
| |
Collapse
|
13
|
Deehan EC, Al Antwan S, Witwer RS, Guerra P, John T, Monheit L. Revisiting the Concepts of Prebiotic and Prebiotic Effect in Light of Scientific and Regulatory Progress-A Consensus Paper From the Global Prebiotic Association. Adv Nutr 2024; 15:100329. [PMID: 39481540 PMCID: PMC11616045 DOI: 10.1016/j.advnut.2024.100329] [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: 07/31/2024] [Revised: 10/18/2024] [Accepted: 10/25/2024] [Indexed: 11/02/2024] Open
Abstract
The term prebiotic has been used for almost 3 decades and has undergone numerous updates over the years. The scientific literature reveals that despite continuous efforts to establish a globally unified definition to guide jurisdictional regulations and product innovations, ambiguity continues to surround the terms prebiotic and prebiotic effect, leading to products that lack in full regulatory adherence being marketed worldwide. Thus, to reflect the current state of scientific research and knowledge and for the continuous advancement of the category, an update to the current prebiotic definition is warranted. This update includes removing the term selectivity, considering additional locations of action besides the gut, highlighting prebiotic performance benefits such as cognitive and athletic, and providing a clear standalone definition for prebiotic effect. The Global Prebiotic Association (GPA) is a leading information and industry hub committed to raising awareness about prebiotics, their emerging and well-established health benefits, and prebiotic product integrity and efficacy. In this position paper, GPA builds on previous prebiotic definitions to propose the following expanded definition for prebiotic: "a compound or ingredient that is utilized by the microbiota producing a health or performance benefit." In addition to prebiotic, GPA also defines prebiotic effect as "a health or performance benefit that arises from alteration of the composition and/or activity of the microbiota, as a direct or indirect result of the utilization of a specific and well-defined compound or ingredient by microorganisms." With these 2 definitions, GPA aims to paint a clearer picture for the term prebiotic, and by incorporating an industry point of view, these updated definitions may be used alongside current scientific and regulatory perspectives to move the category forward.
Collapse
Affiliation(s)
- Edward C Deehan
- Department of Food Science and Technology, University of Nebraska, Lincoln, NE, United States; Nebraska Food for Health Center, University of Nebraska, Lincoln, NE, United States; Scientific & Technical Committee, Global Prebiotic Association, Chicago, IL, United States.
| | | | - Rhonda S Witwer
- Scientific & Technical Committee, Global Prebiotic Association, Chicago, IL, United States; ADM, Decatur, IL, United States
| | - Paula Guerra
- Scientific & Technical Committee, Global Prebiotic Association, Chicago, IL, United States; SGS Nutrasource, Guelph, Ontario, Canada.
| | - Tania John
- Scientific & Technical Committee, Global Prebiotic Association, Chicago, IL, United States; SGS Nutrasource, Guelph, Ontario, Canada
| | - Len Monheit
- Scientific & Technical Committee, Global Prebiotic Association, Chicago, IL, United States; Global Prebiotic Association/Industry Transparency Center, Chicago, IL, United States
| |
Collapse
|
14
|
Singarayar MS, Chandrasekaran A, Balasundaram D, Veerasamy V, Neethirajan V, Thilagar S. Prebiotics: Comprehensive analysis of sources, structural characteristics and mechanistic roles in disease regulation. Microb Pathog 2024; 197:107071. [PMID: 39447658 DOI: 10.1016/j.micpath.2024.107071] [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: 06/27/2024] [Revised: 10/16/2024] [Accepted: 10/21/2024] [Indexed: 10/26/2024]
Abstract
Prebiotics are nondigestible components that comprise short-chain carbohydrates, primarily oligosaccharides, which are converted into beneficial compounds by probiotics. Various plant substances with prebiotic properties provide substantial health benefits and are used to prevent different diseases and for medical and clinical applications. Consuming prebiotics gives impeccable benefits since it aids in gut microbial balance. Prebiotic research is primarily concerned with the influence of intestinal disorders. The proposed review will describe recent data on the sources, structures, implementation of prebiotics and potential mechanisms in preventing and treating various disorders, with an emphasis on the gut microbiome. Prebiotics have a distinctive impact on the gastro intestine by explicitly encouraging the growth of probiotic organisms like Bifidobacteria and Lactobacilli. This in turn augments the body's inherent ability to fend off harmful pathogens. Prebiotic carbohydrates may also provide other non-specific advantages due to their fermentation in the large intestine. Additional in vivo research is needed to fully comprehend the interactions between prebiotics and probiotics ingested by hosts to improve their nutritional and therapeutic benefits.
Collapse
Affiliation(s)
- Magdalin Sylvia Singarayar
- Department of Environmental Biotechnology, Bharathidasan University, Tiruchirappalli, Tamilnadu, 620024, India.
| | - Ajithan Chandrasekaran
- Department of Horticulture, Chungnam National University, Daejeon, 34134, Republic of Korea.
| | | | - Veeramurugan Veerasamy
- Department of Environmental Biotechnology, Bharathidasan University, Tiruchirappalli, Tamilnadu, 620024, India.
| | - Vivek Neethirajan
- Department of Environmental Biotechnology, Bharathidasan University, Tiruchirappalli, Tamilnadu, 620024, India.
| | - Sivasudha Thilagar
- Department of Environmental Biotechnology, Bharathidasan University, Tiruchirappalli, Tamilnadu, 620024, India.
| |
Collapse
|
15
|
Basnet J, Eissa MA, Cardozo LLY, Romero DG, Rezq S. Impact of Probiotics and Prebiotics on Gut Microbiome and Hormonal Regulation. GASTROINTESTINAL DISORDERS 2024; 6:801-815. [PMID: 39649015 PMCID: PMC11623347 DOI: 10.3390/gidisord6040056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/10/2024] Open
Abstract
The gut microbiome plays a crucial role in human health by influencing various physiological functions through complex interactions with the endocrine system. These interactions involve the production of metabolites, signaling molecules, and direct communication with endocrine cells, which modulate hormone secretion and activity. As a result, the microbiome can exert neuroendocrine effects and contribute to metabolic regulation, adiposity, and appetite control. Additionally, the gut microbiome influences reproductive health by altering levels of sex hormones such as estrogen and testosterone, potentially contributing to conditions like polycystic ovary syndrome (PCOS) and hypogonadism. Given these roles, targeting the gut microbiome offers researchers and clinicians novel opportunities to improve overall health and well-being. Probiotics, such as Lactobacillus and Bifidobacterium, are live beneficial microbes that help maintain gut health by balancing the microbiota. Prebiotics, non-digestible fibers, nourish these beneficial bacteria, promoting their growth and activity. When combined, probiotics and prebiotics form synbiotics, which work synergistically to enhance the gut microbiota balance and improve metabolic, immune, and hormonal health. This integrated approach shows promising potential for managing conditions related to hormonal imbalances, though further research is needed to fully understand their specific mechanisms and therapeutic potential.
Collapse
Affiliation(s)
- Jelina Basnet
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS 39216, USA
- Mississippi Center of Excellence in Perinatal Research, University of Mississippi Medical Center, Jackson, MS 39216, USA
- Women’s Health Research Center, University of Mississippi Medical Center, Jackson, MS 39216, USA
- Cardiovascular-Renal Research Center, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Manar A. Eissa
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS 39216, USA
- Mississippi Center of Excellence in Perinatal Research, University of Mississippi Medical Center, Jackson, MS 39216, USA
- Women’s Health Research Center, University of Mississippi Medical Center, Jackson, MS 39216, USA
- Cardiovascular-Renal Research Center, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Licy L. Yanes Cardozo
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS 39216, USA
- Mississippi Center of Excellence in Perinatal Research, University of Mississippi Medical Center, Jackson, MS 39216, USA
- Women’s Health Research Center, University of Mississippi Medical Center, Jackson, MS 39216, USA
- Cardiovascular-Renal Research Center, University of Mississippi Medical Center, Jackson, MS 39216, USA
- Department of Medicine, University of Mississippi Medical Center, 2500 N. State Street, Jackson, MS 39216, USA
| | - Damian G. Romero
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS 39216, USA
- Mississippi Center of Excellence in Perinatal Research, University of Mississippi Medical Center, Jackson, MS 39216, USA
- Women’s Health Research Center, University of Mississippi Medical Center, Jackson, MS 39216, USA
- Cardiovascular-Renal Research Center, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Samar Rezq
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS 39216, USA
- Mississippi Center of Excellence in Perinatal Research, University of Mississippi Medical Center, Jackson, MS 39216, USA
- Women’s Health Research Center, University of Mississippi Medical Center, Jackson, MS 39216, USA
- Cardiovascular-Renal Research Center, University of Mississippi Medical Center, Jackson, MS 39216, USA
| |
Collapse
|
16
|
Salahlou M, Hajimohammadi A, Nazifi S, Rowshan-Ghasrodashti A, Nikzad M, Mirzaei A. Effects of probiotic and yeast extract supplement on liver functionality index and metabolic parameters in transition period of dairy cattle. Vet J 2024; 309:106280. [PMID: 39617095 DOI: 10.1016/j.tvjl.2024.106280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 07/09/2024] [Accepted: 11/26/2024] [Indexed: 12/06/2024]
Abstract
This research sought to evaluate the potential effects of probiotics and yeast cell wall (YCW) supplements on the liver functionality index (LFI) and metabolic parameters of dairy cattle throughout the transitional period. A cohort of forty dry cows was randomly divided into four groups, namely the probiotic group (Pr) receiving a basal diet combined with a blend of Bacillus subtilis, Bacillus lechiniformis, Streptococcus Thermophilis, and Enterococcus faecium; the YCW group receiving a basal diet enriched with Saccharomyces cerevisiae; the probiotic and yeast cell wall extract group (P & Y) receiving a basal diet supplemented with a mixture of probiotic and yeast cell wall extract; and the control group adhering to the basal diet. The intervention was initiated 21 days before calving and persisted until 28 days post-calving, except for the control group. The study entailed the collection of blood samples at four sampling times, encompassing 21 days preceding calving, seven days before calving, seven days post-calving, and four weeks post-calving. Multiple biochemical parameters were assessed, including urea, blood urea nitrogen (BUN), Gamma-glutamyl transferase (GGT), total bilirubin (TB), albumin, total protein (TP), globulin, glucose, triglyceride, cholesterol, and liver functionality index. The results showed that the Pr group exhibited reduced average levels of GGT and glucose compared to the control group (P < 0.05). Similarly, the P & Y group demonstrated lower average BUN, TB, and cholesterol levels than the control (P < 0.05). Notably, the LFI exhibited a discernible trend towards elevation in the Pr group compared to the control group (P = 0.007) and the P & Y group (P = 0.007). In essence, supplementation of YCW and probiotics is associated with advantageous effects on metabolic parameters and liver function.
Collapse
Affiliation(s)
- Mohammad Salahlou
- Resident of Large Animal Internal Medicine, Department of Clinical Sciences, School of Veterinary Medicine, Shiraz University, Shiraz, Iran
| | - Ali Hajimohammadi
- Associate Professor of Large Animal Internal Medicine, Department of Clinical Sciences, School of Veterinary Medicine, Shiraz University, Shiraz, Iran.
| | - Saeed Nazifi
- Professor of Clinical Pathology, Department of Clinical Sciences, School of Veterinary Medicine, Shiraz University, Shiraz, Iran
| | - Abbas Rowshan-Ghasrodashti
- Assistant Professor of Large Animal Internal Medicine, Department of Clinical Studies, School of Veterinary Medicine, Islamic Azad University, Kazerun Branch, Kazerun, Iran
| | - Mohammad Nikzad
- Resident of Large Animal Internal Medicine, Department of Clinical Sciences, School of Veterinary Medicine, Shiraz University, Shiraz, Iran
| | - Ahmadreza Mirzaei
- Post-doctoral associate at College of Veterinary Medicine, University of Florida, FL 32610, USA
| |
Collapse
|
17
|
Bhatnagar K, Jha K, Dalal N, Patki N, Gupta G, Kumar A, Kumar A, Chaudhary S. Exploring micronutrients and microbiome synergy: pioneering new paths in cancer therapy. Front Immunol 2024; 15:1442788. [PMID: 39676876 PMCID: PMC11638209 DOI: 10.3389/fimmu.2024.1442788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Accepted: 11/18/2024] [Indexed: 12/17/2024] Open
Abstract
The human microbiome is the complex ecosystem consisting of trillions of microorganisms that play a key role in developing the immune system and nutrient metabolism. Alterations in the gut microbiome have been linked to cancer initiation, progression, metastasis, and response to treatment. Accumulating evidence suggests that levels of vitamins and minerals influence the gut environment and may have implications for cancer risk and progression. Bifidobacterium has been reported to reduce the colorectal cancer risk by binding to free iron. Additionally, zinc ions have been shown to activate the immune cells and enhance the effectiveness of immunotherapy. Higher selenium levels have been associated with a reduced risk of several cancers, including colorectal cancer. In contrast, enhanced copper uptake has been implicated in promoting cancer progression, including colon cancer. The interaction between cancer and gut bacteria, as well as dysbiosis impact has been studied in animal models. The interplay between prebiotics, probiotics, synbiotics, postbiotics and gut bacteria in cancer offers the diverse physiological benefits. We also explored the particular probiotic formulations like VSL#3, Prohep, Lactobacillus rhamnosus GG (LGG), etc., for their ability to modulate immune responses and reduce tumor burden in preclinical models. Targeting the gut microbiome through antibiotics, bacteriophage, microbiome transplantation-based therapies will offer a new perspective in cancer research. Hence, to understand this interplay, we outline the importance of micronutrients with an emphasis on the immunomodulatory function of the microbiome and highlight the microbiome's potential as a target for precision medicine in cancer treatment.
