1
|
Bhatia A, Sharma D, Mehta J, Kumarasamy V, Begum MY, Siddiqua A, Sekar M, Subramaniyan V, Wong LS, Mat Rani NNI. Probiotics and Synbiotics: Applications, Benefits, and Mechanisms for the Improvement of Human and Ecological Health. J Multidiscip Healthc 2025; 18:1493-1510. [PMID: 40092220 PMCID: PMC11910042 DOI: 10.2147/jmdh.s501056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Accepted: 02/05/2025] [Indexed: 03/19/2025] Open
Abstract
This review explores the multifaceted roles and applications of probiotics, emphasizing their significance in maintaining and enhancing host health through microbial interactions. It includes the concept of holobionts and the symbiotic relationships between hosts and their microbiomes, illustrating how various microbiota can enhance immunity, support growth, and prevent diseases. It delves into the customization of probiotics using molecular and genomic techniques, focusing Enterococcus, Bifidobacterium, and Lactobacillus species. Furthermore, it discusses the symbiotic effects of symbiotics which aids in enhancing the survivability and beneficial effects of probiotics. The role beneficial microbes in gut is emphasized, noting its impact on preventing diseases and maintaining a stable microbial community. The potential therapeutic value of probiotics includes the ability to treat gastrointestinal diseases, as well as to strengthen the immune system and reduce the number of free radicals that are present in the body. Additionally, it explores secondary metabolites produced by bacteria in the gut, such as bacteriocins and exopolysaccharides, and their effect on the health of human, particularly in the gastrointestinal tract. The review concludes by addressing the use of probiotics in traditional medicine and their potential in novel therapeutic applications, including the treatment of endangered wildlife species and various human ailments.
Collapse
Affiliation(s)
- Ankita Bhatia
- Faculty of Applied Sciences and Biotechnology, Shoolini University of Biotechnology and Management Sciences, Solan, Himachal Pradesh, India
| | - Deeksha Sharma
- Faculty of Applied Sciences and Biotechnology, Shoolini University of Biotechnology and Management Sciences, Solan, Himachal Pradesh, India
| | - Jyoti Mehta
- Faculty of Applied Sciences and Biotechnology, Shoolini University of Biotechnology and Management Sciences, Solan, Himachal Pradesh, India
| | - Vinoth Kumarasamy
- Department of Parasitology & Medical Entomology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, Cheras, Kuala Lumpur, Malaysia
| | - M Yasmin Begum
- Department of Pharmaceutics, College of Pharmacy, King Khalid University, Abha, Saudi Arabia
| | - Ayesha Siddiqua
- Department of Clinical Pharmacy, College of Pharmacy, King Khalid University, Abha, Saudi Arabia
| | - Mahendran Sekar
- School of Pharmacy, Monash University Malaysia, Bandar Sunway, Subang Jaya, Selangor, Malaysia
| | - Vetriselvan Subramaniyan
- Department of Medical Sciences, School of Medical and Life Sciences, Sunway University, Bandar Sunway, Selangor, Malaysia
| | - Ling Shing Wong
- Faculty of Health and Life Sciences, INTI International University, Nilai, Malaysia
| | - Nur Najihah Izzati Mat Rani
- Faculty of Pharmacy and Health Sciences, Royal College of Medicine Perak, Universiti Kuala Lumpur, Ipoh, Perak, Malaysia
| |
Collapse
|
2
|
Golshany H, Helmy SA, Morsy NFS, Kamal A, Yu Q, Fan L. The gut microbiome across the lifespan: how diet modulates our microbial ecosystem from infancy to the elderly. Int J Food Sci Nutr 2025; 76:95-121. [PMID: 39701663 DOI: 10.1080/09637486.2024.2437472] [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: 08/11/2024] [Revised: 10/16/2024] [Accepted: 11/28/2024] [Indexed: 12/21/2024]
Abstract
This comprehensive review examines the impact of dietary patterns on gut microbiome composition and diversity from infancy to old age, linking these changes to age-related health outcomes. It investigates how the gut microbiome develops and changes across life stages, focusing on the influence of dietary factors. The review explores how early-life feeding practices, including breastfeeding and formula feeding, shape the infant gut microbiota and have lasting effects. In elderly individuals, alterations in the gut microbiome are associated with increased susceptibility to infections, chronic inflammation, metabolic disorders and cognitive decline. The critical role of diet in modulating the gut microbiome throughout life is emphasised, particularly the potential benefits of probiotics and fortified foods in promoting healthy ageing. By elucidating the mechanisms connecting food systems to gut health, this review provides insights into interventions that could enhance gut microbiome resilience and improve health outcomes across the lifespan.
Collapse
Affiliation(s)
- Hazem Golshany
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, China
- Food Science Department, Faculty of Agriculture, Cairo University, Giza, Egypt
- School of Food Science and Technology, Jiangnan University, Wuxi, China
| | | | | | - Aya Kamal
- Food Science Department, Faculty of Agriculture, Cairo University, Giza, Egypt
| | - Qun Yu
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Liuping Fan
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, China
- Collaborative Innovation Center of Food Safety & Quality Control, Jiangnan University, Wuxi, China
| |
Collapse
|
3
|
Zhang W, Zhang Y, Zhao Y, Li L, Zhang Z, Hettinga K, Yang H, Deng J. A Comprehensive Review on Dietary Polysaccharides as Prebiotics, Synbiotics, and Postbiotics in Infant Formula and Their Influences on Gut Microbiota. Nutrients 2024; 16:4122. [PMID: 39683515 DOI: 10.3390/nu16234122] [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: 11/04/2024] [Revised: 11/25/2024] [Accepted: 11/26/2024] [Indexed: 12/18/2024] Open
Abstract
Human milk contains an abundance of nutrients which benefit the development and growth of infants. However, infant formula has to be used when breastfeeding is not possible. The large differences between human milk and infant formula in prebiotics lead to the suboptimal intestinal health of infant formula-fed infants. This functional deficit of infant formula may be overcome through other dietary polysaccharides that have been characterized. The aim of this review was to summarize the potential applications of dietary polysaccharides as prebiotics, synbiotics, and postbiotics in infant formula to better mimic the functionality of human milk prebiotics for infant gut health. Previous studies have demonstrated the influences of dietary polysaccharides on gut microbiota, SCFA production, and immune system development. Compared to prebiotics, synbiotics and postbiotics showed better application potential in shaping the gut microbiota, the prevention of pathogen infections, and the development of the immune system. Moreover, the safety issues for biotics still require more clinical trials with a large-scale population and long time duration, and the generally accepted regulations are important to regulate related products. Pectin polysaccharides has similar impacts to human milk oligosaccharides on gut microbiota and the repairing of a damaged gut barrier, with similar functions also being observed for inulin and β-glucan. Prebiotics as an encapsulation material combined with probiotics and postbiotics showed better potential applications compared to traditional material in infant formula.
Collapse
Affiliation(s)
- Wenyuan Zhang
- State Key Laboratory of Vegetable Biobreeding, Institute of Vegetables and Flowers, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Yanli Zhang
- State Key Laboratory of Vegetable Biobreeding, Institute of Vegetables and Flowers, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Yaqi Zhao
- State Key Laboratory of Vegetable Biobreeding, Institute of Vegetables and Flowers, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Liang Li
- State Key Laboratory of Vegetable Biobreeding, Institute of Vegetables and Flowers, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Zhanquan Zhang
- State Key Laboratory of Vegetable Biobreeding, Institute of Vegetables and Flowers, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Kasper Hettinga
- Dairy Science and Technology, Food Quality and Design Group, Wageningen University & Research, 6708 WG Wageningen, The Netherlands
| | - Haixia Yang
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Jianjun Deng
- State Key Laboratory of Vegetable Biobreeding, Institute of Vegetables and Flowers, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| |
Collapse
|
4
|
Shafaeizadeh S, Henry CJ, van Helvoort A, Alles M, Abrahamse-Berkeveld M. Tailored recommendations for infant milk formula intake results in more accurate feeding. Eur J Pediatr 2024; 183:4693-4704. [PMID: 39186085 PMCID: PMC11473556 DOI: 10.1007/s00431-024-05726-w] [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: 02/15/2024] [Revised: 08/04/2024] [Accepted: 08/08/2024] [Indexed: 08/27/2024]
Abstract
Currently available guidelines on the daily formula milk requirements of infants are based on the needs of infants with their growth pattern following the 50th percentile of the weight-for-age growth curve. Hence, current recommendations may not thoroughly detail the needs of infants across the broad spectrum of body weight percentiles. This study aimed to provide stratified recommendations for daily formula milk intake of fully formula-fed infants, across different weight-for-age categories from 0 to 4 months. At first, theoretical age- and gender-specific weight ranges were constructed for infants across five pre-defined weight-for-length percentile categories of the WHO growth standard. Thereafter, total daily energy requirements for each category were calculated and converted to daily formula milk needs. Subsequently, these stratified age- and weight-formula milk recommendations were compared to actual daily and relative formula milk of infants in these categories, retrieved from pooled individual infant formula milk intake data derived from 13 clinical intervention trials. A fitted regression model was used to evaluate differences in volume intakes across body weight categories as well as between theoretically derived and actual intake values. Median daily formula milk volume intake (ml/day) of infants differed significantly across the increasing weight-for-age categories at each time point, with significant differences between small and large infants. Interestingly, the relative daily formula milk volume intake (ml/kg/day) was higher for smaller infants compared to larger infants. The mean daily and relative formula milk intakes demonstrated the same pattern based on theoretical calculations as well as for the actual formula milk intake values retrieved from 13 pooled clinical intervention trials. CONCLUSIONS Based on theoretical calculations and actual formula intake data, we conclude that larger infants require a significantly higher daily formula milk intake than smaller infants, and we postulate that infants could benefit from more tailored formula milk intake recommendations. WHAT IS KNOWN • Adequate energy intake during the infancy period is crucial to support optimal growth and organ development, with the potential for long-lasting health effects. • Current available guidelines on the daily formula milk requirements of infants are based on the needs of infants with their growth pattern following the 50th percentile of the weight-for-age growth curve. WHAT IS NEW • Based on using both theoretical calculations and actual formula intake data, larger infants require a significantly higher daily formula milk intake than smaller infants. • Exclusive formula-fed infants could benefit from more tailored formula milk intake recommendations, in early infancy.
