1
|
Ray D, Bose P, Mukherjee S, Roy S, Kaity S. Recent drug delivery systems targeting the gut-brain-microbiome axis for the management of chronic diseases. Int J Pharm 2025; 680:125776. [PMID: 40425058 DOI: 10.1016/j.ijpharm.2025.125776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2025] [Revised: 05/14/2025] [Accepted: 05/24/2025] [Indexed: 05/29/2025]
Abstract
In recent years, the study of microorganisms and the brain has become increasingly connected. The gut-brain-microbiome axis (GBMA), a bi-directional communication system, is the key part of how the body's bacteria and the brain interact. This system can influence the brain and behaviour. Changes in this relationship have been linked to various mental and physical health conditions. The immune system, tryptophan metabolism, the vagus nerve, and the enteric nervous system all facilitate connections between the gut and brain. Microbes produce Peptidoglycans, branched-chain amino acids, and short-chain fatty acids, which are involved in this communication. Studies suggest the gut microbiome may be involved in conditions like autism, anxiety, obesity, schizophrenia, Parkinson's disease, and Alzheimer's disease. Researchers are exploring the gut-brain connection to cure a variety of disorders, such as neurological disorders, cancers, metabolic problems, and liver diseases. Developing novel drug delivery systems is a key focus in GBMA for therapeutic targeting at various disease pathways. Notable platforms attracting significant interest include silica nanoparticle-based delivery systems for probiotic spores, composite hydrogels formulated from protein isolates and citrus pectin, and biomimetic nanosystems designed for targeted therapeutic delivery. This review summarizes different methods of delivering drugs and using dietary interventions to target the GBMA and treat these conditions in a less invasive way. By understanding how the gut and brain communicate, scientists aim to develop new and more effective therapies for these complex chronic diseases.
Collapse
Affiliation(s)
- Debjani Ray
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Kolkata, West Bengal, India
| | - Piyas Bose
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Kolkata, West Bengal, India
| | - Saptarshi Mukherjee
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Kolkata, West Bengal, India
| | - Subhadeep Roy
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Kolkata, West Bengal, India
| | - Santanu Kaity
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Kolkata, West Bengal, India.
| |
Collapse
|
2
|
Liu P, Zuo J, Lu H, Zhang B, Wu C. Bacillus subtilis Fed to Sows Promotes Intestinal Development and Regulates Mucosal Immunity in Offspring. Vet Sci 2025; 12:489. [PMID: 40431582 PMCID: PMC12116194 DOI: 10.3390/vetsci12050489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2025] [Revised: 05/14/2025] [Accepted: 05/14/2025] [Indexed: 05/29/2025] Open
Abstract
Diarrhea in piglets causes intestinal inflammation and epithelial damage. Weaned piglets fed with Bacillus subtilis (B.S) have enhanced intestinal mucosal immunity and reduces diarrhea in piglets. However, the immune system of newborn piglets is immature, and B.S cannot effectively activate the intestinal mucosal reaction when given directly. This research explored the impact of the maternal supplementation of B.S-Dia during the final 35 days of gestation on piglet intestinal development and mucosal immunity. The results demonstrated that B.S-Dia administration significantly increased the body weight, jejunal villus height, and crypt depth in the piglets. In addition, B.S-Dia also significantly increased the proliferative activity of intestinal epithelial cells, as evidenced by proliferating cell nuclear antigen (PCNA) staining and the elevated mRNA expression of the proliferation-related gene (c-Myc). Furthermore, B.S-Dia supplementation also reinforced the intestinal mucosal barrier by increasing goblet cell numbers and upregulating the mRNA expression of antimicrobial peptides, such as Muc2 and Lyz-1. Finally, elevated levels of IL-4 and IFN-γ, along with an increased abundance of CD3+ T cells, revealed that the intestinal mucosal immunity of piglets was improved after B.S-Dia administration. Our study indicates that feeding B.S-Dia to sow spromotes intestinal development and improves intestinal mucosal immunity in piglets.
Collapse
Affiliation(s)
- Peng Liu
- College of Veterinary Medicine, Jiangsu Agri-Animal Husbandry Vocational College, Taizhou 225300, China;
| | - Jinjiao Zuo
- Pet Science and Technology College, Jiangsu Agri-Animal Husbandry Vocational College, Taizhou 225300, China; (J.Z.); (B.Z.)
| | - Hui Lu
- College of Veterinary Medicine, Jiangsu Agri-Animal Husbandry Vocational College, Taizhou 225300, China;
| | - Bin Zhang
- Pet Science and Technology College, Jiangsu Agri-Animal Husbandry Vocational College, Taizhou 225300, China; (J.Z.); (B.Z.)
| | - Caihong Wu
- College of Veterinary Medicine, Jiangsu Agri-Animal Husbandry Vocational College, Taizhou 225300, China;
| |
Collapse
|
3
|
Herrington RTB, Ellenberger DT, Rosenfeld CS. Maternal probiotic supplementation and effects on the fetal placenta. Biol Reprod 2025; 112:807-823. [PMID: 40057969 DOI: 10.1093/biolre/ioaf041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 02/14/2025] [Accepted: 03/03/2025] [Indexed: 05/16/2025] Open
Abstract
Increasing number of pregnant women are consumi ng probiotics to promote their own health and that of their unborn fetuses. Such supplements are presumed to be safe for pregnant mothers and their unborn offspring. For pregnant mothers, such bioactive compounds might lower the risk of constipation, diarrhea, other gastrointestinal conditions, and pre-term birth, and prevent adverse pregnancy outcomes, including gestational diabetes mellitus and depression/anxiety. More research is needed to examine potential safety of probiotic consumption during pregnancy and long-term health consequences to offspring. The conceptus can also be indirectly affected by maternal probiotic supplementation through microorganism production of bioactive compounds. The placenta is in direct communication with the underlying uterine tissue. Thus, compounds in the maternal blood can easily transfer across the placenta and impact this hormonally sensitive organ. Select studies suggest that disruptions to the maternal microbiome dramatically affect the placenta. In the current review, we will therefore consider the evidence to date of how maternal probiotic supplementation affects the placenta. Three potential mechanisms we will explore include the possibility that maternal probiotic supplementation might impact the putative placenta microbiome. The second potential mechanism we will consider is that maternal probiotic consumption alters bacterial-derived metabolites, including short-chained fatty acids, polyamines, Vitamin B9, and Vitamin B12. The third potential mechanism to be discussed is that such supplements affect maternal and placental immune responses. Before probiotics are promoted for healthy pregnant women and those with gestational disorders, more studies, including those examining the effects on the placenta, are essential.
Collapse
Affiliation(s)
- Rosalind T B Herrington
- Biomedical Sciences, University of Missouri, Columbia, Missouri 65211, USA
- Biological Sciences, University of Missouri, Columbia, Missouri 65211, USA
| | - David T Ellenberger
- Biomedical Sciences, University of Missouri, Columbia, Missouri 65211, USA
- Microbiology, University of Missouri Columbia, Missouri 65211, USA
| | - Cheryl S Rosenfeld
- Biomedical Sciences, University of Missouri, Columbia, Missouri 65211, USA
- MU Institute for Data Science and Informatics, University of Missouri, Columbia, Missouri 65211, USA
- Department of Genetics Area Program, University of Missouri, Columbia, Missouri 65211, USA
- Department of Thompson Center for Autism and Neurobehavioral Disorders, University of Missouri, Columbia, Missouri 65211, USA
| |
Collapse
|
4
|
Chen L, Tian L, Zhang Y, Shi Y, Yuan W, Zou Y, Zhang Q, Chen M, Zeng P. Updated Insights into Probiotic Interventions for Metabolic Syndrome: Mechanisms and Evidence. Probiotics Antimicrob Proteins 2025:10.1007/s12602-025-10554-x. [PMID: 40332670 DOI: 10.1007/s12602-025-10554-x] [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] [Accepted: 04/17/2025] [Indexed: 05/08/2025]
Abstract
Metabolic syndrome (MetS) is a disease with complex and diverse etiologies. Extrinsic factors such as diet and lifestyle can induce dysbiosis of gut microbes, compromising intestinal barrier integrity and leading to inflammation and insulin resistance, thereby advancing MetS. Probiotic interventions have shown potential in ameliorating gut microbiota dysbiosis and regulating host metabolism by assimilating lipids, metabolizing carbohydrates, and producing short-chain fatty acids (SCFA), indole compounds, secondary bile acids, conjugated linoleic acid (CLA), and other active ingredients. An increasing number of new strains are being isolated and validated for their effective roles intervening on MetS in animal and population studies. This review aims to provide updated insights into the pathogenic mechanisms of MetS, highlight the newly identified probiotic strains that have demonstrated improvements in MetS, and elucidate their mechanisms of action, with the aim of offering contemporary perspectives for the future use of probiotics in mitigating MetS.