Collapse
Affiliation(s)
- Kartik Bhatnagar
- Department of Biotechnology, School of Engineering and Applied Sciences, Bennett University, Greater Noida, Uttar Pradesh, India
| | - Kanupriya Jha
- Department of Biotechnology, School of Engineering and Applied Sciences, Bennett University, Greater Noida, Uttar Pradesh, India
| | - Nishu Dalal
- Gene Regulation Laboratory, National Institute of Immunology, New Delhi, India
- Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
| | - Ninad Patki
- Department of Biotechnology, School of Engineering and Applied Sciences, Bennett University, Greater Noida, Uttar Pradesh, India
| | - Garima Gupta
- Biological Engineering and Sciences, Indian Institute of Technology Gandhinagar Palaj, Gandhinagar, Gujarat, India
| | - Amit Kumar
- Department of Biotechnology, School of Engineering and Applied Sciences, Bennett University, Greater Noida, Uttar Pradesh, India
| | - Anil Kumar
- Gene Regulation Laboratory, National Institute of Immunology, New Delhi, India
| | - Sarika Chaudhary
- Department of Biotechnology, School of Engineering and Applied Sciences, Bennett University, Greater Noida, Uttar Pradesh, India
| |
Collapse
|
18
|
Alhamlan FS, Albadawi IA, Al-Qahtani AA, Awartani KA, Obeid DA, Tulbah AM. Cervicovaginal and gastrointestinal microbiomes in gynecological cancers and their roles in therapeutic intervention. Front Microbiol 2024; 15:1489942. [PMID: 39664050 PMCID: PMC11631898 DOI: 10.3389/fmicb.2024.1489942] [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: 09/02/2024] [Accepted: 11/13/2024] [Indexed: 12/13/2024] Open
Abstract
Cancer remains a significant global health concern, and understanding factors that regulate cancer development is important. The microbiome, with its potential role in cancer development, progression, and treatment, has garnered increasing attention in recent years. The cervicovaginal and gastrointestinal microbiomes in females constitute complex biological ecosystems. Although the gut microbiome has been extensively studied, little is known about the cervicovaginal microbiome. The microbiome plays a crucial role in maintaining local microenvironments and tissue homeostasis, but dysbiosis can disrupt this fine balance and contribute to pathological ramifications leading to cancer. This review explores the current understanding of the microbiome's correlation with gynecological cancers and highlights the potential of microbiome-based interventions to improve outcomes in these cancers. In addition, this review underscores the gaps and limitations in the literature, such as findings in specific ethnicities compared with understudied ethnicities. In addition, discrepancies in molecular techniques and terminology (microbiome vs. microbiota) used in the literature are addressed. Emerging evidence linking gynecological cancers and dysbiosis underscores microbiota as a potential target for cancer prevention and therapy. Manipulating the microbiome, such as through the use of probiotics, prebiotics, antibiotics, or vaginal and fecal transplantation, has demonstrated benefits in the treatment of chronic and inflammatory conditions. Further translational research in this field is needed to integrate the benefits of beneficial microorganisms in the fight against gynecological cancers.
Collapse
Affiliation(s)
- Fatimah S. Alhamlan
- Department of Infection and Immunity, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
- College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
- Department of Pathology and Laboratory Medicine, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Ismail A. Albadawi
- College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
- Gynecology Oncology, Department of Obstetrics and Gynecology, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Ahmed A. Al-Qahtani
- Department of Infection and Immunity, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
- College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| | - Khalid A. Awartani
- College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
- Reproductive Medicine, Department of Obstetrics and Gynecology, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Dalia A. Obeid
- Department of Infection and Immunity, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
- Organ Transplant Center of Excellence, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Asma M. Tulbah
- Department of Pathology and Laboratory Medicine, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| |
Collapse
|
19
|
Hillerer KM, Gimsa U. Adult neurogenesis and the microbiota-gut-brain axis in farm animals: underestimated and understudied parameters for improving welfare in livestock farming. Front Neurosci 2024; 18:1493605. [PMID: 39664450 PMCID: PMC11631930 DOI: 10.3389/fnins.2024.1493605] [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: 09/09/2024] [Accepted: 11/05/2024] [Indexed: 12/13/2024] Open
Abstract
Welfare in commercial livestock farming is becoming increasingly important in current agriculture research. Unfortunately, there is a lack of understanding about the neuronal mechanisms that underlie well-being on an individual level. Neuroplasticity in the hippocampus, the subventricular zone (SVZ), the olfactory bulb (OB) and the hypothalamus may be essential regulatory components in the context of farm animal behaviour and welfare that may be altered by providing environmental enrichment (EE). The importance of pre-and probiotics as a form of EE and the microbiota-gut-brain axis (MGBA) has come under the spotlight in the last 20 years, particularly in the contexts of research into stress and of stress resilience. However, it could also be an important regulatory system for animal welfare in livestock farming. This review aims to present a brief overview of the effects of EE on physiology and behaviour in farm animals and briefly discusses literature on behavioural flexibility, as well as inter-individual stress-coping styles and their relationship to animal welfare. Most importantly, we will summarise the literature on different forms of neural plasticity in farm animals, focusing on neurogenesis in various relevant brain regions. Furthermore, we will provide a brief outlook connecting these forms of neuroplasticity, stress, EE, the MGBA and welfare measures in modern livestock farming, concentrating on pigs.
Collapse
Affiliation(s)
- Katharina M. Hillerer
- Research Institute for Farm Animal Biology (FBN), Dummerstorf, Mecklenburg-Vorpommern, Germany
| | | |
Collapse
|
20
|
Saleem J, Zakar R, Iqbal S, Arshad M, Shahzad R, Batool M, Nawaz M, Butt MS, Fischer F. Effects of prebiotics on microbial diversity and abundance in young children with acute malnutrition: study protocol for a multi-centered, double-blinded randomized controlled trial. Trials 2024; 25:798. [PMID: 39593072 PMCID: PMC11590257 DOI: 10.1186/s13063-024-08647-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Accepted: 11/21/2024] [Indexed: 11/28/2024] Open
Abstract
BACKGROUND The anti-inflammatory and antimicrobial benefits of prebiotics may present an affordable and cost-effective strategy for not only the prevention but also treatment of malnutrition. Therefore, the present trial has been designed with the aim to evaluate the role of prebiotics on the gut microbiome of severe acute malnourished (SAM) children. METHODS The study is designed as a prospective, double-blinded, triple-armed, multi-centered randomized controlled trial, with 6-59 months old uncomplicated SAM children recruited to the experimental group receiving ready-to-use therapeutic food (RUTF) plus prebiotics and the active comparator group receiving RUTF plus starch for 2 months duration (8 weeks). Healthy children with matching age and gender will be recruited to placebo comparator group and will receive starch as a placebo during the study period. A total of 58 participants will be recruited to each arm with 1:1:1 allocation ratio following a pre-defined inclusion and exclusion criteria. The results of the gut microbiome diversity will serve as the primary outcome, while weight-for-height/length z-score, mid-upper-arm circumference, neurodevelopment assessment, and body mass accumulation will serve as the secondary outcome. Data collection and evaluations will be conducted at baseline and at the end of the trial (week 8), while the safety monitoring will be conducted at every second week. For analysis, the principles of intention-to-treat will be followed. CONCLUSIONS Conclusively, the results of the present trial would provide useful insights and high-quality data for the treatment and management of SAM children by evaluating the effect of RUTF plus prebiotic on the gut microbiome diversity of children, leading to medical evidence for designing the large-scale studies. TRIAL REGISTRATION The present trial is registered at ClinicalTrials.gov with identifier No: NCT06155474 and registration date 4 December 2023.
Collapse
Affiliation(s)
- Javeria Saleem
- Department of Public Health, University of the Punjab, Lahore, Pakistan
| | - Rubeena Zakar
- Department of Public Health, University of the Punjab, Lahore, Pakistan
| | - Sanaullah Iqbal
- Department of Food Science and Human Nutrition, University of Veterinary and Animal Sciences, Lahore, Pakistan
| | - Muhammad Arshad
- Centre for Genomics and Systems Biology, New York University Abu Dhabi, Abu Dhabi, United Arab Emirates
| | - Ruhma Shahzad
- Department of Public Health, University of the Punjab, Lahore, Pakistan
| | - Munazza Batool
- Department of Public Health, University of the Punjab, Lahore, Pakistan
| | - Muhammad Nawaz
- Institute of Microbiology, University of Veterinary and Animal Sciences, Lahore, Pakistan
| | | | - Florian Fischer
- Institute of Public Health, Charité - Universitätsmedizin Berlin, Berlin, Germany.
| |
Collapse
|
21
|
Ayyanar MP, Vijayan M. A review on gut microbiota and miRNA crosstalk: implications for Alzheimer's disease. GeroScience 2024:10.1007/s11357-024-01432-5. [PMID: 39562408 DOI: 10.1007/s11357-024-01432-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Accepted: 11/07/2024] [Indexed: 11/21/2024] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder characterized by cognitive decline and progressive neuronal damage. Recent research has highlighted the significant roles of the gut microbiota and microRNAs (miRNAs) in the pathogenesis of AD. This review explores the intricate interaction between gut microbiota and miRNAs, emphasizing their combined impact on Alzheimer's progression. First, we discuss the bidirectional communication within the gut-brain axis and how gut dysbiosis contributes to neuroinflammation and neurodegeneration in AD. Changes in gut microbiota composition in Alzheimer's patients have been linked to inflammation, which exacerbates disease progression. Next, we delve into the biology of miRNAs, focusing on their roles in gene regulation, neurodevelopment, and neurodegeneration. Dysregulated miRNAs are implicated in AD pathogenesis, influencing key processes like inflammation, tau pathology, and amyloid deposition. We then examine how the gut microbiota modulates miRNA expression, particularly in the brain, potentially altering neuroinflammatory responses and synaptic plasticity. The interplay between gut microbiota and miRNAs also affects blood-brain barrier integrity, further contributing to Alzheimer's pathology. Lastly, we explore therapeutic strategies targeting this gut microbiota-miRNA axis, including probiotics, prebiotics, and dietary interventions, aiming to modulate miRNA expression and improve AD outcomes. While promising, challenges remain in fully elucidating these interactions and translating them into effective therapies. This review highlights the importance of understanding the gut microbiota-miRNA relationship in AD, offering potential pathways for novel therapeutic approaches aimed at mitigating the disease's progression.
Collapse
Affiliation(s)
- Maruthu Pandian Ayyanar
- Department of Biology, The Gandhigram Rural Institute (Deemed to be University), Gandhigram, 624302, Tamil Nadu, India
| | - Murali Vijayan
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, 79430, USA.
| |
Collapse
|
22
|
Alexandrescu L, Suceveanu AP, Stanigut AM, Tofolean DE, Axelerad AD, Iordache IE, Herlo A, Nelson Twakor A, Nicoara AD, Tocia C, Dumitru A, Dumitru E, Condur LM, Aftenie CF, Tofolean IT. Intestinal Insights: The Gut Microbiome's Role in Atherosclerotic Disease: A Narrative Review. Microorganisms 2024; 12:2341. [PMID: 39597729 PMCID: PMC11596410 DOI: 10.3390/microorganisms12112341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Revised: 11/13/2024] [Accepted: 11/14/2024] [Indexed: 11/29/2024] Open
Abstract
Recent advances have highlighted the gut microbiota as a significant contributor to the development and progression of atherosclerosis, which is an inflammatory cardiovascular disease (CVD) characterized by plaque buildup within arterial walls. The gut microbiota, consisting of a diverse collection of microorganisms, impacts the host's metabolism, immune responses, and lipid processing, all of which contribute to atherosclerosis. This review explores the complex mechanisms through which gut dysbiosis promotes atherogenesis. We emphasize the potential of integrating microbiota modulation with traditional cardiovascular care, offering a holistic approach to managing atherosclerosis. Important pathways involve the translocation of inflammatory microbial components, modulation of lipid metabolism through metabolites such as trimethylamine-N-oxide (TMAO), and the production of short-chain fatty acids (SCFAs) that influence vascular health. Studies reveal distinct microbial profiles in atherosclerosis patients, with increased pathogenic bacteria (Megamonas, Veillonella, Streptococcus) and reduced anti-inflammatory genera (Bifidobacterium, Roseburia), highlighting the potential of these profiles as biomarkers and therapeutic targets. Probiotics are live microorganisms that have health benefits on the host. Prebiotics are non-digestible dietary fibers that stimulate the growth and activity of beneficial gut bacteria. Interventions targeting microbiota, such as probiotics, prebiotics, dietary modifications, and faecal microbiota transplantation (FMT), present effective approaches for restoring microbial equilibrium and justifying cardiovascular risk. Future research should focus on longitudinal, multi-omics studies to clarify causal links and refine therapeutic applications.
Collapse
Affiliation(s)
- Luana Alexandrescu
- Gastroenterology Department, “Sf. Apostol Andrei” Emergency County Hospital, 145 Tomis Blvd., 900591 Constanta, Romania; (L.A.); (C.T.); (A.D.); (E.D.); (I.T.T.)
- Medicine Faculty, “Ovidius” University of Constanta, 1 Universitatii Street, 900470 Constanta, Romania; (A.M.S.); (D.E.T.); (A.D.A.); (A.D.N.); (L.M.C.); (C.F.A.)
| | - Adrian Paul Suceveanu
- Gastroenterology Department, “Sf. Apostol Andrei” Emergency County Hospital, 145 Tomis Blvd., 900591 Constanta, Romania; (L.A.); (C.T.); (A.D.); (E.D.); (I.T.T.)
- Medicine Faculty, “Ovidius” University of Constanta, 1 Universitatii Street, 900470 Constanta, Romania; (A.M.S.); (D.E.T.); (A.D.A.); (A.D.N.); (L.M.C.); (C.F.A.)
| | - Alina Mihaela Stanigut
- Medicine Faculty, “Ovidius” University of Constanta, 1 Universitatii Street, 900470 Constanta, Romania; (A.M.S.); (D.E.T.); (A.D.A.); (A.D.N.); (L.M.C.); (C.F.A.)