Collapse
Affiliation(s)
- Shila Shafaeizadeh
- Danone Nutricia Research, Uppsalalaan 12, 3584 CT, Utrecht, The Netherlands.
| | - Christiani Jeyakumar Henry
- Clinical Nutrition Research Centre (CNRC), Singapore Institute of Food and Biotechnology Innovation (SIFBI), Singapore, Singapore
| | - Ardy van Helvoort
- Danone Nutricia Research, Uppsalalaan 12, 3584 CT, Utrecht, The Netherlands
- Department of Respiratory Medicine, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Martine Alles
- Danone Nutricia Research, Uppsalalaan 12, 3584 CT, Utrecht, The Netherlands
| | | |
Collapse
|
5
|
Cappio Barazzone E, Diard M, Hug I, Larsson L, Slack E. Diagnosing and engineering gut microbiomes. EMBO Mol Med 2024; 16:2660-2677. [PMID: 39468301 PMCID: PMC11554810 DOI: 10.1038/s44321-024-00149-4] [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: 05/22/2024] [Revised: 09/18/2024] [Accepted: 09/19/2024] [Indexed: 10/30/2024] Open
Abstract
The microbes, nutrients and toxins that we are exposed to can have a profound effect on the composition and function of the gut microbiome. Thousands of peer-reviewed publications link microbiome composition and function to health from the moment of birth, right through to centenarians, generating a tantalizing glimpse of what might be possible if we could intervene rationally. Nevertheless, there remain relatively few real-world examples where successful microbiome engineering leads to beneficial health effects. Here we aim to provide a framework for the progress needed to turn gut microbiome engineering from a trial-and-error approach to a rational medical intervention. The workflow starts with truly understanding and accurately diagnosing the problems that we are trying to fix, before moving on to developing technologies that can achieve the desired changes.
Collapse
Affiliation(s)
- Elisa Cappio Barazzone
- Laboratory for Mucosal Immunology, Institute for Food, Nutrition and Health, Department of Health Sciences and Technology, ETH Zurich, Zürich, Switzerland
- Basel Research Centre for Child Health, Basel, Switzerland
| | - Médéric Diard
- Basel Research Centre for Child Health, Basel, Switzerland
- Biozentrum, University of Basel, Basel, Switzerland
| | - Isabelle Hug
- Basel Research Centre for Child Health, Basel, Switzerland
- Biozentrum, University of Basel, Basel, Switzerland
| | - Louise Larsson
- Laboratory for Mucosal Immunology, Institute for Food, Nutrition and Health, Department of Health Sciences and Technology, ETH Zurich, Zürich, Switzerland
- Basel Research Centre for Child Health, Basel, Switzerland
| | - Emma Slack
- Laboratory for Mucosal Immunology, Institute for Food, Nutrition and Health, Department of Health Sciences and Technology, ETH Zurich, Zürich, Switzerland.
- Basel Research Centre for Child Health, Basel, Switzerland.
- Sir William Dunn School of Pathology, University of Oxford, Oxford, UK.
| |
Collapse
|
6
|
Sáez‐Fuertes L, Rio‐Aige K, Massot‐Cladera M, Castell M, Knipping K, Garssen J, Bourdet‐Sicard R, Rodríguez‐Lagunas MJ, Collado MC, Pérez‐Cano FJ. Bifidobacterium breve M-16 V and scGOS/lcFOS Supplementation to Dams Ameliorates Infant Rotavirus Infection in Early Life. Mol Nutr Food Res 2024; 68:e2400377. [PMID: 39468988 PMCID: PMC11605786 DOI: 10.1002/mnfr.202400377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 09/29/2024] [Indexed: 10/30/2024]
Abstract
The immune system of newborns is underdeveloped, leaving them susceptible to infections like rotavirus (RV). Despite vaccines, RV remains a leading cause of child mortality, especially in developing countries. Maternal immunity is transferred during pregnancy and breastfeeding to the offspring providing protection against RV infection. This study aims to explore how the maternal diet can enhance the newborn's ability to fight early infections. Pregnant rats received orally Bifidobacterium breve M-16 V and short chain galacto-oligosaccharides (scGOS)/long chain fructo-oligosaccharides (lcFOS). At day 5 of life pups are infected with RV and at day 8, samples are collected for the infection analysis. Pups whose mothers received the synbiotic have lower RV infection severity. The levels of immunoglobulins (Ig) IgG2c and IgA are raised in pups' plasma and digested milk, respectively. Synbiotic supplementation improves intestinal maturation and increases gene expression of immune-related genes. In conclusion, the administration of this synbiotic to gestating and lactating mothers ameliorates the incidence and severity of the pup's diarrhea caused by the RV infection by improving their immunity.
Collapse
Affiliation(s)
- Laura Sáez‐Fuertes
- Physiology Section, Department of Biochemistry and PhysiologyFaculty of Pharmacy and Food ScienceUniversity of Barcelona (UB)Barcelona08028Spain
- Nutrition and Food Safety Research Institute (INSA‐UB)Santa Coloma de Gramenet08921Spain
| | - Karla Rio‐Aige
- Physiology Section, Department of Biochemistry and PhysiologyFaculty of Pharmacy and Food ScienceUniversity of Barcelona (UB)Barcelona08028Spain
- Nutrition and Food Safety Research Institute (INSA‐UB)Santa Coloma de Gramenet08921Spain
| | - Malén Massot‐Cladera
- Physiology Section, Department of Biochemistry and PhysiologyFaculty of Pharmacy and Food ScienceUniversity of Barcelona (UB)Barcelona08028Spain
- Nutrition and Food Safety Research Institute (INSA‐UB)Santa Coloma de Gramenet08921Spain
| | - Margarida Castell
- Physiology Section, Department of Biochemistry and PhysiologyFaculty of Pharmacy and Food ScienceUniversity of Barcelona (UB)Barcelona08028Spain
- Nutrition and Food Safety Research Institute (INSA‐UB)Santa Coloma de Gramenet08921Spain
- Biomedical Research Centre in Physiopathology of Obesity and Nutrition (CIBEROBN), Institute of Salud Carlos IIIMadrid28029Spain
| | - Karen Knipping
- Danone Research & InnovationUtrechtthe Netherlands
- Division of Pharmacology, Faculty of ScienceUtrecht Institute for Pharmaceutical SciencesUtrechtthe Netherlands
| | - Johan Garssen
- Danone Research & InnovationUtrechtthe Netherlands
- Division of Pharmacology, Faculty of ScienceUtrecht Institute for Pharmaceutical SciencesUtrechtthe Netherlands
| | | | - María José Rodríguez‐Lagunas
- Physiology Section, Department of Biochemistry and PhysiologyFaculty of Pharmacy and Food ScienceUniversity of Barcelona (UB)Barcelona08028Spain
- Nutrition and Food Safety Research Institute (INSA‐UB)Santa Coloma de Gramenet08921Spain
| | - María Carmen Collado
- Institute of Agrochemistry and Food Technology (IATA‐CSIC), National Research CouncilValencia46980Spain
| | - Francisco José Pérez‐Cano
- Physiology Section, Department of Biochemistry and PhysiologyFaculty of Pharmacy and Food ScienceUniversity of Barcelona (UB)Barcelona08028Spain
- Nutrition and Food Safety Research Institute (INSA‐UB)Santa Coloma de Gramenet08921Spain
| |
Collapse
|
7
|
Aziz N, Wal P, Patel A, Prajapati H. A comprehensive review on the pharmacological role of gut microbiome in neurodegenerative disorders: potential therapeutic targets. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:7307-7336. [PMID: 38734839 DOI: 10.1007/s00210-024-03109-4] [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: 10/09/2023] [Accepted: 04/17/2024] [Indexed: 05/13/2024]
Abstract
Neurological disorders, including Alzheimer and Parkinson's, pose significant challenges to public health due to their complex etiologies and limited treatment options. Recent advances in research have highlighted the intricate bidirectional communication between the gut microbiome and the central nervous system (CNS), revealing a potential therapeutic avenue for neurological disorders. Thus, this review aims to summarize the current understanding of the pharmacological role of gut microbiome in neurological disorders. Mounting evidence suggests that the gut microbiome plays a crucial role in modulating CNS function through various mechanisms, including the production of neurotransmitters, neuroactive metabolites, and immune system modulation. Dysbiosis, characterized by alterations in gut microbial composition and function, has been observed in many neurological disorders, indicating a potential causative or contributory role. Pharmacological interventions targeting the gut microbiome have emerged as promising therapeutic strategies for neurological disorders. Probiotics, prebiotics, antibiotics, and microbial metabolite-based interventions have shown beneficial effects in animal models and some human studies. These interventions aim to restore microbial homeostasis, enhance microbial diversity, and promote the production of beneficial metabolites. However, several challenges remain, including the need for standardized protocols, identification of specific microbial signatures associated with different neurological disorders, and understanding the precise mechanisms underlying gut-brain communication. Further research is necessary to unravel the intricate interactions between the gut microbiome and the CNS and to develop targeted pharmacological interventions for neurological disorders.
Collapse
Affiliation(s)
- Namra Aziz
- PSIT-Pranveer Singh Institute of Technology (Pharmacy), NH-19, Kanpur, UP, 209305, India
| | - Pranay Wal
- PSIT-Pranveer Singh Institute of Technology (Pharmacy), NH-19, Kanpur, UP, 209305, India.
| | - Aman Patel
- PSIT-Pranveer Singh Institute of Technology (Pharmacy), NH-19, Kanpur, UP, 209305, India
| | - Harshit Prajapati
- PSIT-Pranveer Singh Institute of Technology (Pharmacy), NH-19, Kanpur, UP, 209305, India
| |
Collapse
|
8
|
Taghizadeh Ghassab F, Shamlou Mahmoudi F, Taheri Tinjani R, Emami Meibodi A, Zali MR, Yadegar A. Probiotics and the microbiota-gut-brain axis in neurodegeneration: Beneficial effects and mechanistic insights. Life Sci 2024; 350:122748. [PMID: 38843992 DOI: 10.1016/j.lfs.2024.122748] [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: 12/05/2023] [Revised: 03/21/2024] [Accepted: 05/23/2024] [Indexed: 06/10/2024]
Abstract
Neurodegenerative diseases (NDs) are a group of heterogeneous disorders with a high socioeconomic burden. Although pharmacotherapy is currently the principal therapeutic approach for the management of NDs, mounting evidence supports the notion that the protracted application of available drugs would abate their dopaminergic outcomes in the long run. The therapeutic application of microbiome-based modalities has received escalating attention in biomedical works. In-depth investigations of the bidirectional communication between the microbiome in the gut and the brain offer a multitude of targets for the treatment of NDs or maximizing the patient's quality of life. Probiotic administration is a well-known microbial-oriented approach to modulate the gut microbiota and potentially influence the process of neurodegeneration. Of note, there is a strong need for further investigation to map out the mechanistic prospects for the gut-brain axis and the clinical efficacy of probiotics. In this review, we discuss the importance of microbiome modulation and hemostasis via probiotics, prebiotics, postbiotics and synbiotics in ameliorating pathological neurodegenerative events. Also, we meticulously describe the underlying mechanism of action of probiotics and their metabolites on the gut-brain axis in different NDs. We suppose that the present work will provide a functional direction for the use of probiotic-based modalities in promoting current practical treatments for the management of neurodegenerative-related diseases.