Collapse
Affiliation(s)
- Lili Chen
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, 610000, People's Republic of China
| | - Lvbo Tian
- Sichuan International Travel Health Care Center (Chengdu Customs Port Clinic), Chengdu, 610000, People's Republic of China
| | - Yuqi Zhang
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, 610000, People's Republic of China
| | - Ying Shi
- Sichuan International Travel Health Care Center (Chengdu Customs Port Clinic), Chengdu, 610000, People's Republic of China
| | - Wenyi Yuan
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, 610000, People's Republic of China
| | - Yue Zou
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, 610000, People's Republic of China
| | - Qin Zhang
- Sichuan International Travel Health Care Center (Chengdu Customs Port Clinic), Chengdu, 610000, People's Republic of China
| | - Moutong Chen
- State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangdong 510070, Guangzhou, China
| | - Peibin Zeng
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, 610000, People's Republic of China.
| |
Collapse
|
5
|
Chen Y, Peng Y, Niu Q, Jiang Y, Ni H, Chen L, Lin Y. The impact of probiotics therapy on cognitive and metabolic characteristics in patients with cognitive impairment: An umbrella review of meta-analysis of randomized controlled trials. Eur J Pharmacol 2025; 994:177326. [PMID: 39914784 DOI: 10.1016/j.ejphar.2025.177326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Revised: 01/29/2025] [Accepted: 01/30/2025] [Indexed: 03/15/2025]
Abstract
BACKGROUND While existing meta-analysis suggest that probiotic therapies may enhance cognitive functions and influence metabolic characteristics, the findings have been inconclusive. In light of these discrepancies, our study undertakes an umbrella review to more precisely determine the aggregate effects and rigorously evaluate the credibility and quality of evidence. METHODS We conducted a systematic search across seven databases, including PubMed, Embase, the Cochrane Library, ProQuest, Web of Science, CINAHL, and Scopus, from their inception to June 20, 2024. We utilized the AMSTAR-2 tool to evaluate the quality of the meta-analyses and applied the GRADE system to rate the quality of the evidence. We estimated the final effect sizes (ESs) along with their 95% confidence intervals (CI) and performed both sensitivity and subgroup analyses to explore the sources of heterogeneity. RESULTS Among the 314 articles identified in our search, 13 meta-analysis that met the criteria were included in the study. The quality of the evidence in these studies was graded from high to very low. Our results demonstrate that probiotic treatment significantly enhances cognitive function in patients (ESSMD = 0.39, 95%CI: 0.19 to 0.59, p < 0.001). Moreover, probiotic treatment notably decreased level of serum malondialdehyde (MDA), insulin resistance as detected by homeostasis model assessment method (HOMA-IR) and high-sensitivity C-reactive protein (hs-CRP) (p < 0.001). Conversely, probiotic treatment did not significantly impact triglycerides and very-low-density lipoprotein (VLDL) in patients (p > 0.05). CONCLUSION Although preliminary evidence indicated that probiotic therapy may positively impact cognitive function, MDA, HOMA-IR, and hs-CRP, the overall quality of the existing evidence is insufficient to provide strong support. Therefore, future research must employ more rigorous designs or initiate larger clinical trials to produce more compelling evidence to further validate the efficacy of probiotic therapy on cognitive function of patients with cognitive dysfunction.
Collapse
Affiliation(s)
- Yaqin Chen
- School of Nursing, Fujian Medical University, Fuzhou, Fujian, China
| | - Yanchun Peng
- Department of Nursing, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| | - Qi Niu
- School of Nursing, Fujian Medical University, Fuzhou, Fujian, China
| | - Yan Jiang
- School of Nursing, Fujian Medical University, Fuzhou, Fujian, China
| | - Hong Ni
- Department of Nursing, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| | - Liangwan Chen
- Department of Cardiac Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, China; Fujian Provincial Special Reserve Talents Laboratory, Fuzhou, Fujian, China.
| | - Yanjuan Lin
- Department of Nursing, Fujian Medical University Union Hospital, Fuzhou, Fujian, China; Department of Cardiac Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, China.
| |
Collapse
|
6
|
Kumar H, Dhanjal DS, Dhalaria R, Kimta N, Cimler R, Kuča K. Dysbiosis significantly elevates the probability of altered affective function in Alzheimer disease (AD). INTERNATIONAL REVIEW OF NEUROBIOLOGY 2025; 180:1-24. [PMID: 40414630 DOI: 10.1016/bs.irn.2025.04.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2025]
Abstract
Changes in the makeup of gut microbiota are linked to many neuropsychiatric diseases. Although the exact connection between gut dysbiosis and brain dysfunction is not yet fully understood, but recent data suggests that gut dysbiosis may contribute to the development of Alzheimer's disease (AD) by promoting neuroinflammation, insulin resistance, oxidative stress, and amyloid-beta (Aβ) aggregation. Gut dysbiosis in animal models is primarily characterized by an elevated ratio of Firmicutes/Bacteroidetes which may lead to the accumulation of amyloid precursor protein (APP) in the intestine, in the early stages of AD. Probiotics play a significant role in preventing against the symptoms of AD by restoring gut-brain homeostasis. This chapter provides an overview of the gut microbiota and its dysregulation in etiology of AD. Moreover, novel insights into alteration of the composition of gut microbiota as a preventive or therapeutic approach to AD are discussed.
Collapse
Affiliation(s)
- Harsh Kumar
- Centre of Advanced Technologies, Faculty of Science, University of Hradec Kralove, Rokitanskeho, Hradec Kralove, Czech Republic
| | - Daljeet Singh Dhanjal
- School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, India
| | - Rajni Dhalaria
- School of Biological and Environmental Sciences, Shoolini University of Biotechnology and Management Sciences, Solan, India
| | - Neetika Kimta
- School of Biological and Environmental Sciences, Shoolini University of Biotechnology and Management Sciences, Solan, India
| | - Richard Cimler
- Centre of Advanced Technologies, Faculty of Science, University of Hradec Kralove, Rokitanskeho, Hradec Kralove, Czech Republic
| | - Kamil Kuča
- Department of Chemistry, Faculty of Science, University of Hradec Kralove, Hradec Kralove, Czech Republic.
| |
Collapse
|
7
|
Mendez-Hernandez R, Braga I, Bali A, Yang M, de Lartigue G. Vagal Sensory Gut-Brain Pathways That Control Eating-Satiety and Beyond. Compr Physiol 2025; 15:e70010. [PMID: 40229922 DOI: 10.1002/cph4.70010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2025] [Revised: 03/20/2025] [Accepted: 03/31/2025] [Indexed: 04/16/2025]
Abstract
The vagus nerve is the body's primary sensory conduit from gut to brain, traditionally viewed as a passive relay for satiety signals. However, emerging evidence reveals a far more complex system-one that actively encodes diverse aspects of meal-related information, from mechanical stretch to nutrient content, metabolic state, and even microbial metabolites. This review challenges the view of vagal afferent neurons (VANs) as simple meal-termination sensors and highlights their specialized subpopulations, diverse sensory modalities, and downstream brain circuits, which shape feeding behavior, metabolism, and cognition. We integrate recent advances from single-cell transcriptomics, neural circuit mapping, and functional imaging to examine how VANs contribute to gut-brain communication beyond satiety, including their roles in food reward and memory formation. By synthesizing the latest research and highlighting emerging directions for the field, this review provides a comprehensive update on vagal sensory pathways and their role as integrators of meal information.
Collapse
Affiliation(s)
- Rebeca Mendez-Hernandez
- Monell Chemical Senses Center, Philadelphia, Pennsylvania, USA
- Department of Neuroscience, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Isadora Braga
- Monell Chemical Senses Center, Philadelphia, Pennsylvania, USA
- Department of Neuroscience, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Avnika Bali
- Monell Chemical Senses Center, Philadelphia, Pennsylvania, USA
- Department of Neuroscience, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Mingxin Yang
- Monell Chemical Senses Center, Philadelphia, Pennsylvania, USA
- Department of Neuroscience, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Guillaume de Lartigue
- Monell Chemical Senses Center, Philadelphia, Pennsylvania, USA
- Department of Neuroscience, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
8
|
Zhu J, Peng F, Yang H, Luo J, Zhang L, Chen X, Liao H, Lei H, Liu S, Yang T, Luo G, Chen G, Zhao H. Probiotics and muscle health: the impact of Lactobacillus on sarcopenia through the gut-muscle axis. Front Microbiol 2025; 16:1559119. [PMID: 40160272 PMCID: PMC11952772 DOI: 10.3389/fmicb.2025.1559119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Accepted: 02/24/2025] [Indexed: 04/02/2025] Open
Abstract
Sarcopenia refers to the decline in skeletal muscle mass and function. Due to its increased mortality rate and severe disability, the clinical importance of sarcopenia is becoming increasingly prominent. Although the exact cause of sarcopenia is not fully understood, the gut microbiota (GM) plays a crucial role in the pathogenesis of sarcopenia, and increasing evidence suggests that gut dysbiosis may be associated with disease development. In the past few decades, the use of probiotics has surged, few studies have explored their impact on sarcopenia prevention and treatment. Lactobacillus probiotics are commonly used for gut health and immune support, but their mechanism in sarcopenia via the gut-muscle axis remains uncertain. This review highlights the treatment challenges, GM's role in sarcopenia, and the potential of Lactobacillus as an adjunct therapy. In addition, we also discuss the possible mechanisms by which Lactobacillus affect muscle function, such as alleviating inflammatory states, clearing excessive reactive oxygen species (ROS), improving skeletal muscle metabolism, enhancing intestinal barrier function and modulating the gut microbiota and its metabolites. These mechanisms may collectively contribute to the preservation of muscle mass and function, offering a promising avenue for advancing microbial therapies for sarcopenia.