- Nephrology Department, “Sf. Apostol Andrei” Emergency County Hospital, 145 Tomis Blvd., 900591 Constanta, Romania
| | - Doina Ecaterina Tofolean
- Medicine Faculty, “Ovidius” University of Constanta, 1 Universitatii Street, 900470 Constanta, Romania; (A.M.S.); (D.E.T.); (A.D.A.); (A.D.N.); (L.M.C.); (C.F.A.)
- Pneumology Department, Sf. Apostol Andrei” Emergency County Hospital, 145 Tomis Blvd., 900591 Constanta, Romania
| | - Ani Docu Axelerad
- Medicine Faculty, “Ovidius” University of Constanta, 1 Universitatii Street, 900470 Constanta, Romania; (A.M.S.); (D.E.T.); (A.D.A.); (A.D.N.); (L.M.C.); (C.F.A.)
| | - Ionut Eduard Iordache
- Department of General Surgery, “Sf. Apostol Andrei” Emergency County Hospital, 145 Tomis Blvd., 900591 Constanta, Romania;
| | - Alexandra Herlo
- Department XIII, Discipline of Infectious Diseases, “Victor Babes” University of Medicine and Pharmacy Timisoara, 2 Eftimie Murgu Square, 300041 Timisoara, Romania;
| | - Andreea Nelson Twakor
- Internal Medicine Department, Sf. Apostol Andrei” Emergency County Hospital, 145 Tomis Blvd., 900591 Constanta, Romania;
| | - Alina Doina Nicoara
- Medicine Faculty, “Ovidius” University of Constanta, 1 Universitatii Street, 900470 Constanta, Romania; (A.M.S.); (D.E.T.); (A.D.A.); (A.D.N.); (L.M.C.); (C.F.A.)
- Internal Medicine Department, Sf. Apostol Andrei” Emergency County Hospital, 145 Tomis Blvd., 900591 Constanta, Romania;
| | - Cristina Tocia
- Gastroenterology Department, “Sf. Apostol Andrei” Emergency County Hospital, 145 Tomis Blvd., 900591 Constanta, Romania; (L.A.); (C.T.); (A.D.); (E.D.); (I.T.T.)
- Medicine Faculty, “Ovidius” University of Constanta, 1 Universitatii Street, 900470 Constanta, Romania; (A.M.S.); (D.E.T.); (A.D.A.); (A.D.N.); (L.M.C.); (C.F.A.)
| | - Andrei Dumitru
- Gastroenterology Department, “Sf. Apostol Andrei” Emergency County Hospital, 145 Tomis Blvd., 900591 Constanta, Romania; (L.A.); (C.T.); (A.D.); (E.D.); (I.T.T.)
| | - Eugen Dumitru
- Gastroenterology Department, “Sf. Apostol Andrei” Emergency County Hospital, 145 Tomis Blvd., 900591 Constanta, Romania; (L.A.); (C.T.); (A.D.); (E.D.); (I.T.T.)
- Medicine Faculty, “Ovidius” University of Constanta, 1 Universitatii Street, 900470 Constanta, Romania; (A.M.S.); (D.E.T.); (A.D.A.); (A.D.N.); (L.M.C.); (C.F.A.)
- Academy of Romanian Scientist, 3 Ilfov Street, 050044 Bucharest, Romania
| | - Laura Maria Condur
- Medicine Faculty, “Ovidius” University of Constanta, 1 Universitatii Street, 900470 Constanta, Romania; (A.M.S.); (D.E.T.); (A.D.A.); (A.D.N.); (L.M.C.); (C.F.A.)
| | - Cristian Florentin Aftenie
- Medicine Faculty, “Ovidius” University of Constanta, 1 Universitatii Street, 900470 Constanta, Romania; (A.M.S.); (D.E.T.); (A.D.A.); (A.D.N.); (L.M.C.); (C.F.A.)
| | - Ioan Tiberiu Tofolean
- Gastroenterology Department, “Sf. Apostol Andrei” Emergency County Hospital, 145 Tomis Blvd., 900591 Constanta, Romania; (L.A.); (C.T.); (A.D.); (E.D.); (I.T.T.)
- Medicine Faculty, “Ovidius” University of Constanta, 1 Universitatii Street, 900470 Constanta, Romania; (A.M.S.); (D.E.T.); (A.D.A.); (A.D.N.); (L.M.C.); (C.F.A.)
| |
Collapse
|
23
|
Wang R, Yu Y, Yu W, Sun S, Lei Y, Li Y, Lu C, Zhai J, Bai F, Ren F, Huang J, Chen J. Roles of Probiotics, Prebiotics, and Postbiotics in B-cell mediated Immune Regulation. J Nutr 2024:S0022-3166(24)01178-7. [PMID: 39551357 DOI: 10.1016/j.tjnut.2024.11.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 10/29/2024] [Accepted: 11/13/2024] [Indexed: 11/19/2024] Open
Abstract
Probiotics, prebiotics, and postbiotics can significantly influence B-cell-related diseases through their immunomodulatory effects. They enhance the immune system's function, particularly affecting B cells, which originate in the bone marrow and are crucial for antibody production and immune memory. These substances have therapeutic potential in managing allergies, autoimmune diseases, and inflammatory conditions by regulating the gut microbiota, strengthening epithelial barriers, and directly interacting with various components of the innate and adaptive immune systems. The review highlights the critical need for further research into the precise mechanisms through which probiotics, prebiotics, and postbiotics modulate B cells. Gaining this understanding could facilitate the development of more effective treatments for B-cell-related diseases by harnessing the immunomodulatory properties of these dietary components.
Collapse
Affiliation(s)
- R Wang
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, China
| | - Yf Yu
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, China
| | - Wr Yu
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, China
| | - Sy Sun
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, China
| | - Ym Lei
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, China
| | - Yx Li
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, China
| | - Cx Lu
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, China
| | - Jn Zhai
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, China
| | - Fr Bai
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, China
| | - Fz Ren
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, China
| | - Jq Huang
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, China.
| | - J Chen
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, China.
| |
Collapse
|
24
|
Thacharodi A, Hassan S, Ahmed ZHT, Singh P, Maqbool M, Meenatchi R, Pugazhendhi A, Sharma A. The ruminant gut microbiome vs enteric methane emission: The essential microbes may help to mitigate the global methane crisis. ENVIRONMENTAL RESEARCH 2024; 261:119661. [PMID: 39043353 DOI: 10.1016/j.envres.2024.119661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 06/17/2024] [Accepted: 07/20/2024] [Indexed: 07/25/2024]
Abstract
Ruminants release enteric methane into the atmosphere, significantly increasing greenhouse gas emissions and degrading the environment. A common focus of traditional mitigation efforts is on dietary management and manipulation, which may have limits in sustainability and efficacy, exploring the potential of essential microorganisms as a novel way to reduce intestinal methane emissions in ruminants; a topic that has garnered increased attention in recent years. Fermentation and feed digestion are significantly aided by essential microbes found in the rumen, such as bacteria, fungi, and archaea. The practical implications of the findings reported in various studies conducted on rumen gut concerning methane emissions may pave the way to understanding the mechanisms of CH4 production in the rumen to enhance cattle feed efficiency and mitigate CH4 emissions from livestock. This review discussed using essential bacteria to reduce intestinal methane emissions in ruminants. It investigates how particular microbial strains or consortia can alter rumen fermentation pathways to lower methane output while preserving the health and productivity of animals. We also describe the role of probiotics and prebiotics in managing methane emissions using microbial feed additives. Further, recent studies involving microbial interventions have been discussed. The use of new methods involving functional metagenomics and meta-transcriptomics for exploring the rumen microbiome structure has been highlighted. This review also emphasizes the challenges faced in altering the gut microbiome and future directions in this area.
Collapse
Affiliation(s)
- Aswin Thacharodi
- Dr. Thacharodi's Laboratories, Department of Research and Development, Puducherry, 605005, India
| | - Saqib Hassan
- Department of Biotechnology, School of Bio and Chemical Engineering, Sathyabama Institute of Science and Technology, Chennai, Tamilnadu, 600119, India; Future Leaders Mentoring Fellow, American Society for Microbiology, Washington, 20036, USA
| | - Z H Tawfeeq Ahmed
- Department of Biotechnology, School of Bio and Chemical Engineering, Sathyabama Institute of Science and Technology, Chennai, Tamilnadu, 600119, India
| | - Prabhakar Singh
- Department of Biotechnology, School of Bio and Chemical Engineering, Sathyabama Institute of Science and Technology, Chennai, Tamilnadu, 600119, India
| | - Mohsin Maqbool
- Sidney Kimmel Cancer Center, Jefferson Health, Thomas Jefferson University, Philadelphia, PA, 19107, USA
| | - Ramu Meenatchi
- Department of Biotechnology, SRM Institute of Science and Technology, Chengalpattu, Tamil Nadu, 603203, India
| | - Arivalagan Pugazhendhi
- Tecnologico de Monterrey, Centre of Bioengineering, NatProLab, AgroInnovationLab, School of Engineering and Sciences, Queretaro, 76130, Mexico
| | - Ashutosh Sharma
- Tecnologico de Monterrey, Centre of Bioengineering, NatProLab, AgroInnovationLab, School of Engineering and Sciences, Queretaro, 76130, Mexico.
| |
Collapse
|
25
|
Kang JU, So YS, Kim G, Lee W, Seo DH, Shin H, Yoo SH. Efficient Biosynthesis of Theanderose, a Potent Prebiotic, Using Amylosucrase from Deinococcus deserti. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:25197-25209. [PMID: 39480747 PMCID: PMC11565756 DOI: 10.1021/acs.jafc.4c05763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 10/17/2024] [Accepted: 10/23/2024] [Indexed: 11/02/2024]
Abstract
The study aimed to develop an efficient bioprocess for the discovery and synthesis of theanderose by using amylosucrase from Deinococcus deserti (DdAS). An unknown trisaccharide produced by DdAS was detected by high-performance anion-exchange chromatography-pulsed amperometric detection and high-performance liquid chromatography-evaporative light scattering detection, purified using medium-pressure liquid chromatography, and identified as theanderose (α-d-glucopyranosyl-(1→6)-α-d-glucopyranosyl-(1→2)-β-d-fructofuranoside) through nuclear magnetic resonance and mass spectrometry. DdAS synthesized theanderose with a 25.4% yield (174.1 g/L) using 2.0 M sucrose at 40 °C for 96 h. In an in vitro digestion model, theanderose showed a 6.5% hydrolysis rate over 16 h. Prebiotic efficacy tests confirmed that theanderose significantly enhanced the proliferation of selected Bifidobacterium strains in the culturing medium with theanderose as the main carbon source. Subsequently, fecal fermentation was performed by adding theanderose to the feces of 20 individuals of varying ages to assess its effect on the gut microbiota. Theanderose increased the relative abundance of Bifidobacteriaceae and Prevotellaceae while decreasing the population ratio of Lachnospiraceae and Ruminococcaceae. Conclusively, theanderose displayed excellent prebiotic potential when judged by low digestibility and selective growth of beneficial microbes over harmful microbes.
Collapse
Affiliation(s)
- Jeon-Uk Kang
- Department of Food Science
and Biotechnology, and Carbohydrate Bioproduct Research Center, Sejong University, 209 Neungdong-ro, Gwangjin-gu, Seoul 05006, Republic of Korea
| | - Yun-Sang So
- Department of Food Science
and Biotechnology, and Carbohydrate Bioproduct Research Center, Sejong University, 209 Neungdong-ro, Gwangjin-gu, Seoul 05006, Republic of Korea
| | - Gyungcheon Kim
- Department of Food Science
and Biotechnology, and Carbohydrate Bioproduct Research Center, Sejong University, 209 Neungdong-ro, Gwangjin-gu, Seoul 05006, Republic of Korea
| | - WonJune Lee
- Department of Food Science
and Biotechnology, and Carbohydrate Bioproduct Research Center, Sejong University, 209 Neungdong-ro, Gwangjin-gu, Seoul 05006, Republic of Korea
| | - Dong-Ho Seo
- Department of Food Science
and Biotechnology, and Carbohydrate Bioproduct Research Center, Sejong University, 209 Neungdong-ro, Gwangjin-gu, Seoul 05006, Republic of Korea
| | - Hakdong Shin
- Department of Food Science
and Biotechnology, and Carbohydrate Bioproduct Research Center, Sejong University, 209 Neungdong-ro, Gwangjin-gu, Seoul 05006, Republic of Korea
| | - Sang-Ho Yoo
- Department of Food Science
and Biotechnology, and Carbohydrate Bioproduct Research Center, Sejong University, 209 Neungdong-ro, Gwangjin-gu, Seoul 05006, Republic of Korea
| |
Collapse
|
26
|
Kustrimovic N, Balkhi S, Bilato G, Mortara L. Gut Microbiota and Immune System Dynamics in Parkinson's and Alzheimer's Diseases. Int J Mol Sci 2024; 25:12164. [PMID: 39596232 PMCID: PMC11595203 DOI: 10.3390/ijms252212164] [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: 09/03/2024] [Revised: 10/25/2024] [Accepted: 10/27/2024] [Indexed: 11/28/2024] Open
Abstract
The gut microbiota, a diverse collection of microorganisms in the gastrointestinal tract, plays a critical role in regulating metabolic, immune, and cognitive functions. Disruptions in the composition of these microbial communities, termed dysbiosis, have been linked to various neurodegenerative diseases (NDs), such as Parkinson's disease (PD) and Alzheimer's disease (AD). One of the key pathological features of NDs is neuroinflammation, which involves the activation of microglia and peripheral immune cells. The gut microbiota modulates immune responses through the production of metabolites and interactions with immune cells, influencing the inflammatory processes within the central nervous system. This review explores the impact of gut dysbiosis on neuroinflammation, focusing on the roles of microglia, immune cells, and potential therapeutic strategies targeting the gut microbiota to alleviate neuroinflammatory processes in NDs.