Collapse
Affiliation(s)
- Fatemeh Taghizadeh Ghassab
- Foodborne and Waterborne Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fatemeh Shamlou Mahmoudi
- Foodborne and Waterborne Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Reyhaneh Taheri Tinjani
- Foodborne and Waterborne Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Armitasadat Emami Meibodi
- Foodborne and Waterborne Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Reza Zali
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Abbas Yadegar
- Foodborne and Waterborne Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
9
|
Singh J, Vanlallawmzuali, Singh A, Biswal S, Zomuansangi R, Lalbiaktluangi C, Singh BP, Singh PK, Vellingiri B, Iyer M, Ram H, Udey B, Yadav MK. Microbiota-brain axis: Exploring the role of gut microbiota in psychiatric disorders - A comprehensive review. Asian J Psychiatr 2024; 97:104068. [PMID: 38776563 DOI: 10.1016/j.ajp.2024.104068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 02/28/2024] [Accepted: 05/01/2024] [Indexed: 05/25/2024]
Abstract
Mental illness is a hidden epidemic in modern science that has gradually spread worldwide. According to estimates from the World Health Organization (WHO), approximately 10% of the world's population suffers from various mental diseases each year. Worldwide, financial and health burdens on society are increasing annually. Therefore, understanding the different factors that can influence mental illness is required to formulate novel and effective treatments and interventions to combat mental illness. Gut microbiota, consisting of diverse microbial communities residing in the gastrointestinal tract, exert profound effects on the central nervous system through the gut-brain axis. The gut-brain axis serves as a conduit for bidirectional communication between the two systems, enabling the gut microbiota to affect emotional and cognitive functions. Dysbiosis, or an imbalance in the gut microbiota, is associated with an increased susceptibility to mental health disorders and psychiatric illnesses. Gut microbiota is one of the most diverse and abundant groups of microbes that have been found to interact with the central nervous system and play important physiological functions in the human gut, thus greatly affecting the development of mental illnesses. The interaction between gut microbiota and mental health-related illnesses is a multifaceted and promising field of study. This review explores the mechanisms by which gut microbiota influences mental health, encompassing the modulation of neurotransmitter production, neuroinflammation, and integrity of the gut barrier. In addition, it emphasizes a thorough understanding of how the gut microbiome affects various psychiatric conditions.
Collapse
Affiliation(s)
- Jawahar Singh
- Department of Psychiatry, All India Institute of Medical Sciences (AIIMS), Bathinda, Punjab, India
| | - Vanlallawmzuali
- Department of Biotechnology, Mizoram Central University, Pachhunga University College Campus, Aizawl, Mizoram, India
| | - Amit Singh
- Department of Microbiology Central University of Punjab, Bathinda 151401, India
| | - Suryanarayan Biswal
- Department of Human Genetics and Molecular Medicine, Central University of Punjab, Bathinda 151401, India
| | - Ruth Zomuansangi
- Department of Microbiology Central University of Punjab, Bathinda 151401, India
| | - C Lalbiaktluangi
- Department of Microbiology Central University of Punjab, Bathinda 151401, India
| | - Bhim Pratap Singh
- Department of Agriculture and Environmental Sciences (AES), National Institute of Food Technology Entrepreneurship and Management (NIFTEM), Sonepat, Haryana, India
| | - Prashant Kumar Singh
- Department of Biotechnology, Pachhunga University College Campus, Mizoram University (A Central University), Aizawl 796001, Mizoram, India
| | - Balachandar Vellingiri
- Stem cell and Regenerative Medicine/Translational Research, Department of Zoology, School of Basic Sciences, Central University of Punjab (CUPB), Bathinda, Punjab 151401, India
| | - Mahalaxmi Iyer
- Department of Microbiology Central University of Punjab, Bathinda 151401, India
| | - Heera Ram
- Department of Zoology, Jai Narain Vyas University, Jodhpur, Rajasthan 342001, India
| | - Bharat Udey
- Department of Psychiatry, All India Institute of Medical Sciences (AIIMS), Bathinda, Punjab, India
| | - Mukesh Kumar Yadav
- Department of Microbiology Central University of Punjab, Bathinda 151401, India.
| |
Collapse
|
10
|
Sáez-Fuertes L, Kapravelou G, Grases-Pintó B, Bernabeu M, Knipping K, Garssen J, Bourdet-Sicard R, Castell M, Rodríguez-Lagunas MJ, Collado MC, Pérez-Cano FJ. Early-Life Supplementation Enhances Gastrointestinal Immunity and Microbiota in Young Rats. Foods 2024; 13:2058. [PMID: 38998564 PMCID: PMC11241808 DOI: 10.3390/foods13132058] [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: 05/21/2024] [Revised: 06/16/2024] [Accepted: 06/25/2024] [Indexed: 07/14/2024] Open
Abstract
Immunonutrition, which focuses on specific nutrients in breast milk and post-weaning diets, plays a crucial role in supporting infants' immune system development. This study explored the impact of maternal supplementation with Bifidobacterium breve M-16V and a combination of short-chain galacto-oligosaccharide (scGOS) and long-chain fructo-oligosaccharide (lcFOS) from pregnancy through lactation, extending into the early childhood of the offspring. The synbiotic supplementation's effects were examined at both mucosal and systemic levels. While the supplementation did not influence their overall growth, water intake, or food consumption, a trophic effect was observed in the small intestine, enhancing its weight, length, width, and microscopic structures. A gene expression analysis indicated a reduction in FcRn and Blimp1 and an increase in Zo1 and Tlr9, suggesting enhanced maturation and barrier function. Intestinal immunoglobulin (Ig) A levels remained unaffected, while cecal IgA levels decreased. The synbiotic supplementation led to an increased abundance of total bacteria and Ig-coated bacteria in the cecum. The abundance of Bifidobacterium increased in both the intestine and cecum. Short-chain fatty acid production decreased in the intestine but increased in the cecum due to the synbiotic supplementation. Systemically, the Ig profiles remained unaffected. In conclusion, maternal synbiotic supplementation during gestation, lactation, and early life is established as a new strategy to improve the maturation and functionality of the gastrointestinal barrier. Additionally, it participates in the microbiota colonization of the gut, leading to a healthier composition.
Collapse
Affiliation(s)
- Laura Sáez-Fuertes
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona (UB), 08028 Barcelona, Spain; (L.S.-F.); (G.K.); (B.G.-P.); (M.C.); (F.J.P.-C.)
- Nutrition and Food Safety Research Institute (INSA-UB), 08921 Santa Coloma de Gramenet, Spain
| | - Garyfallia Kapravelou
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona (UB), 08028 Barcelona, Spain; (L.S.-F.); (G.K.); (B.G.-P.); (M.C.); (F.J.P.-C.)
| | - Blanca Grases-Pintó
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona (UB), 08028 Barcelona, Spain; (L.S.-F.); (G.K.); (B.G.-P.); (M.C.); (F.J.P.-C.)
- Nutrition and Food Safety Research Institute (INSA-UB), 08921 Santa Coloma de Gramenet, Spain
| | - Manuel Bernabeu
- Institute of Agrochemisty and Food Technology-National Research Council (IATA-CSIC), 46980 Valencia, Spain; (M.B.); (M.C.C.)
| | - Karen Knipping
- Danone Research & Innovation, 3584 Utrecht, The Netherlands; (K.K.); (J.G.)
- Division of Pharmacology, Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, 3584 CG Utrecht, The Netherlands
| | - Johan Garssen
- Danone Research & Innovation, 3584 Utrecht, The Netherlands; (K.K.); (J.G.)
- Division of Pharmacology, Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, 3584 CG Utrecht, The Netherlands
| | | | - Margarida Castell
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona (UB), 08028 Barcelona, Spain; (L.S.-F.); (G.K.); (B.G.-P.); (M.C.); (F.J.P.-C.)
- Nutrition and Food Safety Research Institute (INSA-UB), 08921 Santa Coloma de Gramenet, Spain
- Center for Biomedical Research Network for the Physiopathology of Obesity and Nutrition (CIBEROBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - María José Rodríguez-Lagunas
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona (UB), 08028 Barcelona, Spain; (L.S.-F.); (G.K.); (B.G.-P.); (M.C.); (F.J.P.-C.)
- Nutrition and Food Safety Research Institute (INSA-UB), 08921 Santa Coloma de Gramenet, Spain
| | - María Carmen Collado
- Institute of Agrochemisty and Food Technology-National Research Council (IATA-CSIC), 46980 Valencia, Spain; (M.B.); (M.C.C.)
| | - Francisco José Pérez-Cano
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona (UB), 08028 Barcelona, Spain; (L.S.-F.); (G.K.); (B.G.-P.); (M.C.); (F.J.P.-C.)