Collapse
Affiliation(s)
- Jingjun Zhu
- Department of Radiology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Fei Peng
- Department of Radiology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Huixin Yang
- Changde Hospital, Xiangya School of Medicine, Central South University (The First People’s Hospital of Changde City), Changde, China
| | - Jing Luo
- Department of Radiology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Li Zhang
- Department of Radiology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Xiaolong Chen
- Department of Radiology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Huazhi Liao
- Department of Radiology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Hao Lei
- Department of Radiology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Shuai Liu
- Department of Hepatobiliary Pancreatic Surgery, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Tingqian Yang
- Department of Radiology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- Department of Radiology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Guanghua Luo
- Department of Radiology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Guodong Chen
- Department of Hepatobiliary Pancreatic Surgery, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Heng Zhao
- Department of Radiology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| |
Collapse
|
9
|
Peña-Durán E, García-Galindo JJ, López-Murillo LD, Huerta-Huerta A, Balleza-Alejandri LR, Beltrán-Ramírez A, Anaya-Ambriz EJ, Suárez-Rico DO. Microbiota and Inflammatory Markers: A Review of Their Interplay, Clinical Implications, and Metabolic Disorders. Int J Mol Sci 2025; 26:1773. [PMID: 40004236 PMCID: PMC11854938 DOI: 10.3390/ijms26041773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 02/12/2025] [Accepted: 02/14/2025] [Indexed: 02/27/2025] Open
Abstract
The human microbiota, a complex ecosystem of microorganisms, plays a pivotal role in regulating host immunity and metabolism. This review investigates the interplay between microbiota and inflammatory markers, emphasizing their impact on metabolic and autoimmune disorders. Key inflammatory biomarkers, such as C-reactive protein (CRP), interleukin-6 (IL-6), lipopolysaccharides (LPS), zonulin (ZO-1), and netrin-1 (Ntn1), are discussed in the context of intestinal barrier integrity and chronic inflammation. Dysbiosis, characterized by alterations in microbial composition and function, directly modulates the levels and activity of these biomarkers, exacerbating inflammatory responses and compromising epithelial barriers. The disruption of microbiota is further correlated with increased intestinal permeability and chronic inflammation, serving as a precursor to conditions like type 2 diabetes (T2D), obesity, and non-alcoholic fatty liver disease. Additionally, this review examines therapeutic strategies, including probiotics and prebiotics, designed to restore microbial balance, mitigate inflammation, and enhance metabolic homeostasis. Emerging evidence positions microbiota-targeted interventions as critical components in the advancement of precision medicine, offering promising avenues for diagnosing and treating inflammatory and metabolic disorders.
Collapse
Affiliation(s)
- Emiliano Peña-Durán
- Licenciatura en Médico Cirujano y Partero, Centro Universitario de Ciencias de la Salud (CUCS), Universidad de Guadalajara, Guadalajara 44340, Mexico
| | - Jesús Jonathan García-Galindo
- Departamento de Fisiología, Centro Universitario de Ciencias de la Salud (CUCS), Universidad de Guadalajara, Calle Sierra Mojada 950, Independencia Oriente, Guadalajara 44340, Mexico
- Departamento Académico Aparatos y Sistemas II, Decanato de Ciencias de la Salud, Universidad Autónoma de Guadalajara, Zapopan 44670, Mexico
| | - Luis Daniel López-Murillo
- Departamento de Fisiología, Centro Universitario de Ciencias de la Salud (CUCS), Universidad de Guadalajara, Calle Sierra Mojada 950, Independencia Oriente, Guadalajara 44340, Mexico
- Departamento Académico Aparatos y Sistemas I, Decanato de Ciencias de la Salud, Universidad Autónoma de Guadalajara, Zapopan 44670, Mexico
| | - Alfredo Huerta-Huerta
- Hospital Medica de la Ciudad, Santa Catalina, Calle. Pablo Valdez 719, La Perla, Guadalajara 44360, Mexico
| | - Luis Ricardo Balleza-Alejandri
- Doctorado en Farmacología, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Mexico
| | - Alberto Beltrán-Ramírez
- Departamento de Fisiología, Centro Universitario de Ciencias de la Salud (CUCS), Universidad de Guadalajara, Calle Sierra Mojada 950, Independencia Oriente, Guadalajara 44340, Mexico
- Departamento Académico Aparatos y Sistemas I, Decanato de Ciencias de la Salud, Universidad Autónoma de Guadalajara, Zapopan 44670, Mexico
| | - Elsa Janneth Anaya-Ambriz
- Departamento de Ciencias de la Salud, Centro Universitario de los Valles, Universidad de Guadalajara, Ameca 46708, Mexico
| | - Daniel Osmar Suárez-Rico
- Departamento de Fisiología, Centro Universitario de Ciencias de la Salud (CUCS), Universidad de Guadalajara, Calle Sierra Mojada 950, Independencia Oriente, Guadalajara 44340, Mexico
- Departamento Académico Aparatos y Sistemas II, Decanato de Ciencias de la Salud, Universidad Autónoma de Guadalajara, Zapopan 44670, Mexico
- Departamento de Farmacobiología, Centro Universitario de Ciencias Exactas e Ingenierías (CUCEI), Universidad de Guadalajara, Guadalajara 44430, Mexico
| |
Collapse
|
10
|
Ramzan H, Bukhari DA, Bibi Z, Arifullah, Isha, Nawaz A, Rehman A. Probiotic supplement for the treatment of polycystic ovarian syndrome. Pharmacol Ther 2025; 266:108785. [PMID: 39719172 DOI: 10.1016/j.pharmthera.2024.108785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 11/24/2024] [Accepted: 12/19/2024] [Indexed: 12/26/2024]
Abstract
Polycystic Ovarian Syndrome is one of the major prevalent causes of infertility reported worldwide nearly 6-26 %, especially in girls hitting puberty and women at their childbearing age. The main clinical manifestations include irregular menstrual cycle, small cysts on one or both ovaries, chronic oligo-anovulation, and hirsutism. The etiological criteria are very complex and related to many factors like obesity, insulin sensitivity, inflammation, hyperandrogenism, diabetes mellitus type II, cardiovascular diseases, and dysbiosis of gut microbiota. The given review focuses on managing PCOS through probiotics by analyzing the effects on the symptoms of the disease. The probiotics effective in treating PCOS belong to Bifidobacterium, Lactobacilli, Clostridium, Enterococcus, and other Lactic acid bacteria. Its significance in PCOS is mainly due to the antagonizing of the growth of pathogenic microorganisms, increasing intestinal mucus layer production, reducing intestinal permeability, and modulating the gastrointestinal immune system. Also, their interaction with certain hormones such as insulin, androgen, and estrogen through short-chain fatty acids influences fertility. More research is necessary to validate these results. Probiotic supplements could be a viable option for treating PCOS in adults.
Collapse
Affiliation(s)
- Habiba Ramzan
- Department of Zoology, Government College University, Lahore, Pakistan
| | | | - Zuhra Bibi
- Department of Zoology, Government College University, Lahore, Pakistan
| | - Arifullah
- Department of Zoology, Government College University, Lahore, Pakistan
| | - Isha
- Department of Zoology, Government College University, Lahore, Pakistan
| | - Atif Nawaz
- Department of Zoology, Government College University, Lahore, Pakistan
| | - Abdul Rehman
- Institute of Microbiology and Molecular Genetics, University of the Punjab, New Campus, Lahore 54590, Pakistan.
| |
Collapse
|
11
|
Misra S, Rajput P, Kaur A. Tirzepatide mitigates cognitive decline in zebrafish model of type 2 diabetes mellitus induced by high-fat diet. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-03827-3. [PMID: 39873719 DOI: 10.1007/s00210-025-03827-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Accepted: 01/17/2025] [Indexed: 01/30/2025]
Abstract
In examining the enduring consequences of diabetes, recent research has focused on the anticipated outcomes of the condition. Specifically, cognitive impairment has been linked to diabetes mellitus dating back to the discovery of insulin. This study delves into the neuroprotective effects of TZP, i.e. tirzepatide a dual GIP and GLP-1 receptor agonist that works by mimicking these two gut hormones, against cognitive impairment associated with type 2 diabetes mellitus (T2DM). T2DM-like zebrafish model of varying age groups was created through a 6-week administration of a high-fat diet (HFD). Parameters such as body weight, body mass index, and blood glucose levels were monitored, and behavioural assessments (T-maze, novel tank diving test, and inhibitory avoidance test) were conducted at the conclusion of the protocol to assess learning and memory. Additionally, lipid profile biochemical parameters (MDA, AChEs, and GSH), molecular markers (IL-1β, IL-10, TNF-α, Bcl-2, Bax, GSK-3β, and AMPK), and histopathological examinations were performed. Treatment with the novel GLP-1 and GIP dual agonist TZP (10 nM/kg, i.p.) significantly ameliorated cognitive impairment, as evidenced by behavioural parameters, and restored antioxidant like GSH (p < 0.05) and catalase (p < 0.05) and anti-inflammatory marker levels, i.e. IL-10 (p < 0.05) compared to the HFD group. TZP also mitigated abnormal glucose (73.2 ± 5.889) and lipid profiles (TG 0.159 ± 0.0075 and TC 0.100 ± 0.0020) in hyperglycaemic zebrafish. This study suggests that the positive effects of TZP on cognition and memory may stem from its neuroprotective capabilities, potentially attributed to its antioxidant, anti-inflammatory, and anti-apoptotic properties, as well as its ability to enhance AMPK levels as GLP-1 agonist has the potential to increase the level of AMPK.
Collapse
Affiliation(s)
- Sakshi Misra
- Department of Pharmacology, ISF College of Pharmacy, Ghal Kalan, GT Road, Moga, 142001, Punjab, India
| | - Prabha Rajput
- Department of Pharmacology, ISF College of Pharmacy, Ghal Kalan, GT Road, Moga, 142001, Punjab, India.