Collapse
Affiliation(s)
- Natasa Kustrimovic
- Department of Biotechnology and Life Sciences, University of Insubria, 21100 Varese, Italy;
| | - Sahar Balkhi
- Immunology and General Pathology Laboratory, Department of Biotechnology and Life Sciences, University of Insubria, 21100 Varese, Italy; (S.B.); (G.B.)
| | - Giorgia Bilato
- Immunology and General Pathology Laboratory, Department of Biotechnology and Life Sciences, University of Insubria, 21100 Varese, Italy; (S.B.); (G.B.)
- Unit of Molecular Pathology, Biochemistry and Immunology, IRCCS MultiMedica, 20138 Milan, Italy
| | - Lorenzo Mortara
- Immunology and General Pathology Laboratory, Department of Biotechnology and Life Sciences, University of Insubria, 21100 Varese, Italy; (S.B.); (G.B.)
- Unit of Molecular Pathology, Biochemistry and Immunology, IRCCS MultiMedica, 20138 Milan, Italy
| |
Collapse
|
27
|
Gościniak A, Lainé E, Cielecka-Piontek J. How Do Cyclodextrins and Dextrans Affect the Gut Microbiome? Review of Prebiotic Activity. Molecules 2024; 29:5316. [PMID: 39598705 PMCID: PMC11596334 DOI: 10.3390/molecules29225316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 10/28/2024] [Accepted: 11/08/2024] [Indexed: 11/29/2024] Open
Abstract
The modulation of the gut microbiome through dietary components has garnered significant attention for its potential health benefits. Prebiotics, non-digestible food ingredients that promote the growth of beneficial gut bacteria, play a crucial role in maintaining gut health, enhancing immune function, and potentially preventing various metabolic and inflammatory disorders. This review explores the prebiotic activity of cyclodextrins and dextrans, focusing on their ability to influence gut microbiota composition and function. Both cyclodextrins and dextrans have demonstrated the capacity to promote the growth of beneficial bacterial populations, while also impacting short-chain fatty acid production, crucial for gut health.
Collapse
Affiliation(s)
- Anna Gościniak
- Department of Pharmacognosy and Biomaterials, Poznan University of Medical Sciences, Rokietnicka 3, 60-806 Poznan, Poland;
| | - Emmanuelle Lainé
- UMR 454 INRAe-UCA, Microbiology, Digestive Environment and Health (MEDIS), Université Clermont Auvergne, 63000 Clermont-Ferrand, France;
| | - Judyta Cielecka-Piontek
- Department of Pharmacognosy and Biomaterials, Poznan University of Medical Sciences, Rokietnicka 3, 60-806 Poznan, Poland;
| |
Collapse
|
28
|
Vásquez P, Stucken K, Garcia-Martin A, Ladero M, Bolivar JM, Bernal C. Enzymatic production, physicochemical characterization, and prebiotic potential of pectin oligosaccharides from pisco grape pomace. Int J Biol Macromol 2024; 281:136302. [PMID: 39370085 DOI: 10.1016/j.ijbiomac.2024.136302] [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: 07/02/2024] [Revised: 09/27/2024] [Accepted: 10/03/2024] [Indexed: 10/08/2024]
Abstract
The prebiotic capacity of Pectin Oligosaccharides (POS) is influenced by structural factors such as molecular size, composition, and degree of esterification, which affect their interaction with the gut microbiota. While existing literature has predominantly examined POS derived from apple and citrus pectins, the extrapolation of these findings to other pectin sources remains complex due to variations in their composition. This study focused on obtaining POS with prebiotic potential from pisco grape pomace through controlled enzymatic hydrolysis, resulting in three molecular size fractions: <3 kDa, 3-10 kDa, and > 10 kDa. The POS fractions were analyzed using FTIR, HPSEC, HPLC, and MALDI-TOF-MS techniques to characterize their physical-chemical properties. Each fraction presented distinct compositions, with the <3 kDa fraction showing a higher concentration of galacturonic acid and glucose, while the >10 kDa fraction was also composed of rhamnose and arabinose. Notably, the <3 kDa fraction supported greater biomass growth of the probiotic strain Lactobacillus casei ATCC 393 compared to the other fractions. In contrast, the non-probiotic strain Escherichia coli ATCC 25922 achieved the lowest biomass with this fraction. Consequently, the <3 kDa POS fraction exhibited the highest prebiotic index. This fraction, composed of oligomers from the rhamnogalacturonan region and arabino-oligosaccharides with a degree of polymerization between two and five, highlights its potential for further research and applications. Therefore, investigating other sources and optimizing extraction conditions could lead to developing novel prebiotic formulations that supply specific probiotic strains for a symbiotic product.
Collapse
Affiliation(s)
- Pamela Vásquez
- Departamento de Ingeniería en Alimentos, Universidad de La Serena, Chile; Laboratorio de Catálisis y Biocatálisis, Departamento de Química, Universidad de La Serena, Chile
| | - Karina Stucken
- Departamento de Ingeniería en Alimentos, Universidad de La Serena, Chile
| | - Alberto Garcia-Martin
- FQPIMA group, Chemical and Materials Engineering Department, Faculty of Chemistry, Complutense University of Madrid, Madrid 28040, Spain
| | - Miguel Ladero
- FQPIMA group, Chemical and Materials Engineering Department, Faculty of Chemistry, Complutense University of Madrid, Madrid 28040, Spain
| | - Juan M Bolivar
- FQPIMA group, Chemical and Materials Engineering Department, Faculty of Chemistry, Complutense University of Madrid, Madrid 28040, Spain
| | - Claudia Bernal
- Laboratorio de Catálisis y Biocatálisis, Departamento de Química, Universidad de La Serena, Chile.
| |
Collapse
|
29
|
Al-Wraikat M, Zhang L, Li L, Abubaker MA, Liu Y. Recent advances in wolfberry polysaccharides and whey protein-based biopolymers for regulating the diversity of gut microbiota and its mechanism: A review. Int J Biol Macromol 2024; 281:136401. [PMID: 39383924 DOI: 10.1016/j.ijbiomac.2024.136401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 09/11/2024] [Accepted: 10/05/2024] [Indexed: 10/11/2024]
Abstract
Imbalances in gut microbiota diversity are associated with various health issues, including obesity and related disorders. There is a growing interest in developing synergistic biopolymers based on wolfberry polysaccharides and whey protein to address these problems due to their potential health benefits. This review explores recent advances in understanding how functional foods based on Lycium barbarum polysaccharides (LBP) and whey protein (WP) influence gut microbiota diversity and their underlying mechanisms. We examine the impact of these biopolymers on microbial composition and functionality, focusing on their roles in improving health by regulating gut microbiota. The combined effects of WP and LBP significantly enhance gut microbiome metabolic activities and taxonomic diversity, offering promising avenues for treating obesity and related disorders.
Collapse
Affiliation(s)
- Majida Al-Wraikat
- College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710119, Shaanxi, China
| | - Lan Zhang
- College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710119, Shaanxi, China
| | - Linqiang Li
- College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710119, Shaanxi, China
| | - Mohamed Aamer Abubaker
- College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710119, Shaanxi, China
| | - Yongfeng Liu
- College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710119, Shaanxi, China.
| |
Collapse
|
30
|
Campagnoli LIM, Marchesi N, Varesi A, Morozzi M, Mascione L, Ricevuti G, Esposito C, Galeotti N, Pascale A. New therapeutic avenues in multiple sclerosis: Is there a place for gut microbiota-based treatments? Pharmacol Res 2024; 209:107456. [PMID: 39389400 DOI: 10.1016/j.phrs.2024.107456] [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: 08/15/2024] [Revised: 09/30/2024] [Accepted: 10/07/2024] [Indexed: 10/12/2024]
Abstract
The bidirectional interaction between the gut and the central nervous system (CNS), the so-called gut microbiota-brain axis, is reported to influence brain functions, thus having a potential impact on the development or the progression of several neurodegenerative disorders. Within this context, it has been documented that multiple sclerosis (MS), an autoimmune inflammatory, demyelinating, and neurodegenerative disease of the CNS, is associated with gastrointestinal symptoms, including constipation, dysphagia, and faecal incontinence. Moreover, some evidence suggests the existence of an altered gut microbiota (GM) composition in MS patients with respect to healthy individuals, as well as the potential influence of GM dysbiosis on typical MS features, including increased intestinal permeability, disruption of blood-brain barrier integrity, chronic inflammation, and altered T cells differentiation. Starting from these assumptions, the possible involvement of GM alteration in MS pathogenesis seems likely, and its restoration could represent a supplemental beneficial strategy against this disabling disease. In this regard, the present review will explore possible preventive approaches (including several dietary interventions, the administration of probiotics, prebiotics, synbiotics, and postbiotics, and the use of faecal microbiota transplantation) to be pursued as prophylaxis or in combination with pharmacological treatments with the aim of re-establishing a proper GM, thus helping to prevent the development of this disease or to manage it by alleviating symptoms or slowing down its progression.
Collapse
Affiliation(s)
| | - Nicoletta Marchesi
- Department of Drug Sciences, Section of Pharmacology, University of Pavia, Pavia, Italy.
| | - Angelica Varesi
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada; Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | - Martina Morozzi
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), Section of Pharmacology and Toxicology, University of Florence, Florence, Italy
| | - Linda Mascione
- Department of Drug Sciences, Section of Pharmacology, University of Pavia, Pavia, Italy
| | | | - Ciro Esposito
- Department of Internal Medicine and Therapeutics, University of Pavia, Italy; Nephrology and dialysis unit, ICS S. Maugeri SPA SB Hospital, Pavia, Italy; High School in Geriatrics, University of Pavia, Italy
| | - Nicoletta Galeotti
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), Section of Pharmacology and Toxicology, University of Florence, Florence, Italy
| | - Alessia Pascale
- Department of Drug Sciences, Section of Pharmacology, University of Pavia, Pavia, Italy.
| |
Collapse
|
31
|
Stanciu D, Staykov H, Dragomanova S, Tancheva L, Pop RS, Ielciu I, Crișan G. Gluten Unraveled: Latest Insights on Terminology, Diagnosis, Pathophysiology, Dietary Strategies, and Intestinal Microbiota Modulations-A Decade in Review. Nutrients 2024; 16:3636. [PMID: 39519469 PMCID: PMC11547711 DOI: 10.3390/nu16213636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 10/21/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024] Open
Abstract
A decade of research on gluten-related disorders (GRDs) is reviewed in this study, with a particular emphasis on celiac disease (CD) and non-celiac gluten sensitivity (NCGS). GRDs are triggered by the ingestion of gluten and gluten-like proteins found in wheat, barley, and rye. These proteins lead to intestinal damage in celiac disease, an autoimmune condition characterized by villous atrophy and a variety of gastrointestinal and extraintestinal symptoms. More enigmatic and less understood, NCGS involves symptoms similar to CD but without the immunological reaction or intestinal damage. Recent years have seen advances in the understanding of GRDs, particularly in connection to how intestinal microbiota influences disease progression and patient outcomes. The gluten-free diet (GFD) is still the standard therapy recommended for GRDs despite significant challenges, as discussed in this article. Precise diagnostic methods, patient education and dietary counseling are critical for improving patients' quality of life. The purpose of this review is to provide a more clear and up-to-date understanding of GRDs, and to help further research on this important topic.
Collapse
Affiliation(s)
- Dana Stanciu
- Department of Pharmaceutical Botany, Faculty of Pharmacy, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania; (D.S.); (I.I.); (G.C.)
| | - Hristian Staykov
- Department of Pharmacology and Toxicology, Faculty of Medicine, Medical University of Sofia, 1431 Sofia, Bulgaria
| | - Stela Dragomanova
- Department of Pharmacology, Toxicology and Pharmacotherapy, Faculty of Pharmacy, Medical University of Varna, 9000 Varna, Bulgaria;
| | - Lyubka Tancheva
- Institute of Neurobiology, Bulgarian Academy of Sciences, 1113 Sofia, Bulgaria;
| | - Radu Samuel Pop
- 3rd Department of Pediatrics, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400217 Cluj-Napoca, Romania;
| | - Irina Ielciu
- Department of Pharmaceutical Botany, Faculty of Pharmacy, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania; (D.S.); (I.I.); (G.C.)
| | - Gianina Crișan
- Department of Pharmaceutical Botany, Faculty of Pharmacy, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania; (D.S.); (I.I.); (G.C.)
| |
Collapse
|
32
|
Provitera L, Tomaselli A, Algieri F, Tripodi M, Raffaeli G, Amodeo I, Raymo L, Bronzoni CV, Fumagalli M, Garrido F, Cavallaro G. Gut Microbiota-Derived Metabolites and Their Role in the Pathogenesis of Necrotizing Enterocolitis in Preterm Infants: A Narrative Review. Metabolites 2024; 14:570. [PMID: 39590806 PMCID: PMC11596930 DOI: 10.3390/metabo14110570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Revised: 10/18/2024] [Accepted: 10/21/2024] [Indexed: 11/28/2024] Open
Abstract
Background: Necrotizing enterocolitis (NEC) is a severe gastrointestinal disease that occurs predominantly in premature infants and is characterized by the inflammation and necrosis of the intestine, showing high morbidity and mortality rates. Despite decades of research efforts, a specific treatment is currently lacking, and preventive strategies are the mainstays of care. This review aims to help understand the complex interplay between gut microbiota and their metabolites in NEC pathogenesis. In particular, we focused on how these factors can influence gut health, immune responses, and intestinal barrier integrity. Discussion: Current research has increasingly focused on the role of the gut microbiota and their metabolites in NEC pathogenesis, thanks to their involvement in modulating gut health, immune responses, and intestinal barrier integrity. Conclusions: A deeper understanding of the interplay between gut microbiota and their metabolites is essential for developing personalized strategies to prevent NEC. By targeting these microbial interactions, new therapeutic approaches may emerge that offer improved outcomes for preterm infants at a high risk of NEC.