- Nutrition and Food Safety Research Institute (INSA-UB), 08921 Santa Coloma de Gramenet, Spain
| |
Collapse
|
11
|
Sáez-Fuertes L, Kapravelou G, Grases-Pintó B, Bernabeu M, Knipping K, Garssen J, Bourdet-Sicard R, Castell M, Collado MC, Pérez-Cano FJ, Rodríguez-Lagunas MJ. Maternal Synbiotic Supplementation with B. breve M-16V and scGOS/lcFOS Shape Offspring Immune Development and Gut Microbiota at the End of Suckling. Nutrients 2024; 16:1890. [PMID: 38931246 PMCID: PMC11206815 DOI: 10.3390/nu16121890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 06/06/2024] [Accepted: 06/12/2024] [Indexed: 06/28/2024] Open
Abstract
Immune system development during gestation and suckling is significantly modulated by maternal environmental and dietary factors. Breastfeeding is widely recognized as the optimal source of nutrition for infant growth and immune maturation, and its composition can be modulated by the maternal diet. In the present work, we investigated whether oral supplementation with Bifidobacterium breve M-16V and short-chain galacto-oligosaccharide (scGOS) and long-chain fructo-oligosaccharide (lcFOS) to rat dams during gestation and lactation has an impact on the immune system and microbiota composition of the offspring at day 21 of life. On that day, blood, adipose tissue, small intestine (SI), mesenteric lymph nodes (MLN), salivary gland (SG), cecum, and spleen were collected. Synbiotic supplementation did not affect the overall body or organ growth of the pups. The gene expression of Tlr9, Muc2, IgA, and Blimp1 were upregulated in the SI, and the increase in IgA gene expression was further confirmed at the protein level in the gut wash. Synbiotic supplementation also positively impacted the microbiota composition in both the small and large intestines, resulting in higher proportions of Bifidobacterium genus, among others. In addition, there was an increase in butanoic, isobutanoic, and acetic acid concentrations in the cecum but a reduction in the small intestine. At the systemic level, synbiotic supplementation resulted in higher levels of immunoglobulin IgG2c in plasma, SG, and MLN, but it did not modify the main lymphocyte subsets in the spleen and MLN. Overall, synbiotic maternal supplementation is able to positively influence the immune system development and microbiota of the suckling offspring, particularly at the gastrointestinal level.
Collapse
Affiliation(s)
- Laura Sáez-Fuertes
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona (UB), 08028 Barcelona, Spain; (L.S.-F.); (G.K.); (B.G.-P.); (M.C.); (M.J.R.-L.)
- Nutrition and Food Safety Research Institute (INSA-UB), 08921 Santa Coloma de Gramenet, Spain
| | - Garyfallia Kapravelou
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona (UB), 08028 Barcelona, Spain; (L.S.-F.); (G.K.); (B.G.-P.); (M.C.); (M.J.R.-L.)
| | - Blanca Grases-Pintó
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona (UB), 08028 Barcelona, Spain; (L.S.-F.); (G.K.); (B.G.-P.); (M.C.); (M.J.R.-L.)
- Nutrition and Food Safety Research Institute (INSA-UB), 08921 Santa Coloma de Gramenet, Spain
| | - Manuel Bernabeu
- Institute of Agrochemisty and Food Technology-National Research Council (IATA-CSIC), 46980 Valencia, Spain; (M.B.); (M.C.C.)
| | - Karen Knipping
- Danone Research & Innovation, 3584 Utrecht, The Netherlands; (K.K.); (J.G.)
- Division of Pharmacology, Faculty of Science, Institute for Pharmaceutical Sciences, 3584 Utrecht, The Netherlands
| | - Johan Garssen
- Danone Research & Innovation, 3584 Utrecht, The Netherlands; (K.K.); (J.G.)
- Division of Pharmacology, Faculty of Science, Institute for Pharmaceutical Sciences, 3584 Utrecht, The Netherlands
| | | | - Margarida Castell
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona (UB), 08028 Barcelona, Spain; (L.S.-F.); (G.K.); (B.G.-P.); (M.C.); (M.J.R.-L.)
- Nutrition and Food Safety Research Institute (INSA-UB), 08921 Santa Coloma de Gramenet, Spain
- Center for Biomedical Research Network for the Physiopathology of Obesity and Nutrition (CIBEROBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - María Carmen Collado
- Institute of Agrochemisty and Food Technology-National Research Council (IATA-CSIC), 46980 Valencia, Spain; (M.B.); (M.C.C.)
| | - Francisco José Pérez-Cano
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona (UB), 08028 Barcelona, Spain; (L.S.-F.); (G.K.); (B.G.-P.); (M.C.); (M.J.R.-L.)
- Nutrition and Food Safety Research Institute (INSA-UB), 08921 Santa Coloma de Gramenet, Spain
| | - María José Rodríguez-Lagunas
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona (UB), 08028 Barcelona, Spain; (L.S.-F.); (G.K.); (B.G.-P.); (M.C.); (M.J.R.-L.)
- Nutrition and Food Safety Research Institute (INSA-UB), 08921 Santa Coloma de Gramenet, Spain
| |
Collapse
|
12
|
Mantri A, Köhlmoos A, Schelski DS, Seel W, Stoffel-Wagner B, Krawitz P, Stehle P, Holst JJ, Weber B, Koban L, Plassmann H, Simon MC. Impact of Synbiotic Intake on Liver Metabolism in Metabolically Healthy Participants and Its Potential Preventive Effect on Metabolic-Dysfunction-Associated Fatty Liver Disease (MAFLD): A Randomized, Placebo-Controlled, Double-Blinded Clinical Trial. Nutrients 2024; 16:1300. [PMID: 38732547 PMCID: PMC11085762 DOI: 10.3390/nu16091300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 04/21/2024] [Accepted: 04/24/2024] [Indexed: 05/13/2024] Open
Abstract
Synbiotics modulate the gut microbiome and contribute to the prevention of liver diseases such as metabolic-dysfunction-associated fatty liver disease (MAFLD). This study aimed to evaluate the effect of a randomized, placebo-controlled, double-blinded seven-week intervention trial on the liver metabolism in 117 metabolically healthy male participants. Anthropometric data, blood parameters, and stool samples were analyzed using linear mixed models. After seven weeks of intervention, there was a significant reduction in alanine aminotransferase (ALT) in the synbiotic group compared to the placebo group (-14.92%, CI: -26.60--3.23%, p = 0.013). A stratified analysis according to body fat percentage revealed a significant decrease in ALT (-20.70%, CI: -40.88--0.53%, p = 0.045) in participants with an elevated body fat percentage. Further, a significant change in microbiome composition (1.16, CI: 0.06-2.25, p = 0.039) in this group was found, while the microbial composition remained stable upon intervention in the group with physiological body fat. The 7-week synbiotic intervention reduced ALT levels, especially in participants with an elevated body fat percentage, possibly due to modulation of the gut microbiome. Synbiotic intake may be helpful in delaying the progression of MAFLD and could be used in addition to the recommended lifestyle modification therapy.
Collapse
Affiliation(s)
- Aakash Mantri
- Institute of Nutrition and Food Science, Nutrition and Microbiota, University of Bonn, 53115 Bonn, Germany; (A.M.); (A.K.); (W.S.)
- Institute for Genomic Statistics and Bioinformatics, University Hospital Bonn, 53127 Bonn, Germany;
| | - Anika Köhlmoos
- Institute of Nutrition and Food Science, Nutrition and Microbiota, University of Bonn, 53115 Bonn, Germany; (A.M.); (A.K.); (W.S.)
| | - Daniela Stephanie Schelski
- Center for Economics and Neuroscience, University of Bonn, 53113 Bonn, Germany; (D.S.S.); (B.W.)
- Institute of Experimental Epileptology and Cognition Research, University of Bonn, 53113 Bonn, Germany
| | - Waldemar Seel
- Institute of Nutrition and Food Science, Nutrition and Microbiota, University of Bonn, 53115 Bonn, Germany; (A.M.); (A.K.); (W.S.)
| | - Birgit Stoffel-Wagner
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, 53127 Bonn, Germany;
| | - Peter Krawitz
- Institute for Genomic Statistics and Bioinformatics, University Hospital Bonn, 53127 Bonn, Germany;
| | - Peter Stehle
- Institute of Nutrition and Food Science, Nutritional Physiology, University of Bonn, 53115 Bonn, Germany;
| | - Jens Juul Holst
- Novo Nordisk Foundation Center for Basic Metabolic Research, Department for Biomedical Sciences, University of Copenhagen, DK-2200 Copenhagen, Denmark;
| | - Bernd Weber
- Center for Economics and Neuroscience, University of Bonn, 53113 Bonn, Germany; (D.S.S.); (B.W.)
- Institute of Experimental Epileptology and Cognition Research, University of Bonn, 53113 Bonn, Germany
| | - Leonie Koban
- Institut Européen d’Administration des Affaires (INSEAD), 77300 Fontainebleau, France; (L.K.); (H.P.)
- Control-Interoception-Attention Team, Paris Brain Institute (ICM), 75013 Paris, France
- Lyon Neuroscience Research Center (CRNL), Centre National de la Recherche Scientifique (CNRS), Institut National de la Santé et de la Recherche Médicale (INSERM), Université Claude Bernard Lyon 1, 69500 Bron, France
| | - Hilke Plassmann
- Institut Européen d’Administration des Affaires (INSEAD), 77300 Fontainebleau, France; (L.K.); (H.P.)
- Control-Interoception-Attention Team, Paris Brain Institute (ICM), 75013 Paris, France
| | - Marie-Christine Simon
- Institute of Nutrition and Food Science, Nutrition and Microbiota, University of Bonn, 53115 Bonn, Germany; (A.M.); (A.K.); (W.S.)
| |
Collapse
|
13
|
Revankar NA, Negi PS. Biotics: An emerging food supplement for health improvement in the era of immune modulation. Nutr Clin Pract 2024; 39:311-329. [PMID: 37466413 DOI: 10.1002/ncp.11036] [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: 12/08/2022] [Revised: 05/27/2023] [Accepted: 06/06/2023] [Indexed: 07/20/2023] Open
Abstract
The involvement of the commensal microbiota in immune function is a multifold process. Biotics, such as probiotics, prebiotics, synbiotics, and paraprobiotics, have been subjected to animal and human trials demonstrating the association between gut microbes and immunity biomarkers leading to improvement in overall health. In recent years, studies on human microbiome interaction have established the multifarious role of biotics in maintaining overall health. The consumption of biotics has been extensively reported to help in maintaining microbial diversity, enhancing gut-associated mucosal immune homeostasis, and providing protection against a wide range of lifestyle disorders. However, the establishment of biotics as an alternative therapy for a range of health conditions is yet to be ascertained. Despite the fact that scientific literature has demonstrated the correlation between biotics and immune modulation, most in vivo and in vitro reports are inconclusive on the dosage required. This review provides valuable insights into the immunomodulatory effects of biotics consumption based on evidence obtained from animal models and clinical trials. Furthermore, we highlight the optimal dosages of biotics that have been reported to deliver maximum health benefits. By identifying critical research gaps, we have suggested a roadmap for future investigations to advance our understanding of the intricate crosstalk between biotics and immune homeostasis.