- Department of Pharmacology, School of Pharmacy & Technology Management, SVKM's NMIMS University, Shirpur Campus, Shirpur, India.
| | - Amandeep Kaur
- Department of Pharmacology, ISF College of Pharmacy, Ghal Kalan, GT Road, Moga, 142001, Punjab, India
| |
Collapse
|
12
|
Lapolla A, Dalfrà MG, Marelli G, Parrillo M, Sciacca L, Sculli MA, Succurro E, Torlone E, Vitacolonna E. Medical nutrition therapy in physiological pregnancy and in pregnancy complicated by obesity and/or diabetes: SID-AMD recommendations. Acta Diabetol 2025:10.1007/s00592-024-02442-7. [PMID: 39841216 DOI: 10.1007/s00592-024-02442-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Accepted: 12/27/2024] [Indexed: 01/23/2025]
Abstract
Proper nutrition is essential during pregnancy to ensure an adequate supply of nutrients to the foetus and adequate maternal weight gain. In pregnancy complicated by diabetes (both gestational and pre-gestational), diet in terms of both the intake and quality of carbohydrates is an essential factor in glycaemic control. Maternal BMI at conception defines the correct weight increase during gestation in order to reduce maternal-foetal complications related to hypo- or hyper-nutrition. The recommendations presented here, which are based on national and international guidelines and the most recently published data on nutrition in physiological pregnancy and pregnancy complicated by hyperglycaemia and/or obesity, are designed to help healthcare professionals prescribe suitable eating patterns to safeguard the health of the mother and the foetus.
Collapse
Affiliation(s)
| | | | - Giuseppe Marelli
- Ordine Ospedaliero San Giovanni di Dio Fatebenefratelli, Erba, CO, Italy
| | - Mario Parrillo
- UOSD Endocrinologia e Malattie del Ricambio, AO Sant'Anna e San Sebastiano, Caserta, Italy
| | - Laura Sciacca
- Dipartimento Medicina Clinica e Sperimentale, Università degli Studi di Catania, Catania, Italy
| | - Maria Angela Sculli
- UOC Diabetologia e Endocrinologia, GOM Bianchi-Melacrino-Morelli, Reggio Calabria, Italy
| | - Elena Succurro
- DPT Scienze Mediche Chirurgiche, Università Magna Grecia, Catanzaro, Italy
| | - Elisabetta Torlone
- AOS Maria della Misericordia SC Endocrinologia e Metabolismo, Università di Perugia, Perugia, Italy
| | - Ester Vitacolonna
- Dipartimento di Medicina e Scienza dell'Invecchiamento, Università di Chieti, Chieti, Italy
| |
Collapse
|
13
|
Denys ME, Kozlova EV, Liu R, Bishay AE, Do EA, Piamthai V, Korde YV, Luna CN, Lam AA, Hsiao A, Currás-Collazo M. Maternal probiotic supplementation protects against PBDE-induced developmental, behavior and metabolic reprogramming in a sexually dimorphic manner: Role of gut microbiome. Arch Toxicol 2025; 99:423-446. [PMID: 39520540 PMCID: PMC11748483 DOI: 10.1007/s00204-024-03882-4] [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: 08/29/2024] [Accepted: 09/23/2024] [Indexed: 11/16/2024]
Abstract
Polybrominated diphenyl ethers (PBDEs) are endocrine-disrupting persistent organic pollutants (POPs) used as flame retardants in a wide range of commercial applications. We have previously reported neurobehavioral and metabolic reprogramming produced by developmental PBDEs. PBDEs perturb the microbiome, an influencer of life-long health, while probiotic supplementation with Limosilactobacillus reuteri (LR) can avert neurobehavioral and endocrine disruption. We, therefore, tested the hypothesis that perinatal maternal LR supplementation would protect gut microbiome richness and diversity, developmental milestones, adult neurobehavior and metabolic homeostasis in PBDE-exposed offspring. C57BL/6N dams were orally exposed to a commercial penta-mixture of PBDEs, DE-71, at 0.1 mg/kg/day, or corn oil vehicle (VEH/CON) during gestation and lactation. Mice offspring received DE-71 or VEH/CON with or without co-administration of LR (ATCC-PTA-6475) indirectly via their mother from gestational day (GD) 0 until postnatal day (P)21 (Cohort 1), or continued to receive LR directly from P22 through adulthood (Cohort 2). Results of fecal 16S rRNA sequencing indicated age- and sex-dependent effects of DE-71 on gut microbial communities. Maternal LR treatment protected against DE-71-induced reduction in α-diversity in P22 females and against β-diversity alterations in P30 males. In females, DE-71 changed the relative abundance of specific bacterial taxa, such as Tenericutes and Cyanobacteria (elevated) and Deferribacterota (reduced). In males, several Firmicutes taxa were elevated, while Proteobacteria, Chlamydiae, and several Bacteroidota taxa were reduced. The number of disrupted taxa normalized by maternal LR supplementation was as follows: 100% in P22 females and 33% in males at P22 and 25% at P30. Maternal LR treatment protected against DE-71-induced delay of postnatal body weight gain in males and ameliorated the abnormal timing of incisor eruption in both sexes. Further, DE-71 produced exaggerated digging in both sexes as well as locomotor hyperactivity in females, effects that were mitigated by maternal LR only in females. Other benefits of LR therapy included normalization of glucose tolerance, insulin-to-glucose ratio and plasma leptin in adult DE-71 females (Cohort 2). This study provides evidence that probiotic supplementation can mitigate POP-induced reprogramming of neurodevelopment, adult neurobehavior, and glucose metabolism in association with modified gut microbial community structure in a sex-dependent manner.
Collapse
Affiliation(s)
- Maximillian E Denys
- Department of Molecular Cell and Systems Biology, University of California, Riverside, CA, 92521, USA
| | - Elena V Kozlova
- Department of Molecular Cell and Systems Biology, University of California, Riverside, CA, 92521, USA
- Neuroscience Graduate Program, University of California, Riverside, CA, USA
| | - Rui Liu
- Department of Microbiology and Plant Pathology, University of California, Riverside, CA, USA
| | - Anthony E Bishay
- Department of Molecular Cell and Systems Biology, University of California, Riverside, CA, 92521, USA
| | - Elyza A Do
- Department of Microbiology and Plant Pathology, University of California, Riverside, CA, USA
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Riverside, CA, USA
| | - Varadh Piamthai
- Department of Microbiology and Plant Pathology, University of California, Riverside, CA, USA
| | - Yash V Korde
- Department of Molecular Cell and Systems Biology, University of California, Riverside, CA, 92521, USA
| | - Crystal N Luna
- Department of Molecular Cell and Systems Biology, University of California, Riverside, CA, 92521, USA
| | - Artha A Lam
- Department of Molecular Cell and Systems Biology, University of California, Riverside, CA, 92521, USA
| | - Ansel Hsiao
- Department of Microbiology and Plant Pathology, University of California, Riverside, CA, USA
| | - Margarita Currás-Collazo
- Department of Molecular Cell and Systems Biology, University of California, Riverside, CA, 92521, USA.
| |
Collapse
|
14
|
López-Yerena A, de Santisteban Villaplana V, Badimon L, Vilahur G, Padro T. Probiotics: A Potential Strategy for Preventing and Managing Cardiovascular Disease. Nutrients 2024; 17:52. [PMID: 39796486 PMCID: PMC11722674 DOI: 10.3390/nu17010052] [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: 12/05/2024] [Revised: 12/22/2024] [Accepted: 12/24/2024] [Indexed: 01/13/2025] Open
Abstract
Probiotics are gaining recognition as a viable strategy for mitigating cardiovascular risk factors. Specifically, recent studies highlight their potential benefits in managing cholesterol levels, blood pressure, and inflammation, which are critical components in the prevention of cardiovascular diseases (CVD). This comprehensive review aims to elucidate the impact of probiotic consumption on major cardiovascular risk factors, including individuals with hypertension, type II diabetes mellitus, metabolic syndrome, hypercholesterolemia, and in secondary prevention in coronary artery disease. Scientific evidence based on human studies suggests that probiotic consumption is associated with positive effects on anthropometric measures, inflammation markers, blood pressure, glucose metabolism markers, lipid profiles, and endothelial function. However, these findings should be interpreted pragmatically and acknowledge the significant variability in results. This variability may be attributed to factors such as probiotic composition (single strain or multiple strains), the characteristics of the delivery matrix (food, capsules, and sachets), the duration of the intervention, the dosage regimen, and baseline health profiles of the participants. Incorporating probiotics as part of a comprehensive and healthy lifestyle approach can be considered a feasible strategy for both the prevention and management of CVD. However, further research is needed on factors influencing the effect of probiotics, such as: (i) optimal probiotic strain(s), (ii) appropriate dosage, (iii) duration of treatment, (iv) optimal delivery vehicle, and (v) sex-specific differences.
Collapse
Affiliation(s)
- Anallely López-Yerena
- Institut Recerca Sant Pau, Sant Quinti 77-79, 08041 Barcelona, Spain; (A.L.-Y.); (V.d.S.V.); (L.B.); (G.V.)
| | - Victoria de Santisteban Villaplana
- Institut Recerca Sant Pau, Sant Quinti 77-79, 08041 Barcelona, Spain; (A.L.-Y.); (V.d.S.V.); (L.B.); (G.V.)
- School of Pharmacy and Food Sciences, University of Barcelona (UB), 08036 Barcelona, Spain
| | - Lina Badimon
- Institut Recerca Sant Pau, Sant Quinti 77-79, 08041 Barcelona, Spain; (A.L.-Y.); (V.d.S.V.); (L.B.); (G.V.)
- Centro de Investigación Biomédica en Red Cardiovascular (CIBER-CV), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Cardiovascular Research Foundation for Health Prevention and Innovation (FICSI), 08017 Barcelona, Spain
| | - Gemma Vilahur
- Institut Recerca Sant Pau, Sant Quinti 77-79, 08041 Barcelona, Spain; (A.L.-Y.); (V.d.S.V.); (L.B.); (G.V.)
- Centro de Investigación Biomédica en Red Cardiovascular (CIBER-CV), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Teresa Padro
- Institut Recerca Sant Pau, Sant Quinti 77-79, 08041 Barcelona, Spain; (A.L.-Y.); (V.d.S.V.); (L.B.); (G.V.)