Collapse
Affiliation(s)
- Livia Provitera
- Neonatal Intensive Care Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (A.T.); (M.T.); (G.R.); (I.A.); (L.R.); (C.V.B.); (M.F.); (G.C.)
| | - Andrea Tomaselli
- Neonatal Intensive Care Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (A.T.); (M.T.); (G.R.); (I.A.); (L.R.); (C.V.B.); (M.F.); (G.C.)
- Department of Clinical Sciences and Community Health, Università degli Studi di Milano, 20122 Milan, Italy
| | - Francesca Algieri
- Research and Development Unit, Postbiotica S.R.L., 20123 Milan, Italy;
| | - Matteo Tripodi
- Neonatal Intensive Care Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (A.T.); (M.T.); (G.R.); (I.A.); (L.R.); (C.V.B.); (M.F.); (G.C.)
| | - Genny Raffaeli
- Neonatal Intensive Care Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (A.T.); (M.T.); (G.R.); (I.A.); (L.R.); (C.V.B.); (M.F.); (G.C.)
| | - Ilaria Amodeo
- Neonatal Intensive Care Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (A.T.); (M.T.); (G.R.); (I.A.); (L.R.); (C.V.B.); (M.F.); (G.C.)
| | - Ludovica Raymo
- Neonatal Intensive Care Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (A.T.); (M.T.); (G.R.); (I.A.); (L.R.); (C.V.B.); (M.F.); (G.C.)
- Department of Clinical Sciences and Community Health, Università degli Studi di Milano, 20122 Milan, Italy
| | - Carolina Vittoria Bronzoni
- Neonatal Intensive Care Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (A.T.); (M.T.); (G.R.); (I.A.); (L.R.); (C.V.B.); (M.F.); (G.C.)
- Department of Clinical Sciences and Community Health, Università degli Studi di Milano, 20122 Milan, Italy
| | - Monica Fumagalli
- Neonatal Intensive Care Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (A.T.); (M.T.); (G.R.); (I.A.); (L.R.); (C.V.B.); (M.F.); (G.C.)
- Department of Clinical Sciences and Community Health, Università degli Studi di Milano, 20122 Milan, Italy
| | - Felipe Garrido
- Department of Pediatrics, Clínica Universidad de Navarra, 28027 Madrid, Spain;
| | - Giacomo Cavallaro
- Neonatal Intensive Care Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (A.T.); (M.T.); (G.R.); (I.A.); (L.R.); (C.V.B.); (M.F.); (G.C.)
| |
Collapse
|
33
|
Serrafi A, Chegdani F, Bennis F, Kepinska M. The Importance of Argan Oil in Medicine and Cosmetology. Nutrients 2024; 16:3573. [PMID: 39458566 PMCID: PMC11510224 DOI: 10.3390/nu16203573] [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: 08/29/2024] [Revised: 10/07/2024] [Accepted: 10/17/2024] [Indexed: 10/28/2024] Open
Abstract
Argan oil, rich in unsaturated fatty acids and polyphenols, exerts beneficial effects on both the intestinal and skin microbiotas. In the gut, it promotes the growth of beneficial bacteria, such as lactobacilli, while reducing pathogenic bacteria, due to its anti-inflammatory properties that help maintain microbial balance. Additionally, it improves the integrity of the intestinal mucosa, reducing the risk of dysbiosis. On the skin, argan oil hydrates and balances the lipid environment, creating a favorable setting for beneficial microorganisms, while also possessing antimicrobial and anti-inflammatory properties that soothe conditions like eczema and acne. Thus, argan oil is valuable for overall health, supporting digestion and skin health. The objective of this review is to provide a summary of the benefits of argan oil for alternative and complementary medicine. An exhaustive search of the literature was carried out using targeted keywords. A set of 83 articles were selected and analyzed. As the mechanisms of action of argan oil are not completely understood, this work highlighted the benefits of this oil by analyzing its nutritional properties and its beneficial effects on the intestinal and skin microbiotas. Indeed, argan oil is valuable for overall health.
Collapse
Affiliation(s)
- Agata Serrafi
- Department of Immunochemistry and Chemistry, Wroclaw Medical University, ul. M. Skłodowskiej-Curie 48/50, 50-369 Wroclaw, Poland;
| | - Fatima Chegdani
- Laboratory of Immunology and Biodiversity, Department of Biology, Faculty of Sciences Aïn Chock, Hassan II University of Casablanca, Route El Jadida, BP 5366 Maarif, Casablanca 20100, Morocco; (F.C.); (F.B.)
| | - Faïza Bennis
- Laboratory of Immunology and Biodiversity, Department of Biology, Faculty of Sciences Aïn Chock, Hassan II University of Casablanca, Route El Jadida, BP 5366 Maarif, Casablanca 20100, Morocco; (F.C.); (F.B.)
| | - Marta Kepinska
- Department of Pharmaceutical Biochemistry, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211a, 50-556 Wroclaw, Poland
| |
Collapse
|
34
|
Mousa WK, Al Ali A. The Gut Microbiome Advances Precision Medicine and Diagnostics for Inflammatory Bowel Diseases. Int J Mol Sci 2024; 25:11259. [PMID: 39457040 PMCID: PMC11508888 DOI: 10.3390/ijms252011259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 10/12/2024] [Accepted: 10/13/2024] [Indexed: 10/28/2024] Open
Abstract
The gut microbiome emerges as an integral component of precision medicine because of its signature variability among individuals and its plasticity, which enables personalized therapeutic interventions, especially when integrated with other multiomics data. This promise is further fueled by advances in next-generation sequencing and metabolomics, which allow in-depth high-precision profiling of microbiome communities, their genetic contents, and secreted chemistry. This knowledge has advanced our understanding of our microbial partners, their interaction with cellular targets, and their implication in human conditions such as inflammatory bowel disease (IBD). This explosion of microbiome data inspired the development of next-generation therapeutics for treating IBD that depend on manipulating the gut microbiome by diet modulation or using live products as therapeutics. The current landscape of artificial microbiome therapeutics is not limited to probiotics and fecal transplants but has expanded to include community consortia, engineered probiotics, and defined metabolites, bypassing several limitations that hindered rapid progress in this field such as safety and regulatory issues. More integrated research will reveal new therapeutic targets such as enzymes or receptors mediating interactions between microbiota-secreted molecules that drive or modulate diseases. With the shift toward precision medicine and the enhanced integration of host genetics and polymorphism in treatment regimes, the following key questions emerge: How can we effectively implement microbiomics to further personalize the treatment of diseases like IBD, leveraging proven and validated microbiome links? Can we modulate the microbiome to manage IBD by altering the host immune response? In this review, we discuss recent advances in understanding the mechanism underpinning the role of gut microbes in driving or preventing IBD. We highlight developed targeted approaches to reverse dysbiosis through precision editing of the microbiome. We analyze limitations and opportunities while defining the specific clinical niche for this innovative therapeutic modality for the treatment, prevention, and diagnosis of IBD and its potential implication in precision medicine.
Collapse
Affiliation(s)
- Walaa K. Mousa
- College of Pharmacy, Al Ain University of Science and Technology, Abu Dhabi 64141, United Arab Emirates;
- College of Pharmacy, Mansoura University, Mansoura 35516, Egypt
- AAU Health and Biomedical Research Center, Al Ain University, Abu Dhabi 112612, United Arab Emirates
| | - Aya Al Ali
- College of Pharmacy, Al Ain University of Science and Technology, Abu Dhabi 64141, United Arab Emirates;
- AAU Health and Biomedical Research Center, Al Ain University, Abu Dhabi 112612, United Arab Emirates
| |
Collapse
|
35
|
Popescu C, Munteanu C, Anghelescu A, Ciobanu V, Spînu A, Andone I, Mandu M, Bistriceanu R, Băilă M, Postoiu RL, Vlădulescu-Trandafir AI, Giuvara S, Malaelea AD, Onose G. Novelties on Neuroinflammation in Alzheimer's Disease-Focus on Gut and Oral Microbiota Involvement. Int J Mol Sci 2024; 25:11272. [PMID: 39457054 PMCID: PMC11508522 DOI: 10.3390/ijms252011272] [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: 09/23/2024] [Revised: 10/05/2024] [Accepted: 10/17/2024] [Indexed: 10/28/2024] Open
Abstract
Recent studies underscore the role of gut and oral microbiota in influencing neuroinflammation through the microbiota-gut-brain axis, including in Alzheimer's disease (AD). This review aims to provide a comprehensive synthesis of recent findings on the involvement of gut and oral microbiota in the neuroinflammatory processes associated with AD, emphasizing novel insights and therapeutic implications. This review reveals that dysbiosis in AD patients' gut and oral microbiota is linked to heightened peripheral and central inflammatory responses. Specific bacterial taxa, such as Bacteroides and Firmicutes in the gut, as well as Porphyromonas gingivalis in the oral cavity, are notably altered in AD, leading to significant changes in microglial activation and cytokine production. Gut microbiota alterations are associated with increased intestinal permeability, facilitating the translocation of endotoxins like lipopolysaccharides (LPS) into the bloodstream and exacerbating neuroinflammation by activating the brain's toll-like receptor 4 (TLR4) pathways. Furthermore, microbiota-derived metabolites, including short-chain fatty acids (SCFAs) and amyloid peptides, can cross the blood-brain barrier and modulate neuroinflammatory responses. While microbial amyloids may contribute to amyloid-beta aggregation in the brain, certain SCFAs like butyrate exhibit anti-inflammatory properties, suggesting a potential therapeutic avenue to mitigate neuroinflammation. This review not only highlights the critical role of microbiota in AD pathology but also offers a ray of hope by suggesting that modulating gut and oral microbiota could represent a novel therapeutic strategy for reducing neuroinflammation and slowing disease progression.
Collapse
Affiliation(s)
- Cristina Popescu
- Faculty of Medicine, University of Medicine and Pharmacy “Carol Davila”, 020022 Bucharest, Romania; (C.P.); (A.A.); (A.S.); (I.A.); (R.B.); (M.B.); (R.-L.P.); (A.-I.V.-T.); (S.G.); (A.-D.M.); (G.O.)
- Neuromuscular Rehabilitation Clinic Division, Clinical Emergency Hospital “Bagdasar-Arseni”, 041915 Bucharest, Romania
| | - Constantin Munteanu
- Neuromuscular Rehabilitation Clinic Division, Clinical Emergency Hospital “Bagdasar-Arseni”, 041915 Bucharest, Romania
- Department of Biomedical Sciences, Faculty of Medical Bioengineering, University of Medicine and Pharmacy “Grigore T. Popa” Iași, 700454 Iași, Romania
| | - Aurelian Anghelescu
- Faculty of Medicine, University of Medicine and Pharmacy “Carol Davila”, 020022 Bucharest, Romania; (C.P.); (A.A.); (A.S.); (I.A.); (R.B.); (M.B.); (R.-L.P.); (A.-I.V.-T.); (S.G.); (A.-D.M.); (G.O.)
- Neuromuscular Rehabilitation Clinic Division, Clinical Emergency Hospital “Bagdasar-Arseni”, 041915 Bucharest, Romania
| | - Vlad Ciobanu
- Department of Computer Science and Engineering, Faculty for Automatic Control and Computers, University Politehnica of Bucharest, 060042 Bucharest, Romania;
| | - Aura Spînu
- Faculty of Medicine, University of Medicine and Pharmacy “Carol Davila”, 020022 Bucharest, Romania; (C.P.); (A.A.); (A.S.); (I.A.); (R.B.); (M.B.); (R.-L.P.); (A.-I.V.-T.); (S.G.); (A.-D.M.); (G.O.)
- Neuromuscular Rehabilitation Clinic Division, Clinical Emergency Hospital “Bagdasar-Arseni”, 041915 Bucharest, Romania
| | - Ioana Andone
- Faculty of Medicine, University of Medicine and Pharmacy “Carol Davila”, 020022 Bucharest, Romania; (C.P.); (A.A.); (A.S.); (I.A.); (R.B.); (M.B.); (R.-L.P.); (A.-I.V.-T.); (S.G.); (A.-D.M.); (G.O.)
- Neuromuscular Rehabilitation Clinic Division, Clinical Emergency Hospital “Bagdasar-Arseni”, 041915 Bucharest, Romania
| | - Mihaela Mandu
- Faculty of Medicine, University of Medicine and Pharmacy “Carol Davila”, 020022 Bucharest, Romania; (C.P.); (A.A.); (A.S.); (I.A.); (R.B.); (M.B.); (R.-L.P.); (A.-I.V.-T.); (S.G.); (A.-D.M.); (G.O.)
- Neuromuscular Rehabilitation Clinic Division, Clinical Emergency Hospital “Bagdasar-Arseni”, 041915 Bucharest, Romania
| | - Roxana Bistriceanu
- Faculty of Medicine, University of Medicine and Pharmacy “Carol Davila”, 020022 Bucharest, Romania; (C.P.); (A.A.); (A.S.); (I.A.); (R.B.); (M.B.); (R.-L.P.); (A.-I.V.-T.); (S.G.); (A.-D.M.); (G.O.)
- Neuromuscular Rehabilitation Clinic Division, Clinical Emergency Hospital “Bagdasar-Arseni”, 041915 Bucharest, Romania
| | - Mihai Băilă
- Faculty of Medicine, University of Medicine and Pharmacy “Carol Davila”, 020022 Bucharest, Romania; (C.P.); (A.A.); (A.S.); (I.A.); (R.B.); (M.B.); (R.-L.P.); (A.-I.V.-T.); (S.G.); (A.-D.M.); (G.O.)