Collapse
Affiliation(s)
- Neelam A Revankar
- Department of Fruit and Vegetables Technology, CSIR-Central Food Technological Research Institute, Mysuru, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Pradeep S Negi
- Department of Fruit and Vegetables Technology, CSIR-Central Food Technological Research Institute, Mysuru, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| |
Collapse
|
14
|
Wang Y, Rui B, Ze X, Liu Y, Yu D, Liu Y, Li Z, Xi Y, Ning X, Lei Z, Yuan J, Li L, Zhang X, Li W, Deng Y, Yan J, Li M. Sialic acid-based probiotic intervention in lactating mothers improves the neonatal gut microbiota and immune responses by regulating sialylated milk oligosaccharide synthesis via the gut-breast axis. Gut Microbes 2024; 16:2334967. [PMID: 38630006 PMCID: PMC11028031 DOI: 10.1080/19490976.2024.2334967] [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: 07/05/2023] [Accepted: 03/21/2024] [Indexed: 04/19/2024] Open
Abstract
Human milk oligosaccharides (HMOs) are vital milk carbohydrates that help promote the microbiota-dependent growth and immunity of infants. Sialic acid (SA) is a crucial component of sialylated milk oligosaccharides (S-MOs); however, the effects of SA supplementation in lactating mothers on S-MO biosynthesis and their breastfed infants are unknown. Probiotic intervention during pregnancy or lactation demonstrates promise for modulating the milk glycobiome. Here, we evaluated whether SA and a probiotic (Pro) mixture could increase S-MO synthesis in lactating mothers and promote the microbiota development of their breastfed neonates. The results showed that SA+Pro intervention modulated the gut microbiota and 6'-SL contents in milk of maternal rats more than the SA intervention, which promoted Lactobacillus reuteri colonization in neonates and immune development. Deficient 6'-SL in the maternal rat milk of St6gal1 knockouts (St6gal1-/-) disturbed intestinal microbial structures in their offspring, thereby impeding immune tolerance development. SA+Pro intervention in lactating St6gal1± rats compromised the allergic responses of neonates by promoting 6'-SL synthesis and the neonatal gut microbiota. Our findings from human mammary epithelial cells (MCF-10A) indicated that the GPR41-PI3K-Akt-PPAR pathway helped regulate 6'-SL synthesis in mammary glands after SA+Pro intervention through the gut - breast axis. We further validated our findings using a human-cohort study, confirming that providing SA+Pro to lactating Chinese mothers increased S-MO contents in their breast milk and promoted gut Bifidobacterium spp. and Lactobacillus spp. colonization in infants, which may help enhance immune responses. Collectively, our findings may help alter the routine supplementation practices of lactating mothers to modulate milk HMOs and promote the development of early-life gut microbiota and immunity.
Collapse
Affiliation(s)
- Yushuang Wang
- Department of Microecology, College of Basic Medical Science, Dalian Medical University, Dalian, China
- Department of Clinical Laboratory, Central Hospital of Dalian University of Technology, Dalian, China
| | - Binqi Rui
- Department of Microecology, College of Basic Medical Science, Dalian Medical University, Dalian, China
| | - Xiaolei Ze
- Microbiome Research and Application Center, BYHEALTH Institute of Nutrition & Health, Guangzhou, China
| | - Yujia Liu
- Department of Microecology, College of Basic Medical Science, Dalian Medical University, Dalian, China
| | - Da Yu
- The Third Ward of Obstetrics and Gynecology at Chunliu District, Dalian Women and Children Medical Center (Group), Dalian, China
| | - Yinhui Liu
- Department of Microecology, College of Basic Medical Science, Dalian Medical University, Dalian, China
| | - Zhi Li
- Department of Clinical Laboratory, Central Hospital of Dalian University of Technology, Dalian, China
| | - Yu Xi
- Microbiome Research and Application Center, BYHEALTH Institute of Nutrition & Health, Guangzhou, China
| | - Xixi Ning
- Department of Microecology, College of Basic Medical Science, Dalian Medical University, Dalian, China
| | - Zengjie Lei
- Department of Microecology, College of Basic Medical Science, Dalian Medical University, Dalian, China
| | - Jieli Yuan
- Department of Microecology, College of Basic Medical Science, Dalian Medical University, Dalian, China
| | - Liang Li
- Microbiome Research and Application Center, BYHEALTH Institute of Nutrition & Health, Guangzhou, China
| | - Xuguang Zhang
- Microbiome Research and Application Center, BYHEALTH Institute of Nutrition & Health, Guangzhou, China
| | - Wenzhe Li
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou University Medical College, Shantou, China
| | - Yanjie Deng
- The Third Ward of Obstetrics and Gynecology at Chunliu District, Dalian Women and Children Medical Center (Group), Dalian, China
| | - Jingyu Yan
- Dalian Institute of Chemical Physics, Chinese Academy of Sciences Key Laboratory of Separation Science for Analytical Chemistry, Dalian, China
| | - Ming Li
- Department of Microecology, College of Basic Medical Science, Dalian Medical University, Dalian, China
| |
Collapse
|
15
|
Roson-Calero N, Ballesté-Delpierre C, Fernández J, Vila J. Insights on Current Strategies to Decolonize the Gut from Multidrug-Resistant Bacteria: Pros and Cons. Antibiotics (Basel) 2023; 12:1074. [PMID: 37370393 DOI: 10.3390/antibiotics12061074] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 06/15/2023] [Accepted: 06/17/2023] [Indexed: 06/29/2023] Open
Abstract
In the last decades, we have witnessed a steady increase in infections caused by multidrug-resistant (MDR) bacteria. These infections are associated with higher morbidity and mortality. Several interventions should be taken to reduce the emergence and spread of MDR bacteria. The eradication of resistant pathogens colonizing specific human body sites that would likely cause further infection in other sites is one of the most conventional strategies. The objective of this narrative mini-review is to compile and discuss different strategies for the eradication of MDR bacteria from gut microbiota. Here, we analyse the prevalence of MDR bacteria in the community and the hospital and the clinical impact of gut microbiota colonisation with MDR bacteria. Then, several strategies to eliminate MDR bacteria from gut microbiota are described and include: (i) selective decontamination of the digestive tract (SDD) using a cocktail of antibiotics; (ii) the use of pre and probiotics; (iii) fecal microbiota transplantation; (iv) the use of specific phages; (v) engineered CRISPR-Cas Systems. This review intends to provide a state-of-the-art of the most relevant strategies to eradicate MDR bacteria from gut microbiota currently being investigated.
Collapse
Affiliation(s)
- Natalia Roson-Calero
- Barcelona Institute for Global Health (ISGlobal), 08036 Barcelona, Spain
- Department of Basic Clinical Practice, School of Medicine, University of Barcelona, 08036 Barcelona, Spain
| | - Clara Ballesté-Delpierre
- Barcelona Institute for Global Health (ISGlobal), 08036 Barcelona, Spain
- CIBER de Enfermedades Infecciosas (CIBERINFEC), Instituto Salud Carlos III, 28029 Madrid, Spain
| | - Javier Fernández
- Liver ICU, Liver Unit, Hospital Clinic, University of Barcelona, IDIBAPS and CIBERehd, 08036 Barcelona, Spain
- European Foundation for the Study of Chronic Liver Failure (EF-Clif), 08021 Barcelona, Spain
| | - Jordi Vila
- Barcelona Institute for Global Health (ISGlobal), 08036 Barcelona, Spain
- Department of Basic Clinical Practice, School of Medicine, University of Barcelona, 08036 Barcelona, Spain
- CIBER de Enfermedades Infecciosas (CIBERINFEC), Instituto Salud Carlos III, 28029 Madrid, Spain
- Department of Clinical Microbiology, Biomedical Diagnostic Center, Hospital Clinic, 08036 Barcelona, Spain
| |
Collapse
|
16
|
Yazdandoust E, Hajifathali A, Roshandel E, Zarif MN, Pourfathollah AA, Parkhideh S, Mehdizadeh M, Amini-Kafiabad S. Gut microbiota intervention by pre and probiotics can induce regulatory T cells and reduce the risk of severe acute GVHD following allogeneic hematopoietic stem cell transplantation. Transpl Immunol 2023; 78:101836. [PMID: 37037266 DOI: 10.1016/j.trim.2023.101836] [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: 01/02/2023] [Revised: 04/02/2023] [Accepted: 04/05/2023] [Indexed: 04/12/2023]
Abstract
BACKGROUND Acute graft-versus-host disease (aGVHD) is one of the leading causes of limitation and mortality after allogeneic hematopoietic stem cell transplantation (allo-HSCT). Numerous studies have shown that changes in the gut microbiome diversity increased post-transplant problems, including the occurrence of aGVHD. Probiotics and prebiotics can reconstitute the gut microbiota and thus increase bacterial metabolites such as short-chain fatty acids (SCFAs) that have immunomodulatory effects preventing aGVHD in recipients of allo-HSCTs. METHODS/STUDY DESIGN We conducted a pilot randomized clinical trial to investigate whether oral synbiotics are associated with the prevention or reduction in occurrence/severity and mitigate complications of aGVHD following allo-HSCT. A commercially available synbiotic mixture containing high levels of 7 safe bacterial strains plus fructo-oligosaccharides as a prebiotic was administered to allo-HSCT recipients. Out of 40 allo-HSCT patients, 20 received daily a synbiotic 21 days prior to transplantation (days -21 to day 0). In contrast, in the control group 20 recipients of allo-HSCT did not receive a symbiotic therapy. RESULTS Within first 100 days of observation, the incidence of severe (grade III/IV) aGVHD in the a synbiotic-therapy group was 0% (0 out of 20 patients), whereas it was 25% (5 out of 20 patients) in the control group (P = 0.047). The median percentage of CD4 + CD25 + Foxp3+ regulatory T cells (Tregs) among CD4+ lymphocytes on day 28 after HSCT in the synbiotic group was higher (2.54%) than in control group (1.73%; P = 0.01). There was no difference in Treg cells on day 7 after HSCT between two groups. However, the median percentage and the absolute count of Tregs in patients who experience aGVHD was significantly lower on days 7 and 28 after HSCT (both P < 0.05). The overall 12-month survival (OS) rate was higher (90%) in the symbiotic-treated patients than in the control group (75%), but the difference was not statistically significant (P = 0.234). CONCLUSION Our preliminary findings suggest that synbiotic intake before and during the conditioning regimen of allo-HSCT patients may lead to a reduction in the incidence and severity of aGVHD through the induction of CD4 + CD25 + Foxp3+ regulatory T cells, thus contributing to the improvement of transplant outcomes. Much larger studies are needed to confirm our observations.