- Centro de Investigación Biomédica en Red Cardiovascular (CIBER-CV), Instituto de Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
15
|
Zolghadrpour MA, Jowshan MR, Heidari Seyedmahalleh M, Karimpour F, Imani H, Asghari S. The effect of a new developed synbiotic yogurt consumption on metabolic syndrome components in adults with metabolic syndrome: a randomized controlled clinical trial. Nutr Diabetes 2024; 14:97. [PMID: 39695092 PMCID: PMC11655839 DOI: 10.1038/s41387-024-00354-7] [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: 03/19/2024] [Revised: 10/26/2024] [Accepted: 12/11/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND Studies have proposed that probiotic intake may ameliorate some of the clinical components of metabolic syndrome (MetS). This study aimed to determine the effects of a new developed synbiotic yogurt containing Lactobacillus plantarum, Lactobacillus pentosus, and Chloromyces marcosianos yeast on the components of MetS in adults with MetS. METHODS In this randomized, placebo-controlled, clinical trial, 44 participants were divided into two groups to receive 300 grams of synbiotic yogurt or regular yogurt daily for 12 weeks. Anthropometric measurements, blood pressure, and biochemical parameters evaluated before and after the intervention. RESULTS Daily consumption of synbiotic yogurt containing L. plantarum, L. pentosus, and C. marcosianos yeast in adults with MetS caused a significant decrease in the levels of fasting blood glucose (FBG) (p = 0.005), fasting insulin (p = 0.001), homeostatic model assessment for insulin resistance (HOMA-IR) index (p < 0.001), waist to hip ratio (WHR) (p = 0.02) and systolic blood pressure (p = 0.008) in the intervention group compared to the control group. CONCLUSIONS According to the findings of this study, daily consumption of the synbiotic yogurt was associated with improvements in insulin resistance, systolic blood pressure and WHR, which could be beneficial in patients with MetS.
Collapse
Affiliation(s)
- Mohammad-Amin Zolghadrpour
- Department of Clinical Nutrition, Faculty of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad-Reza Jowshan
- Department of Clinical Nutrition, Faculty of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Heidari Seyedmahalleh
- Department of Community Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran
| | - Farzad Karimpour
- Social Determinants of Health Research Center, Yasuj University of Medical Science, Yasuj, Iran
| | - Hossein Imani
- Department of Clinical Nutrition, Faculty of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran
| | - Somayyeh Asghari
- Department of Clinical Nutrition, Faculty of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
16
|
Al-Habsi N, Al-Khalili M, Haque SA, Elias M, Olqi NA, Al Uraimi T. Health Benefits of Prebiotics, Probiotics, Synbiotics, and Postbiotics. Nutrients 2024; 16:3955. [PMID: 39599742 PMCID: PMC11597603 DOI: 10.3390/nu16223955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 11/13/2024] [Accepted: 11/14/2024] [Indexed: 11/29/2024] Open
Abstract
The trillions of microbes that constitute the human gut microbiome play a crucial role in digestive health, immune response regulation, and psychological wellness. Maintaining gut microbiota is essential as metabolic diseases are associated with it. Functional food ingredients potentially improving gut health include prebiotics, probiotics, synbiotics, and postbiotics (PPSPs). While probiotics are living bacteria that provide health advantages when ingested sufficiently, prebiotics are non-digestible carbohydrates that support good gut bacteria. Synbiotics work together to improve immunity and intestinal health by combining probiotics and prebiotics. Postbiotics have also demonstrated numerous health advantages, such as bioactive molecules created during probiotic fermentation. According to a recent study, PPSPs can regulate the synthesis of metabolites, improve the integrity of the intestinal barrier, and change the gut microbiota composition to control metabolic illnesses. Additionally, the use of fecal microbiota transplantation (FMT) highlights the potential for restoring gut health through microbiota modulation, reinforcing the benefits of PPSPs in enhancing overall well-being. Research has shown that PPSPs provide several health benefits, such as improved immunological function, alleviation of symptoms associated with irritable bowel disease (IBD), decreased severity of allergies, and antibacterial and anti-inflammatory effects. Despite encouraging results, many unanswered questions remain about the scope of PPSPs' health advantages. Extensive research is required to fully realize the potential of these functional food components in enhancing human health and well-being. Effective therapeutic and prophylactic measures require further investigation into the roles of PPSPs, specifically their immune-system-modulating, cholesterol-lowering, antioxidant, and anti-inflammatory characteristics.
Collapse
Affiliation(s)
- Nasser Al-Habsi
- Department of Food Science and Nutrition, College of Agricultural and Marine Sciences, Sultan Qaboos University, Al-Khodh 123, Muscat P.O. Box 34, Oman; (M.A.-K.); (M.E.); (N.A.O.); (T.A.U.)
| | - Maha Al-Khalili
- Department of Food Science and Nutrition, College of Agricultural and Marine Sciences, Sultan Qaboos University, Al-Khodh 123, Muscat P.O. Box 34, Oman; (M.A.-K.); (M.E.); (N.A.O.); (T.A.U.)
| | - Syed Ariful Haque
- Department of Marine Science and Fisheries, College of Agricultural and Marine Sciences, Sultan Qaboos University, Al-Khodh 123, Muscat P.O. Box 34, Oman
- Department of Fisheries, Bangamata Sheikh Fojilatunnesa Mujib Science and Technology University, Melandah, Jamalpur 2012, Bangladesh
| | - Moussa Elias
- Department of Food Science and Nutrition, College of Agricultural and Marine Sciences, Sultan Qaboos University, Al-Khodh 123, Muscat P.O. Box 34, Oman; (M.A.-K.); (M.E.); (N.A.O.); (T.A.U.)
| | - Nada Al Olqi
- Department of Food Science and Nutrition, College of Agricultural and Marine Sciences, Sultan Qaboos University, Al-Khodh 123, Muscat P.O. Box 34, Oman; (M.A.-K.); (M.E.); (N.A.O.); (T.A.U.)
| | - Tasnim Al Uraimi
- Department of Food Science and Nutrition, College of Agricultural and Marine Sciences, Sultan Qaboos University, Al-Khodh 123, Muscat P.O. Box 34, Oman; (M.A.-K.); (M.E.); (N.A.O.); (T.A.U.)
| |
Collapse
|
17
|
Ganjayi MS, Sankaran KR, Meriga B, Bhatia R, Sharma S, Kondepudi KK. Astragalin and rutin restore gut microbiota dysbiosis, alleviate obesity and insulin resistance in high-fat diet-fed C57BL/6J mice. FOOD SCIENCE AND HUMAN WELLNESS 2024; 13:3256-3265. [DOI: 10.26599/fshw.2023.9250012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2025]
|
18
|
Wang H, Guo Y, Han W, Liang M, Xiao X, Jiang X, Yu W. Tauroursodeoxycholic Acid Improves Nonalcoholic Fatty Liver Disease by Regulating Gut Microbiota and Bile Acid Metabolism. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:20194-20210. [PMID: 39193771 DOI: 10.1021/acs.jafc.4c04630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/29/2024]
Abstract
Tauroursodeoxycholic acid (TUDCA) is a synthetic bile salt that has demonstrated efficacy in the management of hepatobiliary disorders. However, its specific mechanism of action in preventing and treating nonalcoholic fatty liver disease (NAFLD) remains incompletely understood. This research revealed that TUDCA treatment can reduce obesity and hepatic lipid buildup, enhance intestinal barrier function and microbial balance, and increase the presence of Allobaculum and Bifidobacterium in NAFLD mouse models. TUDCA can influence the activity of farnesoid X receptor (FXR) and cholesterol 7α-hydroxylase (CYP7A1), resulting in higher hepatic bile acid levels and increased expression of sodium taurocholate cotransporting polypeptide (NTCP), leading to elevated concentrations of liver-bound bile acids in mice. Furthermore, TUDCA can inhibit the expression of FXR and fatty acid transport protein 5 (FATP5), thereby reducing fatty acid absorption and hepatic lipid accumulation. This investigation provides new insights into the potential of TUDCA for preventing and treating NAFLD.
Collapse
Affiliation(s)
- Huan Wang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Yi Guo
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Weiting Han
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Meng Liang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Xiao Xiao
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Xiaowen Jiang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Wenhui Yu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
- Institute of Chinese Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
- Key Laboratory for Prevention and Treatment of Common Animal Diseases in Heilongjiang Province General Universities, Harbin 150030, China
| |
Collapse
|
19
|
Forte N, Marfella B, Nicois A, Palomba L, Paris D, Motta A, Pina Mollica M, Di Marzo V, Cristino L. The short-chain fatty acid acetate modulates orexin/hypocretin neurons: A novel mechanism in gut-brain axis regulation of energy homeostasis and feeding. Biochem Pharmacol 2024; 226:116383. [PMID: 38908530 DOI: 10.1016/j.bcp.2024.116383] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 06/19/2024] [Accepted: 06/20/2024] [Indexed: 06/24/2024]
Abstract
The short-chain fatty acids (SCFAs) acetate, propionate and butyrate, the major products of intestinal microbial fermentation of dietary fibres, are involved in fine-tuning brain functions via the gut-brain axis. However, the effects of SCFAs in the hypothalamic neuronal network regulating several autonomic-brain functions are still unknown. Using NMR spectroscopy, we detected a reduction in brain acetate concentrations in the hypothalamus of obese leptin knockout ob/ob mice compared to lean wild-type littermates. Therefore, we investigated the effect of acetate on orexin/hypocretin neurons (hereafter referred as OX or OX-A neurons), a subset of hypothalamic neurons regulating energy homeostasis, which we have characterized in previous studies to be over-activated by the lack of leptin and enhancement of endocannabinoid tone in the hypothalamus of ob/ob mice. We found that acetate reduces food-intake in concomitance with a reduction of orexin neuronal activity in ob/ob mice. This was demonstrated by evaluating food-intake behaviour and orexin-A/c-FOS immunoreactivity coupled with patch-clamp recordings in Hcrt-eGFP neurons, quantification of prepro-orexin mRNA, and immunolabeling of GPR-43, the main acetate receptor. Our data provide new insights into the mechanisms of the effects of chronic dietary supplementation with acetate, or complex carbohydrates, on energy intake and body weight, which may be partly mediated by inhibition of orexinergic neuron activity.