- Neuromuscular Rehabilitation Clinic Division, Clinical Emergency Hospital “Bagdasar-Arseni”, 041915 Bucharest, Romania
| | - Ruxandra-Luciana Postoiu
- Faculty of Medicine, University of Medicine and Pharmacy “Carol Davila”, 020022 Bucharest, Romania; (C.P.); (A.A.); (A.S.); (I.A.); (R.B.); (M.B.); (R.-L.P.); (A.-I.V.-T.); (S.G.); (A.-D.M.); (G.O.)
- Neuromuscular Rehabilitation Clinic Division, Clinical Emergency Hospital “Bagdasar-Arseni”, 041915 Bucharest, Romania
| | - Andreea-Iulia Vlădulescu-Trandafir
- Faculty of Medicine, University of Medicine and Pharmacy “Carol Davila”, 020022 Bucharest, Romania; (C.P.); (A.A.); (A.S.); (I.A.); (R.B.); (M.B.); (R.-L.P.); (A.-I.V.-T.); (S.G.); (A.-D.M.); (G.O.)
- Neuromuscular Rehabilitation Clinic Division, Clinical Emergency Hospital “Bagdasar-Arseni”, 041915 Bucharest, Romania
| | - Sebastian Giuvara
- Faculty of Medicine, University of Medicine and Pharmacy “Carol Davila”, 020022 Bucharest, Romania; (C.P.); (A.A.); (A.S.); (I.A.); (R.B.); (M.B.); (R.-L.P.); (A.-I.V.-T.); (S.G.); (A.-D.M.); (G.O.)
- Neuromuscular Rehabilitation Clinic Division, Clinical Emergency Hospital “Bagdasar-Arseni”, 041915 Bucharest, Romania
| | - Alin-Daniel Malaelea
- Faculty of Medicine, University of Medicine and Pharmacy “Carol Davila”, 020022 Bucharest, Romania; (C.P.); (A.A.); (A.S.); (I.A.); (R.B.); (M.B.); (R.-L.P.); (A.-I.V.-T.); (S.G.); (A.-D.M.); (G.O.)
- Neuromuscular Rehabilitation Clinic Division, Clinical Emergency Hospital “Bagdasar-Arseni”, 041915 Bucharest, Romania
| | - Gelu Onose
- Faculty of Medicine, University of Medicine and Pharmacy “Carol Davila”, 020022 Bucharest, Romania; (C.P.); (A.A.); (A.S.); (I.A.); (R.B.); (M.B.); (R.-L.P.); (A.-I.V.-T.); (S.G.); (A.-D.M.); (G.O.)
- Neuromuscular Rehabilitation Clinic Division, Clinical Emergency Hospital “Bagdasar-Arseni”, 041915 Bucharest, Romania
| |
Collapse
|
36
|
Zhao Z, Liswaniso S, Qin N, Cao S, Wu X, Ma C, Yan C, Xu R, Sun X. Effects of a novel synbiotics-enzyme complex as a replacement for antibiotics on growth performance, slaughter and meat characteristics, immune organ index, and intestinal morphology of broilers. Front Vet Sci 2024; 11:1468847. [PMID: 39484028 PMCID: PMC11524961 DOI: 10.3389/fvets.2024.1468847] [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: 07/22/2024] [Accepted: 09/23/2024] [Indexed: 11/03/2024] Open
Abstract
Introduction Antibiotic use in broilers is being discouraged globally due to the challenges it poses. This study was conducted to assess the effects of supplementing broilers with a Symbiotic-Enzyme complex (SEC) containing prebiotics (mannose oligosaccharides), probiotics (Clostridium butyricum and Bacillus subtilis), and enzymes (glucose oxidase, and α-galactosidase) as an alternative to antibiotics on growth performance, carcass and meat quality traits, mortality, linear body measurements, intestinal morphology and immune organ indexes. Method A total of 864 mixed-sex 1-day-old arbor acres (AA+) broilers were allocated to 8 experimental groups replicated 9 times with 12 chickens per replicate. These included 6 treatment groups with SEC inclusion levels of 0.025, 0.04, 0.05, 0.06, 0.08, and 0.10%, respectively, and two control groups: a negative control group containing a basal diet only and the positive control group (Antibiotics group) containing a basal diet and antibiotic oxytetracycline added at 0.2%. Growth performance was measured on day 21 and 42, and the mortality, carcass, meat quality traits, linear body measurements, intestinal morphology, and organ size indexes were measured on day 42. Results The results indicated that supplementing broilers with 0.1% SEC resulted in insignificant (P > 0.05) increases in average daily feed intake (ADFI), significant (P < 0.05) increases in the average daily gains (ADG), and significant (P < 0.05) reduction in a feed-to-gain ratio (F/G) in all the phases compared to the control and antibiotics groups. Supplementation of broilers with 0.1% SEC inclusion levels also significantly (P < 0.05) increased the body slope length, chest width, chest depth, keel length, and shank circumference. Furthermore, broilers on diets containing 0.1% SEC inclusion level also had significantly (P < 0.05) higher dressed, semi-evisceration, evisceration, and breast muscle percentages. Including SEC at 0.1% also significantly (P < 0.05) increased villus height and villus-to-crypt ratio (V/C) but reduced crypt depth in the duodenum, jejunum, and ileum compared to the control groups. SEC inclusion at 0.1% significantly (P < 0.05) increased the spleen, bursal, and thymus indexes, respectively. Conclusion Supplementation of broilers with 0.1% SEC can be used as an antibiotic alternative because it increases the F/G, improves the carcass and meat quality, increases the body conformation, improves the small intestines' functions, and immune organ size.
Collapse
Affiliation(s)
- Zihao Zhao
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
- Joint International Research Laboratory of Modern Agricultural Technology, Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Simushi Liswaniso
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
- Joint International Research Laboratory of Modern Agricultural Technology, Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Ning Qin
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
- Joint International Research Laboratory of Modern Agricultural Technology, Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Shengxiao Cao
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
- Joint International Research Laboratory of Modern Agricultural Technology, Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Xin Wu
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
- Joint International Research Laboratory of Modern Agricultural Technology, Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Chang Ma
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
- Joint International Research Laboratory of Modern Agricultural Technology, Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Chunchi Yan
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
- Joint International Research Laboratory of Modern Agricultural Technology, Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Rifu Xu
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
- Joint International Research Laboratory of Modern Agricultural Technology, Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Xue Sun
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
- Joint International Research Laboratory of Modern Agricultural Technology, Ministry of Education, Jilin Agricultural University, Changchun, China
| |
Collapse
|
37
|
Huang J, Xu T, Quan G, Li Y, Yang X, Xie W. Current progress on the microbial therapies for acute liver failure. Front Microbiol 2024; 15:1452663. [PMID: 39479215 PMCID: PMC11521890 DOI: 10.3389/fmicb.2024.1452663] [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: 08/29/2024] [Accepted: 09/30/2024] [Indexed: 11/02/2024] Open
Abstract
Acute liver failure (ALF), associated with a clinical fatality rate exceeding 80%, is characterized by severe liver damage resulting from various factors in the absence of pre-existing liver disease. The role of microbiota in the progression of diverse liver diseases, including ALF, has been increasingly recognized, with the interactions between the microbiota and the host significantly influencing both disease onset and progression. Despite growing interest in the microbiological aspects of ALF, comprehensive reviews remain limited. This review critically examines the mechanisms and efficacy of microbiota-based treatments for ALF, focusing on their role in prevention, treatment, and prognosis over the past decade.
Collapse
Affiliation(s)
- Jiayuan Huang
- Department of Gastroenterology, Research Center for Engineering Techniques of Microbiota-Targeted Therapies of Guangdong Province, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
| | - Tianyu Xu
- Department of Gastroenterology, Research Center for Engineering Techniques of Microbiota-Targeted Therapies of Guangdong Province, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
| | - Guoqiao Quan
- Department of Gastroenterology, Research Center for Engineering Techniques of Microbiota-Targeted Therapies of Guangdong Province, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
| | - Yuange Li
- Department of Gastroenterology, Research Center for Engineering Techniques of Microbiota-Targeted Therapies of Guangdong Province, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
| | - Xiaoya Yang
- Department of Physiology, Guangzhou Health Science College, Guangzhou, China
| | - Wenrui Xie
- Department of Gastroenterology, Research Center for Engineering Techniques of Microbiota-Targeted Therapies of Guangdong Province, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
| |
Collapse
|
38
|
Moon H, Kang K, Kim M. Potential Prebiotic Effects of Artemisia capillaris-Derived Transglycosylated Product. Foods 2024; 13:3267. [PMID: 39456329 PMCID: PMC11507088 DOI: 10.3390/foods13203267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 10/11/2024] [Accepted: 10/13/2024] [Indexed: 10/28/2024] Open
Abstract
This study investigated the impact of a transglycosylated product (ACOD) catalyzed by Leuconostoc mesenteroides MKSR dextransucrase using sucrose as a glucosyl donor and both maltose and Artemisia capillaris as acceptors on gut microbiota through fecal fermentation. ACOD promoted the growth of probiotics such as Lactiplantibacillus plantarum, Lacticaseibacillus casei, Lacticaseibacillus rhamnosus GG, and Leuconostoc mesenteroides MKSR, while inhibiting the growth of pathogenic bacteria such as Escherichia coli, E. coli O157:H7, Enterococcus faecalis, Listeria monocytogenes, Staphylococcus aureus, Shigella flexneri, Streptococcus mutans, Pseudomonas aeruginosa, and Bacillus cereus during independent cultivation. Fecal fermentation for 24 h revealed that ACOD significantly increased the production of short-chain fatty acids (SCFAs) compared to the blank and fructoooligosaccharide (FOS) groups. Specifically, ACOD led to a 4.5-fold increase in acetic acid production compared to FOSs and a 3.3-fold increase in propionic acid production. Both the ACOD and FOS groups exhibited higher levels of butyric acid than the blank. Notably, ACOD significantly modulated the composition of the gut microbiota by increasing the relative abundances of Lactobacillus and decreasing Escherichia/Shigella and Salmonella. In contrast, FOSs remarkably promoted the growth of Salmonella. These findings suggest that ACOD is a potential candidate for prebiotics that improve the intestinal environment by being actively used by beneficial bacteria.
Collapse
Affiliation(s)
- Heewon Moon
- Department of Food Science and Nutrition, Dankook University, Cheonan 31116, Republic of Korea;
| | - Keunsoo Kang
- Department of Microbiology, Dankook University, Cheonan 31116, Republic of Korea;
| | - Misook Kim
- Department of Food Science and Nutrition, Dankook University, Cheonan 31116, Republic of Korea;
| |
Collapse
|
39
|
Piccioni A, Spagnuolo F, Candelli M, Voza A, Covino M, Gasbarrini A, Franceschi F. The Gut Microbiome in Sepsis: From Dysbiosis to Personalized Therapy. J Clin Med 2024; 13:6082. [PMID: 39458032 PMCID: PMC11508704 DOI: 10.3390/jcm13206082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 09/21/2024] [Accepted: 10/09/2024] [Indexed: 10/28/2024] Open
Abstract
Sepsis is a complex clinical syndrome characterized by an uncontrolled inflammatory response to an infection that may result in septic shock and death. Recent research has revealed a crucial link between sepsis and alterations in the gut microbiota, showing that the microbiome could serve an essential function in its pathogenesis and prognosis. In sepsis, the gut microbiota undergoes significant dysbiosis, transitioning from a beneficial commensal flora to a predominance of pathobionts. This transformation can lead to a dysfunction of the intestinal barrier, compromising the host's immune response, which contributes to the severity of the disease. The gut microbiota is an intricate system of protozoa, fungi, bacteria, and viruses that are essential for maintaining immunity and metabolic balance. In sepsis, there is a reduction in microbial heterogeneity and a predominance of pathogenic bacteria, such as proteobacteria, which can exacerbate inflammation and negatively influence clinical outcomes. Microbial compounds, such as short-chain fatty acids (SCFAs), perform a crucial task in modulating the inflammatory response and maintaining intestinal barrier function. However, the role of other microbiota components, such as viruses and fungi, in sepsis remains unclear. Innovative therapeutic strategies aim to modulate the gut microbiota to improve the management of sepsis. These include selective digestive decontamination (SDD), probiotics, prebiotics, synbiotics, postbiotics, and fecal microbiota transplantation (FMT), all of which have shown potential, although variable, results. The future of sepsis management could benefit greatly from personalized treatment based on the microbiota. Rapid and easy-to-implement tests to assess microbiome profiles and metabolites associated with sepsis could revolutionize the disease's diagnosis and management. These approaches could not only improve patient prognosis but also reduce dependence on antibiotic therapies and promote more targeted and sustainable treatment strategies. Nevertheless, there is still limited clarity regarding the ideal composition of the microbiota, which should be further characterized in the near future. Similarly, the benefits of therapeutic approaches should be validated through additional studies.
Collapse
Affiliation(s)
- Andrea Piccioni
- Department of Emergency Medicine, Fondazione Policlinico Universitario Agostino Gemelli-IRCCS, 00168 Rome, Italy; (A.P.); (M.C.); (M.C.); (F.F.)
| | - Fabio Spagnuolo
- Faculty of Medicine and Surgery, Università Cattolica del Sacro Cuore, 00168 Rome, Italy;
| | - Marcello Candelli
- Department of Emergency Medicine, Fondazione Policlinico Universitario Agostino Gemelli-IRCCS, 00168 Rome, Italy; (A.P.); (M.C.); (M.C.); (F.F.)
| | - Antonio Voza
- Department of Emergency Medicine, IRCCS-Humanitas Research Hospital, Rozzano, 20089 Milan, Italy;
| | - Marcello Covino
- Department of Emergency Medicine, Fondazione Policlinico Universitario Agostino Gemelli-IRCCS, 00168 Rome, Italy; (A.P.); (M.C.); (M.C.); (F.F.)