Collapse
Affiliation(s)
- Ehsan Yazdandoust
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran
| | - Abbas Hajifathali
- Hematopoietic Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Elham Roshandel
- Hematopoietic Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mahin Nikougoftar Zarif
- Center for Hematology and Regenerative Medicine, Karolinska Institutet, Department of Medicine, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Ali Akbar Pourfathollah
- Department of Immunology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Sayeh Parkhideh
- Hematopoietic Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mahshid Mehdizadeh
- Hematopoietic Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sedigheh Amini-Kafiabad
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran.
| |
Collapse
|
17
|
Halemani K, Shetty AP, Thimmappa L, Issac A, Dhiraaj S, Radha K, Mishra P, Mathias EG. Impact of probiotic on anxiety and depression symptoms in pregnant and lactating women and microbiota of infants: A systematic review and meta-analysis. J Glob Health 2023; 13:04038. [PMID: 37218177 PMCID: PMC10173681 DOI: 10.7189/jogh.13.04038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/24/2023] Open
Abstract
Background Probiotics are non-invasive therapies composed of live bacteria and yeast. Administration of prebiotics improved the health status of pregnant and lactating women, as well as newborns. This review aimed to appraise the evidence concerning the effectiveness of probiotics on the mental health of pregnant women, lactating mother and the microbiota of the newborn. Methods This systematic review and meta-analysis ascertained quantitative studies published in Medline (PubMed), Clinical Key, EMBASE, Cumulative Index to Nursing and Allied Health Literature (CINAHL), the Cochrane Library, and Google scholar. Two authors independently screened and extracted the data from the primary studies that analysed the efficacy of probiotics on the mental health of pregnant and lactating women and the microbiota of the newborn. We adopted Cochrane Collaboration guidelines and reported using the Preferred Reporting Items for Systematic review and Meta-Analysis (PRISMA) statement. The qualities of included trials were assessed by Cochrane collaboration's risk of bias tool (ROB-2). Results Sixteen trials comprised 946 pregnant women, 524 were lactating mothers, and 1678 were infants. The sample size of primary studies ranged from 36 to 433. Probiotics were administered as interventions, using either a single strain of Bifidobacterium or Lactobacillus or a double-strain combination of Lactobacillus and Bifidobacterium. Probiotics supplementation reduced anxiety in pregnant (n = 676, standardised mean difference (SMD) = 0.01; 95% confidence interval (CI) = -0.28,0.30, P = 0.04, I2 = 70) and lactating women (n = 514, SMD = -0.17; 95% CI = -1.62,1.27, P = 0.98, I2 = 0). Similarly, probiotics decreased depression in pregnant (n = 298, SMD = 0.05; 95% CI = -0.24,0.35, P = 0.20, I2 = 40) and lactating women (n = 518, SMD = -0.10; 95% CI = -1.29,-1.05, P = 0.11, I2 = 60%). Similarly, probiotics supplementation improved the gut microbiota and reduced the duration of crying, abdominal distension, abdominal colic and diarrhoea. Conclusion Non-invasive probiotic therapies are more useful to pregnant and lactating women and newborns. Registration The review protocol was registered with PROSPERO (CRD42022372126).
Collapse
Affiliation(s)
- Kurvatteppa Halemani
- Sanjay Gandhi Post Graduate Institute of Medical Sciences, Lucknow, Uttar Pradesh, India
| | - Asha P Shetty
- All India Institute of Medical Sciences, Bhubaneswar, Odisha, India
| | - Latha Thimmappa
- All India Institute of Medical Sciences, Kalyani, West- Bengal, India
| | - Alwin Issac
- All India Institute of Medical Sciences, Bhubaneswar, Odisha, India
| | - Sanjay Dhiraaj
- Sanjay Gandhi Post Graduate Institute of Medical Sciences, Lucknow, Uttar Pradesh, India
| | - K Radha
- Sanjay Gandhi Post Graduate Institute of Medical Sciences, Lucknow, Uttar Pradesh, India
| | - Prabhaker Mishra
- Sanjay Gandhi Post Graduate Institute of Medical Sciences, Lucknow, Uttar Pradesh, India
| | | |
Collapse
|
18
|
Early life gut microbiota profiles linked to synbiotic formula effects: a randomized clinical trial in European infants. Am J Clin Nutr 2023; 117:326-339. [PMID: 36811568 DOI: 10.1016/j.ajcnut.2022.11.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 11/05/2022] [Accepted: 11/17/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Microbial colonization of the gastrointestinal tract after birth is an essential event that influences infant health with life-long consequences. Therefore, it is important to investigate strategies to positively modulate colonization in early life. OBJECTIVES This randomized, controlled intervention study included 540 infants to investigate the effects of a synbiotic intervention formula (IF) containing Limosilactobacillus fermentum CECT5716 and galacto-oligosaccharides on the fecal microbiome. METHODS The fecal microbiota from infants was analyzed by 16S rRNA amplicon sequencing at 4, 12, and 24 months of age. Metabolites (e.g., short-chain fatty acids) and other milieu parameters (e.g., pH, humidity, and IgA) were also measured in stool samples. RESULTS Microbiota profiles changed with age, with major differences in diversity and composition. Significant effects of the synbiotic IF compared with control formula (CF) were visible at month 4, including higher occurrence of Bifidobacterium spp. and Lactobacillaceae and lower occurrence of Blautia spp., as well as Ruminoccocus gnavus and relatives. This was accompanied by lower fecal pH and concentrations of butyrate. After de novo clustering at 4 months of age, overall phylogenetic profiles of the infants receiving IF were closer to reference profiles of those fed with human milk than infants fed CF. The changes owing to IF were associated with fecal microbiota states characterized by lower occurrence of Bacteroides compared with higher levels of Firmicutes (valid name Bacillota), Proteobacteria (valid name Pseudomonadota), and Bifidobacterium at 4 months of age. These microbiota states were linked to higher prevalence of infants born by Cesarean section. CONCLUSIONS The synbiotic intervention influenced fecal microbiota and milieu parameters at an early age depending on the overall microbiota profiles of the infants, sharing a few similarities with breastfed infants. This trial was registered at clinicaltrials.gov as NCT02221687.
Collapse
|
19
|
Multifaceted role of synbiotics as nutraceuticals, therapeutics and carrier for drug delivery. Chem Biol Interact 2022; 368:110223. [DOI: 10.1016/j.cbi.2022.110223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 09/29/2022] [Accepted: 10/12/2022] [Indexed: 11/15/2022]
|
20
|
Nguyen TT, Nguyen PT, Pham MN, Razafindralambo H, Hoang QK, Nguyen HT. Synbiotics: a New Route of Self-production and Applications to Human and Animal Health. Probiotics Antimicrob Proteins 2022; 14:980-993. [PMID: 35650337 DOI: 10.1007/s12602-022-09960-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/25/2022] [Indexed: 01/17/2023]
Abstract
Synbiotics are preparations in which prebiotics are added to probiotics to achieve superior performance and benefits on the host. A new route of their formation is to induce the prebiotic biosynthesis within the probiotic for synbiotic self-production or autologous synbiotics. The aim of this review paper is first to overview the basic concept and (updated) definitions of synergistic synbiotics, and then to focus particularly on the prebiotic properties of probiotic wall components while describing the environmental factors/stresses that stimulate autologous synbiotics, that is, the biosynthesis of prebiotic-forming microcapsule by probiotic bacteria, and finally to present some of their applications to human and animal health.
Collapse
Affiliation(s)
- Thi-Tho Nguyen
- Hutech Institute of Applied Science, HUTECH University, Ho Chi Minh City, Vietnam
| | - Phu-Tho Nguyen
- An Giang University, An Giang, Vietnam
- Vietnam National University, Ho Chi Minh City, Vietnam
| | - Minh-Nhut Pham
- Hutech Institute of Applied Science, HUTECH University, Ho Chi Minh City, Vietnam
| | | | - Quoc-Khanh Hoang
- Institute of Tropical Biology, Vietnam Academy of Science and Technology, Ho Chi Minh City, Vietnam
| | - Huu-Thanh Nguyen
- An Giang University, An Giang, Vietnam.
- Vietnam National University, Ho Chi Minh City, Vietnam.
| |
Collapse
|
21
|
Khangwal I, Shukla P. A Comparative Analysis for the Production of Xylooligosaccharides via Enzymatic Hydrolysis from Sugarcane Bagasse and Coconut Coir. Indian J Microbiol 2022; 62:317-321. [PMID: 35462709 PMCID: PMC8980137 DOI: 10.1007/s12088-022-01010-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2021] [Accepted: 02/04/2022] [Indexed: 11/05/2022] Open
Abstract
Xylooligosaccharides are known prebiotics that various foods and feed industries can utilize. In the present study, the xylan was extracted from sugarcane bagasse and coconut coir by NaOH treatment and it was further structurally characterized by FTIR. Furthermore, Thermomyces lanuginosus VAPS-24 was used to produce endo-xylanase, which induced the production of XOS from both of these substrates. The maximum production of XOS in 8 h using 20U enzyme concentration and 2% substrate concentration was found as 10.10 mg/ml, 5.43 mg/ml for sugarcane bagasse and coconut coir, respectively. Further, the produced XOS was structurally characterized by the NMR, HPLC and FTIR analysis. This study gives a clue that the sugarcane bagasse can be used as the potent producer for the xylooligosaccharides as compared to the coconut coir and can be utilized at the industrial level. Supplementary Information The online version contains supplementary material available at 10.1007/s12088-022-01010-3.
Collapse
Affiliation(s)
- Ishu Khangwal
- Enzyme Technology and Protein Bioinformatics Laboratory, Department of Microbiology, Maharshi Dayanand University, Rohtak, Haryana 124001 India
| | - Pratyoosh Shukla
- Enzyme Technology and Protein Bioinformatics Laboratory, Department of Microbiology, Maharshi Dayanand University, Rohtak, Haryana 124001 India
- Present Address: School of Biotechnology, Institute of Science, Banaras Hindu University, Varanasi, 221005 India
| |
Collapse
|
22
|
Linehan K, Dempsey EM, Ryan CA, Ross RP, Stanton C. First encounters of the microbial kind: perinatal factors direct infant gut microbiome establishment. MICROBIOME RESEARCH REPORTS 2022; 1:10. [PMID: 38045649 PMCID: PMC10688792 DOI: 10.20517/mrr.2021.09] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 12/28/2021] [Accepted: 01/11/2022] [Indexed: 12/05/2023]
Abstract
The human gut microbiome harbors a diverse range of microbes that play a fundamental role in the health and well-being of their host. The early-life microbiome has a major influence on human development and long-term health. Perinatal factors such as maternal nutrition, antibiotic use, gestational age and mode of delivery influence the initial colonization, development, and function of the neonatal gut microbiome. The perturbed early-life gut microbiome predisposes infants to diseases in early and later life. Understanding how perinatal factors guide and shape the composition of the early-life microbiome is essential to improving infant health. The following review provides a synopsis of perinatal factors with the most decisive influences on initial microbial colonization of the infant gut.