Collapse
Affiliation(s)
- Nicola Forte
- Institute of Biomolecular Chemistry, National Research Council of Italy, Pozzuoli, Naples, Italy
| | - Brenda Marfella
- Institute of Biomolecular Chemistry, National Research Council of Italy, Pozzuoli, Naples, Italy; Department of Biology, University of Naples Federico II, Naples, Italy
| | - Alessandro Nicois
- Institute of Biomolecular Chemistry, National Research Council of Italy, Pozzuoli, Naples, Italy; Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Letizia Palomba
- Institute of Biomolecular Chemistry, National Research Council of Italy, Pozzuoli, Naples, Italy; Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Debora Paris
- Institute of Biomolecular Chemistry, National Research Council of Italy, Pozzuoli, Naples, Italy
| | - Andrea Motta
- Institute of Biomolecular Chemistry, National Research Council of Italy, Pozzuoli, Naples, Italy
| | - Maria Pina Mollica
- Department of Biology, University of Naples Federico II, Naples, Italy; Centro Servizi Metrologici e Tecnologici Avanzati (CeSMA), Complesso Universitario di Monte Sant'Angelo, 80126 Naples, Italy; Task Force on Microbiome Studies, University of Naples Federico II, 80138 Naples, Italy
| | - Vincenzo Di Marzo
- Canada Excellence Research Chair on the Microbiome-Endocannabinoidome Axis in Metabolic Health, Faculty of Medicine and Faculty of Agricultural and Food Sciences, Université Laval, Québec City, QC, Canada.
| | - Luigia Cristino
- Institute of Biomolecular Chemistry, National Research Council of Italy, Pozzuoli, Naples, Italy.
| |
Collapse
|
20
|
Li S, Liu Z, Zhang Q, Su D, Wang P, Li Y, Shi W, Zhang Q. The Antidiabetic Potential of Probiotics: A Review. Nutrients 2024; 16:2494. [PMID: 39125375 PMCID: PMC11313988 DOI: 10.3390/nu16152494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 07/26/2024] [Accepted: 07/26/2024] [Indexed: 08/12/2024] Open
Abstract
Diabetes has become one of the most prevalent global epidemics, significantly impacting both the economy and the health of individuals. Diabetes is associated with numerous complications, such as obesity; hyperglycemia; hypercholesterolemia; dyslipidemia; metabolic endotoxemia; intestinal barrier damage; insulin-secretion defects; increased oxidative stress; and low-grade, systemic, and chronic inflammation. Diabetes cannot be completely cured; therefore, current research has focused on developing various methods to control diabetes. A promising strategy is the use of probiotics for diabetes intervention. Probiotics are a class of live, non-toxic microorganisms that can colonize the human intestine and help improve the balance of intestinal microbiota. In this review, we summarize the current clinical studies on using probiotics to control diabetes in humans, along with mechanistic studies conducted in animal models. The primary mechanism by which probiotics regulate diabetes is improved intestinal barrier integrity, alleviated oxidative stress, enhanced immune response, increased short-chain fatty acid production, etc. Therefore, probiotic supplementation holds great potential for the prevention and management of diabetes.
Collapse
Affiliation(s)
- Shiming Li
- Department of Nutrition and Health, China Agricultural University, Beijing 100193, China; (S.L.); (Z.L.); (Q.Z.); (P.W.); (Y.L.)
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100193, China
| | - Zichao Liu
- Department of Nutrition and Health, China Agricultural University, Beijing 100193, China; (S.L.); (Z.L.); (Q.Z.); (P.W.); (Y.L.)
| | - Qi Zhang
- Department of Nutrition and Health, China Agricultural University, Beijing 100193, China; (S.L.); (Z.L.); (Q.Z.); (P.W.); (Y.L.)
| | - Dan Su
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY 14850, USA;
| | - Pengjie Wang
- Department of Nutrition and Health, China Agricultural University, Beijing 100193, China; (S.L.); (Z.L.); (Q.Z.); (P.W.); (Y.L.)
| | - Yixuan Li
- Department of Nutrition and Health, China Agricultural University, Beijing 100193, China; (S.L.); (Z.L.); (Q.Z.); (P.W.); (Y.L.)
| | - Wenbiao Shi
- Department of Nutrition and Health, China Agricultural University, Beijing 100193, China; (S.L.); (Z.L.); (Q.Z.); (P.W.); (Y.L.)
| | - Qian Zhang
- Department of Nutrition and Health, China Agricultural University, Beijing 100193, China; (S.L.); (Z.L.); (Q.Z.); (P.W.); (Y.L.)
| |
Collapse
|
21
|
Vinothkanna A, Shi‐Liang X, Karthick Rajan D, Prathiviraj R, Sekar S, Zhang S, Wang B, Liu Z, Jia A. Feasible mechanisms and therapeutic potential of food probiotics to mitigate diabetes‐associated cancers: A comprehensive review and in silico validation. FOOD FRONTIERS 2024; 5:1476-1511. [DOI: 10.1002/fft2.406] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2025] Open
Abstract
AbstractPeople with diabetes mellitus (DM) and hyperglycemia are linked with cancer risk. Diabetes and cancer have been corroborated by high morbidity and mortality rates. Studies revealed that elevated levels of insulin secretions trigger insulin‐like growth factor 1 (IGF‐1) production. Moreover, IGF‐1 is a key regulator involved in promoting cancer cell progression and is linked with DM. Cancer drug resistance and ototoxic effects can adversely affect the health and lifespan of an individual. However, naturally derived bioactive compounds are gaining attention for their nontoxic properties and specific behavior. Likewise, probiotics have also been regarded as safe and successful alternatives to treat DM‐linked cancers. The present review aims to highlight the therapeutic potential and feasible functions of probiotics to mitigate or inhibit DM‐associated cancers. Meanwhile, the intracellular signaling cascades involved in promoting DM‐linked cancer are enumerated for future prospective research. However, metabolomics interactions and protein–protein interactions are to be discussed for deeper insights into affirmative principles in diabetic‐linked cancers. Drug discovery and innovative preclinical evaluation need further adjuvant and immune‐enhancement therapies. Furthermore, the results of the in silico assessment could provide scientific excellence of IGF‐1 in diabetes and cancer. Overall, this review summarizes the mechanistic insights and therapeutic targets for diabetes‐associated cancer.
Collapse
Affiliation(s)
- Annadurai Vinothkanna
- Hainan Affiliated Hospital of Hainan Medical University Hainan General Hospital Haikou China
- School of Life and Health Sciences Hainan University Haikou China
| | - Xiang Shi‐Liang
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences Hainan University Haikou China
| | - Durairaj Karthick Rajan
- Department of Cell Biology, School of Life Sciences Central South University Changsha Hunan China
| | | | - Soundarapandian Sekar
- Department of Biotechnology Bharathidasan University Tiruchirappalli Tamil Nadu India
| | - Shubing Zhang
- Department of Cell Biology, School of Life Sciences Central South University Changsha Hunan China
| | - Bo Wang
- Hainan Affiliated Hospital of Hainan Medical University Hainan General Hospital Haikou China
| | - Zhu Liu
- School of Life and Health Sciences Hainan University Haikou China
| | - Ai‐Qun Jia
- Hainan Affiliated Hospital of Hainan Medical University Hainan General Hospital Haikou China
| |
Collapse
|
22
|
Chandrasekaran P, Weiskirchen S, Weiskirchen R. Effects of Probiotics on Gut Microbiota: An Overview. Int J Mol Sci 2024; 25:6022. [PMID: 38892208 PMCID: PMC11172883 DOI: 10.3390/ijms25116022] [Citation(s) in RCA: 44] [Impact Index Per Article: 44.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 05/24/2024] [Accepted: 05/29/2024] [Indexed: 06/21/2024] Open
Abstract
The role of probiotics in regulating intestinal flora to enhance host immunity has recently received widespread attention. Altering the human gut microbiota may increase the predisposition to several disease phenotypes such as gut inflammation and metabolic disorders. The intestinal microbiota converts dietary nutrients into metabolites that serve as biologically active molecules in modulating regulatory functions in the host. Probiotics, which are active microorganisms, play a versatile role in restoring the composition of the gut microbiota, helping to improve host immunity and prevent intestinal disease phenotypes. This comprehensive review provides firsthand information on the gut microbiota and their influence on human health, the dietary effects of diet on the gut microbiota, and how probiotics alter the composition and function of the human gut microbiota, along with their corresponding effects on host immunity in building a healthy intestine. We also discuss the implications of probiotics in some of the most important human diseases. In summary, probiotics play a significant role in regulating the gut microbiota, boosting overall immunity, increasing the abundance of beneficial bacteria, and helping ameliorate the symptoms of multiple diseases.