- Faculty of Medicine and Surgery, Università Cattolica del Sacro Cuore, 00168 Rome, Italy;
| | - Antonio Gasbarrini
- Faculty of Medicine and Surgery, Università Cattolica del Sacro Cuore, 00168 Rome, Italy;
- Medical and Surgical Science Department, Fondazione Policlinico Universitario A. Gemelli-IRCCS, 00168 Rome, Italy
| | - Francesco Franceschi
- Department of Emergency Medicine, Fondazione Policlinico Universitario Agostino Gemelli-IRCCS, 00168 Rome, Italy; (A.P.); (M.C.); (M.C.); (F.F.)
- Faculty of Medicine and Surgery, Università Cattolica del Sacro Cuore, 00168 Rome, Italy;
| |
Collapse
|
40
|
Iatcu OC, Hamamah S, Covasa M. Harnessing Prebiotics to Improve Type 2 Diabetes Outcomes. Nutrients 2024; 16:3447. [PMID: 39458444 PMCID: PMC11510484 DOI: 10.3390/nu16203447] [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: 09/18/2024] [Revised: 10/08/2024] [Accepted: 10/09/2024] [Indexed: 10/28/2024] Open
Abstract
The gut microbiota, a complex ecosystem of microorganisms in the human gastrointestinal tract (GI), plays a crucial role in maintaining metabolic health and influencing disease susceptibility. Dysbiosis, or an imbalance in gut microbiota, has been linked to the development of type 2 diabetes mellitus (T2DM) through mechanisms such as reduced glucose tolerance and increased insulin resistance. A balanced gut microbiota, or eubiosis, is associated with improved glucose metabolism and insulin sensitivity, potentially reducing the risk of diabetes-related complications. Various strategies, including the use of prebiotics like inulin, fructooligosaccharides, galactooligosaccharides, resistant starch, pectic oligosaccharides, polyphenols, β-glucan, and Dendrobium officinale have been shown to improve gut microbial composition and support glycemic control in T2DM patients. These prebiotics can directly impact blood sugar levels while promoting the growth of beneficial bacteria, thus enhancing glycemic control. Studies have shown that T2DM patients often exhibit a decrease in beneficial butyrate-producing bacteria, like Roseburia and Faecalibacterium, and an increase in harmful bacteria, such as Escherichia and Prevotella. This review aims to explore the effects of different prebiotics on T2DM, their impact on gut microbiota composition, and the potential for personalized dietary interventions to optimize diabetes management and improve overall health outcomes.
Collapse
Affiliation(s)
- Oana C. Iatcu
- Department of Biomedical Sciences, College of Medicine and Biological Science, University of Suceava, 720229 Suceava, Romania;
| | - Sevag Hamamah
- Department of Basic Medical Sciences, College of Osteopathic Medicine, Western University of Health Sciences, Pomona, CA 91766, USA;
- Department of Internal Medicine, Scripps Mercy Hospital, San Diego, CA 92103, USA
| | - Mihai Covasa
- Department of Biomedical Sciences, College of Medicine and Biological Science, University of Suceava, 720229 Suceava, Romania;
- Department of Basic Medical Sciences, College of Osteopathic Medicine, Western University of Health Sciences, Pomona, CA 91766, USA;
| |
Collapse
|
41
|
Alswat AS. The Influence of the Gut Microbiota on Host Health: A Focus on the Gut-Lung Axis and Therapeutic Approaches. Life (Basel) 2024; 14:1279. [PMID: 39459579 PMCID: PMC11509314 DOI: 10.3390/life14101279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 10/01/2024] [Accepted: 10/03/2024] [Indexed: 10/28/2024] Open
Abstract
The human gut microbiota is a complex ecosystem harboring thousands of microbial strains that play a crucial role in maintaining the overall well-being of its host. The composition of the gut microbiota varies with age from infancy to adulthood and is influenced by dietary habits, environment, and genetic disposition. Recent advances in culture-independent techniques and nucleic acid sequencing have improved our understanding of the diversity of the gut microbiota. The microbial species present in the gut release short-chain fatty acids (SCFAs), which have anti-inflammatory properties. The gut microbiota also plays a substantial role in modulating the host's immune system, promoting immune tolerance, and maintaining homeostasis. The impact of the gut microbiota on the health of the host is quite evident, as gut dysbiosis has been linked to various diseases, including metabolic disorders, autoimmune diseases, allergies, and inflammatory bowel diseases. The gut microbiota has bidirectional communication with the respiratory system, creating the gut-lung axis, which has been associated with different respiratory diseases. Therapeutic approaches targeting the gut microbiota, such as probiotics, prebiotics, dietary interventions, and fecal microbiota transplantation (FMT), aim to restore microbial balance and promote the growth of beneficial strains in the gut. Nonetheless, gaining knowledge of the complex interactions between the gut microbiota and the host is necessary to develop personalized medicine approaches and microbiota-based therapies for various conditions. This review summarizes studies related to the gut-lung axis with particular emphasis on the role of the microbiota. Future research directions are also discussed.
Collapse
Affiliation(s)
- Amal S Alswat
- Department of Biotechnology, College of Science, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
| |
Collapse
|
42
|
Acharya B, Tofthagen M, Maciej-Hulme ML, Suissa MR, Karlsson NG. Limited support for a direct connection between prebiotics and intestinal permeability - a systematic review. Glycoconj J 2024; 41:323-342. [PMID: 39287885 PMCID: PMC11522178 DOI: 10.1007/s10719-024-10165-8] [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: 02/07/2024] [Revised: 08/20/2024] [Accepted: 08/29/2024] [Indexed: 09/19/2024]
Abstract
The intestinal barrier is a selective interface between the body´s external and the internal environment. Its layer of epithelial cells is joined together by tight junction proteins. In intestinal permeability (IP), the barrier is compromised, leading to increased translocation of luminal contents such as large molecules, toxins and even microorganisms. Numerous diseases including Inflammatory Bowel Disease (IBD), Coeliac disease (CD), autoimmune disorders, and diabetes are believed to be associated with IP. Dietary interventions, such as prebiotics, may improve the intestinal barrier. Prebiotics are non-digestible food compounds, that promote the growth and activity of beneficial bacteria in the gut. This systematic review assesses the connection between prebiotic usage and IP. PubMed and Trip were used to identify relevant studies conducted between 2010-2023. Only six studies were found, which all varied in the characteristics of the population, study design, and types of prebiotics interventions. Only one study showed a statistically significant effect of prebiotics on IP. Alteration of intestinal barrier function was measured by lactulose/mannitol, chromium-labelled Ethylenediaminetetraacetic acid (51Cr-EDTA), lactulose/rhamnose, and sucralose/erythritol excretion as well as zonulin and glucagon-like peptide 2 levels. Three studies also conducted gut microbiota assessment, and one of them showed statistically significant improvement of the gut microbiome. This study also reported a decrease in zonulin level. The main conclusion from this review is that there is a lack of human studies in this important field. Futhermore, large population studies and using standardized protocols, would be required to properly assess the impact of prebiotic intervention and improvement on IP.
Collapse
Affiliation(s)
- Binayak Acharya
- Department of Life Sciences and Health, Faculty of Health Sciences, Oslo Metropolitan University, St. Olavs Plass, P.O. Box 4, N-0130, Oslo, Norway
| | - Marthe Tofthagen
- Department of Life Sciences and Health, Faculty of Health Sciences, Oslo Metropolitan University, St. Olavs Plass, P.O. Box 4, N-0130, Oslo, Norway
| | - Marissa L Maciej-Hulme
- Department of Life Sciences and Health, Faculty of Health Sciences, Oslo Metropolitan University, St. Olavs Plass, P.O. Box 4, N-0130, Oslo, Norway
| | - Michal Rachel Suissa
- Department of Life Sciences and Health, Faculty of Health Sciences, Oslo Metropolitan University, St. Olavs Plass, P.O. Box 4, N-0130, Oslo, Norway
| | - Niclas G Karlsson
- Department of Life Sciences and Health, Faculty of Health Sciences, Oslo Metropolitan University, St. Olavs Plass, P.O. Box 4, N-0130, Oslo, Norway.
| |
Collapse
|
43
|
Lakshmana Senthil S. A comprehensive review to assess the potential, health benefits and complications of fucoidan for developing as functional ingredient and nutraceutical. Int J Biol Macromol 2024; 277:134226. [PMID: 39074709 DOI: 10.1016/j.ijbiomac.2024.134226] [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: 12/24/2023] [Revised: 07/21/2024] [Accepted: 07/26/2024] [Indexed: 07/31/2024]
Abstract
Polysaccharides from seaweeds or macroalgae are garnering significant interest from pharmaceutical and food industries due to their bioactivities and promising therapeutic effects. Among the diverse agal polysaccharides, fucoidan is a well-documented and stands out as a well-researched sulphated heteropolysaccharide found in brown seaweeds. It primarily consists of l-fucose and sulfate ester groups, along with other monosaccharides like xylose, mannose, uronic acid, rhamnose, arabinose, and galactose. Recent scientific investigations have unveiled the formidable inhibitory prowess of fucoidan against SARS-CoV-2, offering a promising avenue for therapeutic intervention in our current landscape. Moreover, fucoidan has demonstrated remarkable abilities in safeguarding the gastrointestinal tract, regulating angiogenesis, mitigating metabolic syndrome, and fortifying bone health. Despite the abundance of studies underscoring fucoidan's potential as a vital component sourced from nature, its exploitation remains constrained by inherent limitations. Thus, the primary objective of this article is to furnish a comprehensive discourse on the structural attributes, health-enhancing properties, safety parameters, and potential toxicity associated with fucoidan. Furthermore, the discourse extends to elucidating the practical applications and developmental prospects of fucoidan as a cornerstone in the realm of functional foods and nutraceuticals.
Collapse
|
44
|
Hao L, Yan Y, Huang G, Li H. From gut to bone: deciphering the impact of gut microbiota on osteoporosis pathogenesis and management. Front Cell Infect Microbiol 2024; 14:1416739. [PMID: 39386168 PMCID: PMC11461468 DOI: 10.3389/fcimb.2024.1416739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Accepted: 09/06/2024] [Indexed: 10/12/2024] Open
Abstract
Osteoporosis (OP) is characterized by decreased bone mineral density (BMD) and increased fracture risk, poses a significant global health burden. Recent research has shed light on the bidirectional relationship between gut microbiota (GM) and bone health, presenting a novel avenue for understanding OP pathogenesis and developing targeted therapeutic interventions. This review provides a comprehensive overview of the GM-bone axis, exploring the impact of GM on OP development and management. We elucidate established risk factors and pathogenesis of OP, delve into the diversity and functional changes of GM in OP. Furthermore, we examine experimental evidence and clinical observations linking alterations in GM composition or function with variations in BMD and fracture risk. Mechanistic insights into microbial mediators of bone health, such as microbial metabolites and products, are discussed. Therapeutic implications, including GM-targeted interventions and dietary strategies, are also explored. Finally, we identify future research directions and challenges in translating these findings into clinical practice.
Collapse
Affiliation(s)
- Linjie Hao
- Department of Joint Surgery, Honghui Hospital, Xi’an Jiaotong University, Xi’an, China
| | - Yuzhu Yan
- Clinical Laboratory of Honghui Hospital, Xi’an Jiaotong University, Xi’an, China
| | - Guilin Huang
- Department of Joint Surgery, Honghui Hospital, Xi’an Jiaotong University, Xi’an, China
| | - Hui Li
- Department of Joint Surgery, Honghui Hospital, Xi’an Jiaotong University, Xi’an, China
| |
Collapse
|
45
|
Panchal SK, Heimann K, Brown L. Improving Undernutrition with Microalgae. Nutrients 2024; 16:3223. [PMID: 39339823 PMCID: PMC11435262 DOI: 10.3390/nu16183223] [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: 08/03/2024] [Revised: 09/15/2024] [Accepted: 09/18/2024] [Indexed: 09/30/2024] Open
Abstract
Undernutrition is an important global health problem, especially in children and older adults. Both reversal of maternal and child undernutrition and heathy ageing have become United Nations-supported global initiatives, leading to increased attention to nutritional interventions targeting undernutrition. One feasible option is microalgae, the precursor of all terrestrial plants. Most commercially farmed microalgae are photosynthetic single-celled organisms producing organic carbon compounds and oxygen. This review will discuss commercial opportunities to grow microalgae. Microalgae produce lipids (including omega-3 fatty acids), proteins, carbohydrates, pigments and micronutrients and so can provide a suitable and underutilised alternative for addressing undernutrition. The health benefits of nutrients derived from microalgae have been identified, and thus they are suitable candidates for addressing nutritional issues globally. This review will discuss the potential benefits of microalgae-derived nutrients and opportunities for microalgae to be converted into food products. The advantages of microalgae cultivation include that it does not need arable land or pesticides. Additionally, most species of microalgae are still unexplored, presenting options for further development. Further, the usefulness of microalgae for other purposes such as bioremediation and biofuels will increase the knowledge of these microorganisms, allowing the development of more efficient production of these microalgae as nutritional interventions.