Collapse
Affiliation(s)
- Kevin Linehan
- Teagasc Food Research Centre, Moorepark, Fermoy, Co. Cork P61 C996, Ireland
- APC Microbiome Ireland, Biosciences Institute, University College Cork, Lee Maltings, Cork, Cork T12 YT20, Ireland
- School of Microbiology, University College Cork, Cork T12 YN60, Ireland
| | - Eugene M. Dempsey
- APC Microbiome Ireland, Biosciences Institute, University College Cork, Lee Maltings, Cork, Cork T12 YT20, Ireland
- Department of Paediatrics & Child Health and INFANT Centre, University College Cork, Cork T12 YN60, Ireland
| | - C. Anthony Ryan
- APC Microbiome Ireland, Biosciences Institute, University College Cork, Lee Maltings, Cork, Cork T12 YT20, Ireland
- Department of Paediatrics & Child Health and INFANT Centre, University College Cork, Cork T12 YN60, Ireland
| | - R. Paul Ross
- APC Microbiome Ireland, Biosciences Institute, University College Cork, Lee Maltings, Cork, Cork T12 YT20, Ireland
- School of Microbiology, University College Cork, Cork T12 YN60, Ireland
| | - Catherine Stanton
- Teagasc Food Research Centre, Moorepark, Fermoy, Co. Cork P61 C996, Ireland
- APC Microbiome Ireland, Biosciences Institute, University College Cork, Lee Maltings, Cork, Cork T12 YT20, Ireland
| |
Collapse
|
23
|
The Impact of Probiotics, Prebiotics, and Synbiotics during Pregnancy or Lactation on the Intestinal Microbiota of Children Born by Cesarean Section: A Systematic Review. Nutrients 2022; 14:nu14020341. [PMID: 35057522 PMCID: PMC8778982 DOI: 10.3390/nu14020341] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 01/07/2022] [Accepted: 01/10/2022] [Indexed: 01/27/2023] Open
Abstract
The gut microbiota is a key factor in the correct development of the gastrointestinal immune system. Studies have found differences between the gut microbiota of newborns delivered by cesarean section compared to those vaginally delivered. Our objective was to evaluate the effect of ingestion of probiotics, prebiotics, or synbiotics during pregnancy and/or lactation on the development of the gut microbiota of the C-section newborns. We selected experimental studies in online databases from their inception to October 2021. Of the 83 records screened, 12 met the inclusion criteria. The probiotics used belonged to the genera Lactobacillus, Bifidobacterium, Propionibacterium, and Streptococcus, or a combination of those, with dosages varying between 2 × 106 and 9 × 1011 CFU per day, and were consumed during pregnancy and/or lactation. Probiotic strains were combined with galacto-oligosaccharides, fructo-oligosaccharides, or bovine milk-derived oligosaccharides in the synbiotic formulas. Probiotic, prebiotic, and synbiotic interventions led to beneficial gut microbiota in cesarean-delivered newborns, closer to that in vaginally delivered newborns, especially regarding Bifidobacterium colonization. This effect was more evident in breastfed infants. The studies indicate that this beneficial effect is achieved when the interventions begin soon after birth, especially the restoration of bifidobacterial population. Changes in the infant microbial ecosystem due to the interventions seem to continue after the end of the intervention in most of the studies. More interventional studies are needed to elucidate the optimal synbiotic combinations and the most effective strains and doses for achieving the optimal gut microbiota colonization of C-section newborns.
Collapse
|
24
|
Fabiano V, Indrio F, Verduci E, Calcaterra V, Pop TL, Mari A, Zuccotti GV, Cullu Cokugras F, Pettoello-Mantovani M, Goulet O. Term Infant Formulas Influencing Gut Microbiota: An Overview. Nutrients 2021; 13:4200. [PMID: 34959752 PMCID: PMC8708119 DOI: 10.3390/nu13124200] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 11/19/2021] [Accepted: 11/20/2021] [Indexed: 02/05/2023] Open
Abstract
Intestinal colonization of the neonate is highly dependent on the term of pregnancy, the mode of delivery, the type of feeding [breast feeding or formula feeding]. Postnatal immune maturation is dependent on the intestinal microbiome implementation and composition and type of feeding is a key issue in the human gut development, the diversity of microbiome, and the intestinal function. It is well established that exclusive breastfeeding for 6 months or more has several benefits with respect to formula feeding. The composition of the new generation of infant formulas aims in mimicking HM by reproducing its beneficial effects on intestinal microbiome and on the gut associated immune system (GAIS). Several approaches have been developed currently for designing new infant formulas by the addition of bioactive ingredients such as human milk oligosaccharides (HMOs), probiotics, prebiotics [fructo-oligosaccharides (FOSs) and galacto-oligosaccharides (GOSs)], or by obtaining the so-called post-biotics also known as milk fermentation products. The aim of this article is to guide the practitioner in the understanding of these different types of Microbiota Influencing Formulas by listing and summarizing the main concepts and characteristics of these different models of enriched IFs with bioactive ingredients.
Collapse
Affiliation(s)
- Valentina Fabiano
- Department of Pediatrics, Vittore Buzzi Children’s Hospital, Università degli Studi di Milano, 20154 Milan, Italy; (V.F.); (E.V.); (V.C.); (A.M.); (G.V.Z.)
| | - Flavia Indrio
- Department of Medical and Surgical Sciences, University of Foggia, 71100 Foggia, Italy
- European Paediatric Association/Union of National European Paediatric Societies and Associations, 10115 Berlin, Germany; (T.L.P.); (F.C.C.); (M.P.-M.)
| | - Elvira Verduci
- Department of Pediatrics, Vittore Buzzi Children’s Hospital, Università degli Studi di Milano, 20154 Milan, Italy; (V.F.); (E.V.); (V.C.); (A.M.); (G.V.Z.)
| | - Valeria Calcaterra
- Department of Pediatrics, Vittore Buzzi Children’s Hospital, Università degli Studi di Milano, 20154 Milan, Italy; (V.F.); (E.V.); (V.C.); (A.M.); (G.V.Z.)
- Pediatric and Adolescent Unit, Department of Internal Medicine, University of Pavia, 27100 Pavia, Italy
| | - Tudor Lucian Pop
- European Paediatric Association/Union of National European Paediatric Societies and Associations, 10115 Berlin, Germany; (T.L.P.); (F.C.C.); (M.P.-M.)
- Second Paediatric Clinic, Department of Mother and Child, University of Medicine and Pharmacy Iuliu Hatieganu, 400177 Cluj-Napoca, Romania
| | - Alessandra Mari
- Department of Pediatrics, Vittore Buzzi Children’s Hospital, Università degli Studi di Milano, 20154 Milan, Italy; (V.F.); (E.V.); (V.C.); (A.M.); (G.V.Z.)
| | - Gian Vincenzo Zuccotti
- Department of Pediatrics, Vittore Buzzi Children’s Hospital, Università degli Studi di Milano, 20154 Milan, Italy; (V.F.); (E.V.); (V.C.); (A.M.); (G.V.Z.)
| | - Fugen Cullu Cokugras
- European Paediatric Association/Union of National European Paediatric Societies and Associations, 10115 Berlin, Germany; (T.L.P.); (F.C.C.); (M.P.-M.)
- Paediatric Gastroenterology, Hepatology and Nutrition, Cerrahpasa Medical Faculty, Istanbul University, Istanbul 34000, Turkey
| | - Massimo Pettoello-Mantovani
- European Paediatric Association/Union of National European Paediatric Societies and Associations, 10115 Berlin, Germany; (T.L.P.); (F.C.C.); (M.P.-M.)
- Department of Pediatrics, Scientific Institute ‘Casa Sollievo della Sofferenza’, University of Foggia, 71122 Foggia, Italy
- Association pour l’Activité et la Recherche Scìentifiques, EPA-UNEPSA/ARS, 2000 Neuchâtel, Switzerland
| | - Olivier Goulet
- Department of Paediatric Gastroenterology, and Nutrition, Intestinal Failure Rehabilitation Centre, National Reference Centre for Rare Digestive Diseases, Necker-Enfants Malades Hospital, Paris Centre University and Paris-Descartes School of Medicine, 75000 Paris, France;
| |
Collapse
|
25
|
Ma M, Geng S, Liu M, Zhao L, Zhang J, Huang S, Ma Q. Effects of Different Methionine Levels in Low Protein Diets on Production Performance, Reproductive System, Metabolism, and Gut Microbiota in Laying Hens. Front Nutr 2021; 8:739676. [PMID: 34692750 PMCID: PMC8526799 DOI: 10.3389/fnut.2021.739676] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2021] [Accepted: 09/06/2021] [Indexed: 12/19/2022] Open
Abstract
This study investigated the effects of different levels of methionine (Met) in a low protein diet on the production performance, reproductive system, metabolism, and gut microbial composition of laying hens to reveal the underlying molecular mechanism of Met in a low protein diet on the host metabolism and gut microbial composition and function of hens. A total of 360 healthy 38-week-old Peking Pink laying hens with similar body conditions and egg production (EP) were randomly divided into four groups with nine replicates per treatment and 10 hens per replicate. The hens in each treatment group were fed low protein diets containing different levels of Met (0.25, 0.31, 0.38, and 0.47%, respectively) for 12 weeks. Feed and water were provided ad libitum throughout the trial period. The results showed that, compared with the 0.25% Met group, the final body weight (FBW), average daily gain (ADG), EP, egg weight (EW), and average daily feed intake (ADFI) in the other groups were significantly increased and feed egg ratio (FER) was decreased. Meanwhile, the EW and yield of abdominal fat (AFY) in the 0.47% Met group were higher than those in other groups. The triglyceride (TG), estradiol (E2), total protein (TP), albumin (ALB), and immunoglobulin A (IgA) in the 0.38 and 0.47% Met groups were higher than those in other groups. In addition, 16S rRNA gene sequencing revealed that there was no difference in the Sobs index, ACE index, and Shannon index among all groups. However, it is worth noting that feeding low protein diets with Met changed the gut microbial composition (e.g., the supplementation of Met increased the level of Lactobacillus and decreased the proportion of Faecalibacterium). Also, our results showed that the changes in gut microbial composition induced by the diets with different levels of Met were closely related to the changes of key parameters: ADFI, EW, FBW, TG, EM, EP, ADG, FER, and uric acid (UA). Our results highlight the role of adding an appropriate amount of Met to the low protein diet in laying hens, which could improve the gut microbial composition, production performance, reproductive system, and nutrient metabolism of laying hens. In conclusion, this study suggested that when the Met level was 0.38%, the production performance of the laying hens was pretty good.