Collapse
Affiliation(s)
- Preethi Chandrasekaran
- UT Southwestern Medical Center Dallas, 5323 Harry Hines Blvd. ND10.504, Dallas, TX 75390-9014, USA
| | - Sabine Weiskirchen
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry (IFMPEGKC), Rheinisch-Westfälische Technische Hochschule (RWTH) University Hospital Aachen, D-52074 Aachen, Germany;
| | - Ralf Weiskirchen
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry (IFMPEGKC), Rheinisch-Westfälische Technische Hochschule (RWTH) University Hospital Aachen, D-52074 Aachen, Germany;
| |
Collapse
|
23
|
Huang J, Wang X, Li Q, Zhang P, Jing Z, Zhang J, Su H, Sun X. Effect of Mixed Probiotics on Ovalbumin-Induced Atopic Dermatitis in Juvenile Mice. Int J Microbiol 2024; 2024:7172386. [PMID: 38590774 PMCID: PMC10999295 DOI: 10.1155/2024/7172386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 03/13/2024] [Accepted: 03/18/2024] [Indexed: 04/10/2024] Open
Abstract
Atopic dermatitis is one of the most common dermatologic problems, especially in children. Given the ability of symbiotic microorganisms in modulating the immune system, probiotics administration has been studied in previous research in the management of atopic dermatitis. However, there are conflicting results between studies. In this study, we aimed to assess the effectiveness of mixed probiotics as a treatment option for atopic dermatitis induced by ovalbumin. BALB/c juvenile mice were classified and divided into the ovalbumin group, mixed probiotic group (ovalbumin + LK), and control group. Except for the control group, all mice were sensitized with ovalbumin to establish a model of atopic dermatitis. The mixed probiotics were given by gavage for 14 days. Mice body weight, skin lesions, skin inflammation, ovalbumin-specific Ig, the number of Treg and CD103+DC, and the expression level of PD-1/PD-L1 were examined. The results showed that mixed probiotics can improve body weight and alleviate skin symptoms. Mixed probiotics reduced serum Th2 inflammatory factors, eosinophils, mast cell degranulation, mast cell count, and the expression of ovalbumin-specific immunoglobulin E/G1 and increased the anti-inflammatory cytokine interleukin-10, Treg cells, CD103+DC cells, and the expression level of PD-1/PD-L1. These findings suggest that mixed probiotics could be a viable treatment option for atopic dermatitis and provide insight into the underlying mechanisms involved.
Collapse
Affiliation(s)
- Jinli Huang
- Department of Pediatrics, Xijing Hospital, The Fourth Military Medical University, Xi'an 710032, China
| | - Xingzhi Wang
- Department of Pediatrics, Xijing Hospital, The Fourth Military Medical University, Xi'an 710032, China
| | - Qiuhong Li
- Department of Pediatrics, Xijing Hospital, The Fourth Military Medical University, Xi'an 710032, China
| | - Panpan Zhang
- Department of Pediatrics, Xijing Hospital, The Fourth Military Medical University, Xi'an 710032, China
| | - Zenghui Jing
- Department of Pediatrics, Xijing Hospital, The Fourth Military Medical University, Xi'an 710032, China
| | - Juan Zhang
- Department of Pediatrics, Xijing Hospital, The Fourth Military Medical University, Xi'an 710032, China
| | - Hui Su
- Department of Geriatrics, Xijing Hospital, The Fourth Military Medical University, Xi'an 710032, China
| | - Xin Sun
- Department of Pediatrics, Xijing Hospital, The Fourth Military Medical University, Xi'an 710032, China
| |
Collapse
|
24
|
Bhatia Z, Kumar S, Seshadri S. Composition and interaction of maternal microbiota with immune mediators during pregnancy and their outcome: A narrative review. Life Sci 2024; 340:122440. [PMID: 38278350 DOI: 10.1016/j.lfs.2024.122440] [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: 10/18/2023] [Revised: 12/26/2023] [Accepted: 01/14/2024] [Indexed: 01/28/2024]
Abstract
The connection between maternal microbiota and infant health has been greatly garnered interest for therapeutic purposes. The early resident microbiota perpetually exhibits much more flexibility as compared to that of the adults, and therefore, constant need of understanding the infant as well as maternal microbiota and their implications however has increased. In this review, we focus mainly on the diversity of overall maternal microbiota including the gut, vaginal, colostrum microbiota and how inflammatory markers fluctuate throughout the normal pregnancy as well in pregnancy with complications. The maternal body undergoes a cascade of physiological changes including hormonal, immunological and metabolic events to support the fetal development. These changes at the time of pregnancy have been correlated with alteration in the composition and diversity of maternal microbiota. Along with alteration in microbiome, the levels of circulatory cytokines fluctuate by complex network of inflammation, in order to prevent the fetal allograft throughout the pregnancy. The dynamic relationship of gut microbiota with the host and its immune system allows one to have greater insights of their role in pregnancy and newborn's health. Emerging evidence suggests that the vertical transmission of bacterial community from mother to newborn may begin in-utero which contributes in developing the immune system and infant gut microbiota.
Collapse
Affiliation(s)
- Zeel Bhatia
- Institute of Science, Nirma University, Ahmedabad, Gujarat 382481, India
| | - Sunny Kumar
- Institute of Science, Nirma University, Ahmedabad, Gujarat 382481, India
| | - Sriram Seshadri
- Institute of Science, Nirma University, Ahmedabad, Gujarat 382481, India.
| |
Collapse
|
25
|
Barakat GM, Ramadan W, Assi G, Khoury NBE. Satiety: a gut-brain-relationship. J Physiol Sci 2024; 74:11. [PMID: 38368346 PMCID: PMC10874559 DOI: 10.1186/s12576-024-00904-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2023] [Accepted: 01/30/2024] [Indexed: 02/19/2024]
Abstract
Many hormones act on the hypothalamus to control hunger and satiety through various pathways closely associated with several factors. When food is present in the gastro intestinal (GI) tract, enteroendocrine cells (EECs) emit satiety signals such as cholecystokinin (CCK), glucagon like peptide-1 (GLP-1) and peptide YY (PYY), which can then communicate with the vagus nerve to control food intake. More specifically, satiety has been shown to be particularly affected by the GLP-1 hormone and its receptor agonists that have lately been acknowledged as a promising way to reduce weight. In addition, there is increasing evidence that normal flora is also involved in the peripheral, central, and reward system that impact satiety. Moreover, neurologic pathways control satiety through neurotransmitters. In this review, we discuss the different roles of each of the GLP-1 hormone and its agonist, gut microbiomes, as well as neurotransmitters and their interconnected relation in the regulation of body's satiety homeostasis.
Collapse
Affiliation(s)
- Ghinwa M Barakat
- Biological and Chemical Sciences Department, School of Arts and Sciences, Lebanese International University, Beirut, Lebanon.
| | - Wiam Ramadan
- Biological and Chemical Sciences Department, School of Arts and Sciences, Lebanese International University, Beirut, Lebanon
- Nutrition and Food Sciences Department, School of Arts and Sciences, Lebanese International University, Beirut, Lebanon
| | - Ghaith Assi
- Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, Beirut, Lebanon
| | - Noura B El Khoury
- Psychology department, Faculty of Arts and Sciences, University of Balamand, Balamand, Lebanon
| |
Collapse
|
26
|
Suastika AV, Widiana IGR, Fatmawati NND, Suastika K, Paulus IB, Sujaya IN. The role of probiotics and synbiotics on treatment of gestational diabetes: systematic review and meta-analysis. AJOG GLOBAL REPORTS 2024; 4:100285. [PMID: 38322777 PMCID: PMC10844859 DOI: 10.1016/j.xagr.2023.100285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2024] Open
Abstract
OBJECTIVE This review investigated the efficacy of probiotics and/or synbiotics in gestational diabetes mellitus treatment by targeting insulin resistance, lipid metabolism, and anti-inflammatory effects in an updated trial. DATA SOURCES The literature review was performed using the key words "Probiotics," "Synbiotics," and "Gestational Diabetes" in several databases, including PubMed, ScienceDirect, and the Cochrane Central Register of Controlled Trials. STUDY ELIGIBILITY CRITERIA Eligible publication was screened independently by 2 reviewers. Studies included provided at least 1 of the following outcomes: (1) blood glucose marker, including fasting blood glucose level, fasting serum insulin level, and homeostasis model assessment insulin resistance; (2) blood lipid profiles, including triglycerides, low-density lipoprotein cholesterol, and high-density lipoprotein cholesterol; and (3) nitric oxide and C-reactive protein. METHODS All studies were reviewed using the critical appraisal Cochrane risk-of-bias tool for randomized trials. The descriptions of the extracted data were guided by the Preferred Reporting Items for Systematic Reviews 2020 statement with the Grading of Recommendations Assessment, Development, and Evaluation approach. This study was registered on the International Prospective Register of Systematic Reviews database (identification number: CRD42022375665). RESULTS From 13 randomized controlled trials involving 896 patients, individuals with probiotic had significant reduction on homeostasis model assessment insulin resistance (mean difference, -0.72; 95% confidence interval, -1.07 to -0.38; I2, 96%; P=.00), fasting blood glucose level (mean difference, -3.79; 95% confidence interval, -6.24 to -1.34; I2, 93%; P=.00), and insulin level (mean difference, -2.43 mg/dL; 95% confidence interval, -3.37 to -1.48; I2, 54%; P=.00). Meanwhile for profile lipid, significant reduction of the mean difference was observed in the triglyceride (mean difference, -17.73 mg/dL; 95% confidence interval, -29.55 to - 5.9; P=.003) and C-reactive protein (mean difference, -1.93 dL; 95% confidence interval, -2.3 to -1.56; P=.00). CONCLUSION Probiotic and synbiotic supplementations reduced the risk of insulin resistance and improved glycemic control, blood lipid profiles, and inflammation in women with gestational diabetes mellitus. Probiotics may be a viable option for gestational diabetes mellitus treatment; however, large-scale, well-designed randomized controlled trials with longer follow-up periods are required before they can be recommended to patients.