Collapse
Affiliation(s)
- Sunil K Panchal
- School of Science, Western Sydney University, Richmond, NSW 2753, Australia
| | - Kirsten Heimann
- College of Medicine and Public Health, Flinders University, Health Science Building, Building 4, Registry Road, Bedford Park, Adelaide, SA 5042, Australia
| | - Lindsay Brown
- School of Pharmacy and Medical Sciences, Griffith University, Gold Coast, QLD 4222, Australia
| |
Collapse
|
46
|
Kerstens R, Ng YZ, Pettersson S, Jayaraman A. Balancing the Oral-Gut-Brain Axis with Diet. Nutrients 2024; 16:3206. [PMID: 39339804 PMCID: PMC11435118 DOI: 10.3390/nu16183206] [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: 08/04/2024] [Revised: 09/14/2024] [Accepted: 09/18/2024] [Indexed: 09/30/2024] Open
Abstract
Background: The oral microbiota is the second largest microbial community in humans. It contributes considerably to microbial diversity and health effects, much like the gut microbiota. Despite physical and chemical barriers separating the oral cavity from the gastrointestinal tract, bidirectional microbial transmission occurs between the two regions, influencing overall host health. Method: This review explores the intricate interplay of the oral-gut-brain axis, highlighting the pivotal role of the oral microbiota in systemic health and ageing, and how it can be influenced by diet. Results: Recent research suggests a relationship between oral diseases, such as periodontitis, and gastrointestinal problems, highlighting the broader significance of the oral-gut axis in systemic diseases, as well as the oral-gut-brain axis in neurological disorders and mental health. Diet influences microbial diversity in the oral cavity and the gut. While certain diets/dietary components improve both gut and oral health, others, such as fermentable carbohydrates, can promote oral pathogens while boosting gut health. Conclusions: Understanding these dynamics is key for promoting a healthy oral-gut-brain axis through dietary interventions that support microbial diversity and mitigate age-related health risks.
Collapse
Affiliation(s)
- Rebecca Kerstens
- ASEAN Microbiome Nutrition Centre, National Neuroscience Institute, 11 Jalan Tan Tock Seng, Singapore 308433, Singapore
| | - Yong Zhi Ng
- ASEAN Microbiome Nutrition Centre, National Neuroscience Institute, 11 Jalan Tan Tock Seng, Singapore 308433, Singapore
- Duke-NUS Medical School, 8 College Rd., Singapore 169857, Singapore
| | - Sven Pettersson
- ASEAN Microbiome Nutrition Centre, National Neuroscience Institute, 11 Jalan Tan Tock Seng, Singapore 308433, Singapore
- Faculty of Medical Sciences, Sunway University, Subang Jaya 47500, Selangor, Malaysia
- Department of Microbiology and Immunology, National University Singapore, Singapore 117545, Singapore
| | - Anusha Jayaraman
- ASEAN Microbiome Nutrition Centre, National Neuroscience Institute, 11 Jalan Tan Tock Seng, Singapore 308433, Singapore
| |
Collapse
|
47
|
Tîrziu AT, Susan M, Susan R, Sonia T, Harich OO, Tudora A, Varga NI, Tiberiu-Liviu D, Avram CR, Boru C, Munteanu M, Horhat FG. From Gut to Eye: Exploring the Role of Microbiome Imbalance in Ocular Diseases. J Clin Med 2024; 13:5611. [PMID: 39337098 PMCID: PMC11432523 DOI: 10.3390/jcm13185611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 09/16/2024] [Accepted: 09/18/2024] [Indexed: 09/30/2024] Open
Abstract
Background: The gut microbiome plays a crucial role in human health, and recent research has highlighted its potential impact on ocular health through the gut-eye axis. Dysbiosis, or an imbalance in the gut microbiota, has been implicated in various ocular diseases. Methods: A comprehensive literature search was conducted using relevant keywords in major electronic databases, prioritizing recent peer-reviewed articles published in English. Results: The gut microbiota influences ocular health through immune modulation, maintenance of the blood-retinal barrier, and production of beneficial metabolites. Dysbiosis can disrupt these mechanisms, contributing to ocular inflammation, tissue damage, and disease progression in conditions such as uveitis, age-related macular degeneration, diabetic retinopathy, dry eye disease, and glaucoma. Therapeutic modulation of the gut microbiome through probiotics, prebiotics, synbiotics, and fecal microbiota transplantation shows promise in preclinical and preliminary human studies. Conclusions: The gut-eye axis represents a dynamic and complex interplay between the gut microbiome and ocular health. Targeting the gut microbiome through innovative therapeutic strategies holds potential for improving the prevention and management of various ocular diseases.
Collapse
Affiliation(s)
- Andreea-Talida Tîrziu
- Department of General Medicine, Doctoral School, "Victor Babes" University of Medicine and Pharmacy, 300041 Timisoara, Romania
- Department of Ophthalmology, "Victor Babes" University of Medicine and Pharmacy, 300041 Timisoara, Romania
| | - Monica Susan
- Centre for Preventive Medicine, Department of Internal Medicine, "Victor Babes" University of Medicine and Pharmacy, Eftimie Murgu Square, No. 2, 300041 Timisoara, Romania
| | - Razvan Susan
- Centre for Preventive Medicine, Department of Family Medicine, "Victor Babes" University of Medicine and Pharmacy, Eftimie Murgu Square, No. 2, 300041 Timisoara, Romania
| | - Tanasescu Sonia
- Department of Pediatrics, "Victor Babes" University of Medicine and Pharmacy, Eftimie Murgu Sq. No. 2, 300041 Timisoara, Romania
| | - Octavia Oana Harich
- Department of Functional Sciences, "Victor Babes" University of Medicine and Pharmacy Timisoara, Eftimie Murgu Sq. No. 2, 300041 Timisoara, Romania
| | - Adelina Tudora
- Multidisciplinary Doctoral School, Vasile Goldis Western University of Arad, Strada Liviu Rebreanu 86, 310419 Arad, Romania
| | - Norberth-Istvan Varga
- Department of General Medicine, Doctoral School, "Victor Babes" University of Medicine and Pharmacy, 300041 Timisoara, Romania
| | - Dragomir Tiberiu-Liviu
- Medical Semiology II Discipline, Internal Medicine Department, "Victor Babes" University of Medicine and Pharmacy, Eftimie Murgu Square 2, 300041 Timisoara, Romania
| | - Cecilia Roberta Avram
- Department of Residential Training and Post-University Courses, "Vasile Goldis" Western University, 310414 Arad, Romania
| | - Casiana Boru
- Department of Medicine, "Vasile Goldis" University of Medicine and Pharmacy, 310414 Arad, Romania
| | - Mihnea Munteanu
- Department of Ophthalmology, "Victor Babes" University of Medicine and Pharmacy, 300041 Timisoara, Romania
| | - Florin George Horhat
- Multidisciplinary Research Center on Antimicrobial Resistance (MULTI-REZ), Microbiology Department, "Victor Babes" University of Medicine and Pharmacy, 300041 Timisoara, Romania
| |
Collapse
|
48
|
Patel BK, Patel KH, Lee CN, Moochhala S. Intestinal Microbiota Interventions to Enhance Athletic Performance-A Review. Int J Mol Sci 2024; 25:10076. [PMID: 39337561 PMCID: PMC11432184 DOI: 10.3390/ijms251810076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 09/17/2024] [Accepted: 09/18/2024] [Indexed: 09/30/2024] Open
Abstract
Recent years have witnessed an uptick in research highlighting the gut microbiota's role as a primary determinant of athletes' health, which has piqued interest in the hypothesis that it correlates with athletes' physical performance. Athletes' physical performances could be impacted by the metabolic activity of the assortment of microbes found in their gut. Intestinal microbiota impacts multiple facets of an athlete's physiology, including immune response, gut membrane integrity, macro- and micronutrient absorption, muscle endurance, and the gut-brain axis. Several physiological variables govern the gut microbiota; hence, an intricately tailored and complex framework must be implemented to comprehend the performance-microbiota interaction. Emerging evidence underscores the intricate relationship between the gut microbiome and physical fitness, revealing that athletes who engage in regular physical activity exhibit a richer diversity of gut microbes, particularly within the Firmicutes phylum, e.g., Ruminococcaceae genera, compared to their sedentary counterparts. In elite sport, it is challenging to implement an unconventional strategy whilst simultaneously aiding an athlete to accomplish feasible, balanced development. This review compiles the research on the effects of gut microbiota modulation on performance in sports and illustrates how different supplementation strategies for gut microbiota have the ability to improve athletic performance by enhancing physical capacities. In addition to promoting athletes' overall health, this study evaluates the existing literature in an effort to shed light on how interventions involving the gut microbiota can dramatically improve performance on the field. The findings should inform both theoretical and practical developments in the fields of sports nutrition and training.
Collapse
Affiliation(s)
- Bharati Kadamb Patel
- Department of Surgery, Yong Loo Lin School of Medicine, Level 8, NUHS Tower Block, Singapore 119278, Singapore; (B.K.P.); (C.N.L.)
| | - Kadamb Haribhai Patel
- Temasek Polytechnic, School of Applied Sciences, 21 Tampines Ave 1, Singapore 529757, Singapore;
| | - Chuen Neng Lee
- Department of Surgery, Yong Loo Lin School of Medicine, Level 8, NUHS Tower Block, Singapore 119278, Singapore; (B.K.P.); (C.N.L.)
| | - Shabbir Moochhala
- Department of Surgery, Yong Loo Lin School of Medicine, Level 8, NUHS Tower Block, Singapore 119278, Singapore; (B.K.P.); (C.N.L.)
- Department of Pharmacology, Yong Loo Lin School of Medicine, Block MD3, 16 Medical Drive, Singapore 117600, Singapore
| |
Collapse
|
49
|
Jangid AK, Noh KM, Kim S, Kim K. Engineered inulin-based hybrid biomaterials for augmented immunomodulatory responses. Carbohydr Polym 2024; 340:122311. [PMID: 38858027 DOI: 10.1016/j.carbpol.2024.122311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 05/20/2024] [Accepted: 05/21/2024] [Indexed: 06/12/2024]
Abstract
Modified biopolymers that are based on prebiotics have been found to significantly contribute to immunomodulatory events. In recent years, there has been a growing use of modified biomaterials and polymer-functionalized nanomaterials in the treatment of various tumors by activating immune cells. However, the effectiveness of immune cells against tumors is hindered by several biological barriers, which highlights the importance of harnessing prebiotic-based biopolymers to enhance host defenses against cancer, thus advancing cancer prevention strategies. Inulin, in particular, plays a crucial role in activating immune cells and promoting the secretion of cytokines. Therefore, this mini-review aims to emphasize the importance of inulin in immunomodulatory responses, the development of inulin-based hybrid biopolymers, and the role of inulin in enhancing immunity and modifying cell surfaces. Furthermore, we discuss the various approaches of chemical modification for inulin and their potential use in cancer treatment, particularly in the field of cancer immunotherapy.
Collapse
Affiliation(s)
- Ashok Kumar Jangid
- Department of Chemical & Biochemical Engineering, Dongguk University, Seoul 04620, Republic of Korea
| | - Kyung Mu Noh
- Department of Chemical & Biochemical Engineering, Dongguk University, Seoul 04620, Republic of Korea
| | - Sungjun Kim
- Department of Chemical & Biochemical Engineering, Dongguk University, Seoul 04620, Republic of Korea
| | - Kyobum Kim
- Department of Chemical & Biochemical Engineering, Dongguk University, Seoul 04620, Republic of Korea.
| |
Collapse
|
50
|
Kodešová T, Mašlejová A, Vlková E, Musilová Š, Horváthová K, Šubrtová Salmonová H. In Vitro Utilization of Prebiotics by Listeria monocytogenes. Microorganisms 2024; 12:1876. [PMID: 39338550 PMCID: PMC11433794 DOI: 10.3390/microorganisms12091876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 09/08/2024] [Accepted: 09/10/2024] [Indexed: 09/30/2024] Open
Abstract
Listeria monocytognes is an emerging pathogen responsible for the serious foodborne disease, listeriosis. The commensal gut microbiota is the first line of defense against pathogen internalization. The gut microbiome can be modified by prebiotic substrates, which are frequently added to food products and dietary supplements. Prebiotics should selectively support the growth of beneficial microbes and thus improve host health. Nevertheless, little is known about their effect on the growth of L. monocytogenes. The aim of this study was to evaluate the growth ability of four L. monocytogenes strains, representing the most common serotypes, on prebiotic oligosaccharides (beta-(1,3)-D-glucan, inulin, fructooligosaccharides, galactooligosaccharides, lactulose, raffinose, stachyose and 2'-fucosyllactose and a mixture of human milk oligosaccharides) as a sole carbon source. The results showed that only beta-(1,3)-D-glucan was metabolized by L. monocytogenes. These cell culture data suggest that beta-(1,3)-D-glucan may not be selectively utilized by healthy commensal bacteria, and its role in intestinal pathogen growth warrants further exploration in vivo.
Collapse
Affiliation(s)
- Tereza Kodešová
- Department of Microbiology, Nutrition and Dietetics, Faculty of Agrobiology, Food and Natural Resources, Czech University of Life Sciences Prague, Kamýcká 129, 165 21 Prague, Czech Republic
| | - Anna Mašlejová
- Department of Microbiology, Nutrition and Dietetics, Faculty of Agrobiology, Food and Natural Resources, Czech University of Life Sciences Prague, Kamýcká 129, 165 21 Prague, Czech Republic
| | - Eva Vlková
- Department of Microbiology, Nutrition and Dietetics, Faculty of Agrobiology, Food and Natural Resources, Czech University of Life Sciences Prague, Kamýcká 129, 165 21 Prague, Czech Republic
| | - Šárka Musilová
- Department of Microbiology, Nutrition and Dietetics, Faculty of Agrobiology, Food and Natural Resources, Czech University of Life Sciences Prague, Kamýcká 129, 165 21 Prague, Czech Republic
| | - Kristýna Horváthová
- Department of Microbiology, Nutrition and Dietetics, Faculty of Agrobiology, Food and Natural Resources, Czech University of Life Sciences Prague, Kamýcká 129, 165 21 Prague, Czech Republic
| | - Hana Šubrtová Salmonová
- Department of Microbiology, Nutrition and Dietetics, Faculty of Agrobiology, Food and Natural Resources, Czech University of Life Sciences Prague, Kamýcká 129, 165 21 Prague, Czech Republic
| |
Collapse
|