Collapse
Affiliation(s)
- Miaolin Ma
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Shunju Geng
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Meiling Liu
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Lihong Zhao
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Jianyun Zhang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Shimeng Huang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Qiugang Ma
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| |
Collapse
|
26
|
Yang Y, Li X, Yang Y, Shoaie S, Zhang C, Ji B, Wei Y. Advances in the Relationships Between Cow's Milk Protein Allergy and Gut Microbiota in Infants. Front Microbiol 2021; 12:716667. [PMID: 34484158 PMCID: PMC8415629 DOI: 10.3389/fmicb.2021.716667] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Accepted: 07/22/2021] [Indexed: 12/24/2022] Open
Abstract
Cow's milk protein allergy (CMPA) is an immune response to cow's milk proteins, which is one of the most common food allergies in infants and young children. It is estimated that 2-3% of infants and young children have CMPA. The diet, gut microbiota, and their interactions are believed to be involved in the alterations of mucosal immune tolerance, which might lead to the development of CMPA and other food allergies. In this review, the potential molecular mechanisms of CMPA, including omics technologies used for analyzing microbiota, impacts of early microbial exposures on CMPA development, and microbiota-host interactions, are summarized. The probiotics, prebiotics, synbiotics, fecal microbiota transplantation, and other modulation strategies for gut microbiota and the potential application of microbiota-based design of diets for the CMPA treatment are also discussed. This review not only summarizes the current studies about the interactions of CMPA with gut microbiota but also gives insights into the possible CMPA treatment strategies by modulating gut microbiota, which might help in improving the life quality of CMPA patients in the future.
Collapse
Affiliation(s)
- Yudie Yang
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, School of Pharmaceutical Sciences, Henan Children’s Hospital, Zhengzhou Children’s Hospital, Children’s Hospital Affiliated to Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Xiaoqi Li
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, School of Pharmaceutical Sciences, Henan Children’s Hospital, Zhengzhou Children’s Hospital, Children’s Hospital Affiliated to Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Ying Yang
- Jing’an District Central Hospital of Shanghai, Jing’an Branch, Huashan Hospital, Fudan University, Shanghai, China
| | - Saeed Shoaie
- Science for Life Laboratory, KTH Royal Institute of Technology, Stockholm, Sweden
- Faculty of Dentistry, Oral and Craniofacial Sciences, Centre for Host-Microbiome Interactions, King’s College London, London, United Kingdom
| | - Cheng Zhang
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, School of Pharmaceutical Sciences, Henan Children’s Hospital, Zhengzhou Children’s Hospital, Children’s Hospital Affiliated to Zhengzhou University, Zhengzhou University, Zhengzhou, China
- Science for Life Laboratory, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Boyang Ji
- Department of Biology and Biological Engineering, Chalmers University of Technology, Gothenburg, Sweden
| | - Yongjun Wei
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, School of Pharmaceutical Sciences, Henan Children’s Hospital, Zhengzhou Children’s Hospital, Children’s Hospital Affiliated to Zhengzhou University, Zhengzhou University, Zhengzhou, China
- Laboratory of Synthetic Biology, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
27
|
Ojima MN, Yoshida K, Sakanaka M, Jiang L, Odamaki T, Katayama T. Ecological and molecular perspectives on responders and non-responders to probiotics and prebiotics. Curr Opin Biotechnol 2021; 73:108-120. [PMID: 34375845 DOI: 10.1016/j.copbio.2021.06.023] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 06/22/2021] [Accepted: 06/23/2021] [Indexed: 12/20/2022]
Abstract
Bifidobacteria are widely used as a probiotic for their health-promoting effects. To promote their growth, bifidogenic prebiotics, including human milk oligosaccharides (HMOs), have been added to supplements and infant formula. However, the efficacy of both probiotic and prebiotic interventions is often debated, as clinical responses vary significantly by case. Here, we review clinical studies that aimed to proliferate human-residential Bifidobacterium (HRB) strains in the gut, and we highlight the difference between responders and non-responders to such interventions through an ecological, niche-based perspective and an examination of the prevalence of genes responsible for prebiotic assimilation in HRB genomes. We discuss the criteria necessary to better evaluate the efficacy of probiotic and prebiotic interventions and the recent therapeutic potential shown by synbiotics.
Collapse
Affiliation(s)
- Miriam N Ojima
- Graduate School of Biostudies, Kyoto University, Kyoto, 606-8502, Japan
| | - Keisuke Yoshida
- Next Generation Science Institute, Morinaga Milk Industry Co., Ltd., Zama, Kanagawa, 252-8583, Japan
| | - Mikiyasu Sakanaka
- Graduate School of Biostudies, Kyoto University, Kyoto, 606-8502, Japan
| | - Lin Jiang
- School of Biology, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Toshitaka Odamaki
- Graduate School of Biostudies, Kyoto University, Kyoto, 606-8502, Japan; Next Generation Science Institute, Morinaga Milk Industry Co., Ltd., Zama, Kanagawa, 252-8583, Japan
| | - Takane Katayama
- Graduate School of Biostudies, Kyoto University, Kyoto, 606-8502, Japan.
| |
Collapse
|
28
|
Co-Encapsulated Synbiotics and Immobilized Probiotics in Human Health and Gut Microbiota Modulation. Foods 2021; 10:foods10061297. [PMID: 34200108 PMCID: PMC8230215 DOI: 10.3390/foods10061297] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 05/31/2021] [Accepted: 06/02/2021] [Indexed: 12/20/2022] Open
Abstract
Growing interest in the development of innovative functional products as ideal carriers for synbiotics, e.g., nutrient bars, yogurt, chocolate, juice, ice cream, and cheese, to ensure the daily intake of probiotics and prebiotics, which are needed to maintain a healthy gut microbiota and overall well-being, is undeniable and inevitable. This review focuses on the modern approaches that are currently being developed to modulate the gut microbiota, with an emphasis on the health benefits mediated by co-encapsulated synbiotics and immobilized probiotics. The impact of processing, storage, and simulated gastrointestinal conditions on the viability and bioactivity of probiotics together with prebiotics such as omega-3 polyunsaturated fatty acids, phytochemicals, and dietary fibers using various delivery systems are considered. Despite the proven biological properties of synbiotics, research in this area needs to be focused on the proper selection of probiotic strains, their prebiotic counterparts, and delivery systems to avoid suppression of their synergistic or complementary effect on human health. Future directions should lead to the development of functional food products containing stable synbiotics tailored for different age groups or specifically designed to fulfill the needs of adjuvant therapy.
Collapse
|
29
|
Sorensen K, Cawood AL, Gibson GR, Cooke LH, Stratton RJ. Amino Acid Formula Containing Synbiotics in Infants with Cow's Milk Protein Allergy: A Systematic Review and Meta-Analysis. Nutrients 2021; 13:935. [PMID: 33799379 PMCID: PMC7998621 DOI: 10.3390/nu13030935] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 03/08/2021] [Accepted: 03/10/2021] [Indexed: 12/15/2022] Open
Abstract
Cow's milk protein allergy (CMPA) is associated with dysbiosis of the infant gut microbiome, with allergic and immune development implications. Studies show benefits of combining synbiotics with hypoallergenic formulae, although evidence has never been systematically examined. This review identified seven publications of four randomised controlled trials comparing an amino acid formula (AAF) with an AAF containing synbiotics (AAF-Syn) in infants with CMPA (mean age 8.6 months; 68% male, mean intervention 27.3 weeks, n = 410). AAF and AAF-Syn were equally effective in managing allergic symptoms and promoting normal growth. Compared to AAF, significantly fewer infants fed AAF-Syn had infections (OR 0.35 (95% CI 0.19-0.67), p = 0.001). Overall medication use, including antibacterials and antifectives, was lower among infants fed AAF-Syn. Significantly fewer infants had hospital admissions with AAF-Syn compared to AAF (8.8% vs. 20.2%, p = 0.036; 56% reduction), leading to potential cost savings per infant of £164.05-£338.77. AAF-Syn was associated with increased bifidobacteria (difference in means 31.75, 95% CI 26.04-37.45, p < 0.0001); reduced Eubacterium rectale and Clostridium coccoides (difference in means -19.06, 95% CI -23.15 to -14.97, p < 0.0001); and reduced microbial diversity (p < 0.05), similar to that described in healthy breastfed infants, and may be associated with the improved clinical outcomes described. This review provides evidence that suggests combining synbiotics with AAF produces clinical benefits with potential economic implications.
Collapse
Affiliation(s)
- Katy Sorensen
- Medical Affairs, Nutricia Ltd., White Horse Business Park, Trowbridge BA14 0XQ, UK
| | - Abbie L. Cawood
- Medical Affairs, Nutricia Ltd., White Horse Business Park, Trowbridge BA14 0XQ, UK
- Institute of Human Nutrition, Faculty of Medicine, Mailpoint 113, Southampton General Hospital, Tremona Road, Southampton SO16 6YD, UK; (A.L.C.); (R.J.S.)
| | - Glenn R. Gibson
- Department of Food and Nutritional Sciences, University of Reading, Whiteknights, Reading RG6 6AP, UK;
| | - Lisa H. Cooke
- Department of Nutrition and Dietetics, Bristol Royal Hospital for Children, Upper Maudlin Street, Bristol BS2 8BJ, UK;
| | - Rebecca J. Stratton
- Medical Affairs, Nutricia Ltd., White Horse Business Park, Trowbridge BA14 0XQ, UK
- Institute of Human Nutrition, Faculty of Medicine, Mailpoint 113, Southampton General Hospital, Tremona Road, Southampton SO16 6YD, UK; (A.L.C.); (R.J.S.)
| |
Collapse
|