Collapse
Affiliation(s)
| | - I Gde Raka Widiana
- Department of Internal Medicine, Udayana University, Bali, Indonesia (Drs I Widiana and K Suastika)
| | - Ni Nengah Dwi Fatmawati
- Faculty of Medicine, Department of Microbiology, Udayana University, Bali, Indonesia (Dr N Fatmawati)
| | - Ketut Suastika
- Department of Internal Medicine, Udayana University, Bali, Indonesia (Drs I Widiana and K Suastika)
| | | | - I Nengah Sujaya
- Faculty of Medicine, School of Public Health, Udayana University, Bali, Indonesia (N Sujaya PhD)
| |
Collapse
|
27
|
Rangel-Torres BE, García-Montoya IA, Rodríguez-Tadeo A, Jiménez-Vega F. The Symbiosis Between Lactobacillus acidophilus and Inulin: Metabolic Benefits in an Obese Murine Model. Probiotics Antimicrob Proteins 2024; 16:26-34. [PMID: 36443558 DOI: 10.1007/s12602-022-10012-y] [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] [Accepted: 11/16/2022] [Indexed: 11/30/2022]
Abstract
Obesity is defined as having an excess of adipose tissue and is associated with the development of diabetes, hypertension, and atherosclerosis, which are the main causes of death worldwide. Research shows that probiotics and prebiotics reduce the metabolic alterations caused by high-fat diets. Therefore, this work evaluated the effect of the incorporation of Lactobacillus acidophilus (probiotic) and inulin (prebiotic) in the diet through obesity markers (biochemical, anthropometric, and molecular markers) in an obese murine model. Four treatments were administered: (1) hypocaloric diet (HD), (2) HD + L. acidophilus, (3) HD + inulin, and (4) DH supplemented with L. acidophilus + inulin for 8 weeks. After treatment, glucose, triglycerides, total cholesterol, HDL-C, and LDL-C in plasma were determined. In addition, the total body weight and adipose tissue were taken to calculate the body mass index. Following RNA extraction from adipose tissue, the expression of PPAR gamma, PPAR alpha, and transforming growth factor beta 1 (TGF1β) was evaluated by semiquantitative PCR. All treatments showed an improvement in biochemical markers compared to the values of the obese model (p < 0.05). Optimal values for blood glucose (133.2 ± 14.3 mg/dL), triglycerides (71 ± 4.6 mg/dL), total cholesterol (48.9 ± 6 mg/dL), HDL-C (40.9 ± 4.8 mg/dL), and LDL-C (8.4 ± 1.7 mg/dL) were obtained in the mixed treatment. Regarding fat mass index (FMI), prebiotic treatment caused the greatest reduction. On the other hand, mixed treatment increased the gene expression of PPARα and TGF1β in adipose tissue with DH with L. acidophilus and inulin treatment. This work demonstrates that the use of L. acidophilus and inulin as a complementary treatment is a viable alternative for prevention and action as a complementary treatment for obesity given the reduction in biochemical parameters and anthropometric indices; these reductions were greater than those found in the classic treatment of obesity due to the induction of the expression of genes related to lipid metabolism and anti-inflammatory cytokines, which contribute to reducing the high levels of glucose, triglycerides, and cholesterol caused by obesity.
Collapse
Affiliation(s)
- Brian Eduardo Rangel-Torres
- Departamento Ciencias Químico-Biológicas, Instituto de Ciencias Biomédicas, Universidad Autónoma de Ciudad Juárez, Ciudad Juárez, México
| | - Isui Abril García-Montoya
- Departamento Ciencias Químico-Biológicas, Instituto de Ciencias Biomédicas, Universidad Autónoma de Ciudad Juárez, Ciudad Juárez, México
| | - Alejandra Rodríguez-Tadeo
- Departamento de Ciencias de La Salud, Instituto de Ciencias Biomédicas, Universidad Autónoma de Ciudad Juárez, Ciudad Juárez, México
| | - Florinda Jiménez-Vega
- Departamento Ciencias Químico-Biológicas, Instituto de Ciencias Biomédicas, Universidad Autónoma de Ciudad Juárez, Ciudad Juárez, México.
| |
Collapse
|
28
|
Hasaniani N, Ghasemi-Kasman M, Halaji M, Rostami-Mansoor S. Bifidobacterium breve Probiotic Compared to Lactobacillus casei Causes a Better Reduction in Demyelination and Oxidative Stress in Cuprizone-Induced Demyelination Model of Rat. Mol Neurobiol 2024; 61:498-509. [PMID: 37639065 DOI: 10.1007/s12035-023-03593-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 08/17/2023] [Indexed: 08/29/2023]
Abstract
Despite the anatomical separation, strong evidence suggested a bidirectional association between gut microbiota and central nervous system. Cross-talk between gut microbiota and brain has an important role in the pathophysiology of neurodegenerative disorders and regenerative processes. However, choosing the appropriate probiotics and combination therapy of probiotics to provide a synergistic effect is very crucial. In the present study, we investigated the effect of Lactobacillus casei (L. casei) and Bifidobacterium breve (B. breve) on alternation performance, oxidant/antioxidant biomarkers, the extent of demyelination, and the expression level of HO-1, Nrf-2, Olig2, MBP, PDGFRα, and BDNF in cuprizone (CPZ)-induced demyelination model of rat corpus callosum. In order to induce this model, rats received oral administration of CPZ 0.6% w/w in corn oil for 28 days. Then, L. casei, B. breve, or their combinations were orally administrated for 28 days. Y maze test was performed to investigate the alternation performance. Oxidant/antioxidant biomarkers were determined by colorimetric methods. Extent of demyelination was investigated using FluoroMyelin staining. The genes' expression levels of antioxidant and myelin lineage cells were assessed by quantitative real time PCR. The results showed the probiotics supplementation significantly improve the alternation performance and antioxidant capacity in demyelinated corpus callosum. Interestingly, B. breve supplementation alleviated demyelination and oxidative stress levels more than the administration of L. casei alone or the combination of two probiotics. These observations suggest that B. breve could provide a supplementary strategy for the treatment of multiple sclerosis by increasing antioxidant capacity and remyelination.
Collapse
Affiliation(s)
- Nima Hasaniani
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran
- Department of Clinical Biochemistry, Faculty of Medicine, Babol University of Medical Sciences, Babol, Iran
| | - Maryam Ghasemi-Kasman
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
- Department of Physiology, Faculty of Medicine, Babol University of Medical Sciences, Babol, Iran
| | - Mehrdad Halaji
- Infectious Diseases and Tropical Medicine Research Center, Babol University of Medical Sciences, Babol, Iran
| | - Sahar Rostami-Mansoor
- Department of Clinical Biochemistry, Faculty of Medicine, Babol University of Medical Sciences, Babol, Iran.
- Department of Laboratory Sciences, Faculty of Paramedical Sciences, Babol University of Medical Sciences, Babol, Iran.
| |
Collapse
|
29
|
Yu M, Yu B, Chen D. The effects of gut microbiota on appetite regulation and the underlying mechanisms. Gut Microbes 2024; 16:2414796. [PMID: 39501848 PMCID: PMC11542600 DOI: 10.1080/19490976.2024.2414796] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 09/23/2024] [Accepted: 09/30/2024] [Indexed: 11/09/2024] Open
Abstract
Appetite, a crucial aspect regulated by both the central nervous system and peripheral hormones, is influenced by the composition and dynamics of the intestinal microbiota, as evidenced by recent research. This review highlights the role of intestinal microbiota in appetite regulation, elucidating the involvement of various pathways. Notably, the metabolites generated by intestinal microorganisms, including short-chain fatty acids, bile acids, and amino acid derivatives, play a pivotal role in this intricate process. Furthermore, intestinal microorganisms contribute to appetite regulation by modulating nutritional perception, neural signal transmission, and hormone secretion within the digestive system. Consequently, manipulating and modulating the intestinal microbiota represent innovative strategies for ameliorating appetite-related disorders. This paper provides a comprehensive review of the effects of gut microbes and their metabolites on the central nervous system and host appetite. By exploring their potential regulatory pathways and mechanisms, this study aims to enhance our understanding of how gut microbes influence appetite regulation in the host.
Collapse
Affiliation(s)
- Miao Yu
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, Sichuan Province, China
- DadHank(Chengdu)Biotech Corp, Chengdu, Sichuan Province, China
| | - Bing Yu
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, Sichuan Province, China
| | - Daiwen Chen
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, Sichuan Province, China
| |
Collapse
|
30
|
de Brito Avelino L, Rodrigues KT, da Silva Cruz NT, Martins AA, de Aquino Martins ARL. Effectiveness of Probiotic Therapy in the Management of PeriodontalDisease in Diabetic Patients: A Scoping Review. Curr Diabetes Rev 2024; 20:e281123223961. [PMID: 38018184 DOI: 10.2174/0115733998271193231108054254] [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: 07/20/2023] [Revised: 09/21/2023] [Accepted: 09/27/2023] [Indexed: 11/30/2023]
Abstract
BACKGROUND Probiotics can compete with periodontal pathogens in the formation of dental biofilm, and they are able to modulate local and systemic immune responses. Thus, its use in diabetic patients with periodontal disease (PD) can overcome the limitations of conventional periodontal treatment. OBJECTIVE This scoping review aimed to understand the extent and type of evidence in relation to the effects of probiotic therapy on periodontal and glycaemic parameters of diabetic patients with PD. METHODS An electronic search was performed in the following databases: Cochrane Library, EMBASE, Virtual Health Library (including LILACS and BBO), PubMed (including Medline), Scopus, Web of Science, and Google Scholar. The review included clinical trials on patients with type 2 diabetes, diagnosed with gingivitis or periodontitis, who received probiotic therapy as a single therapy or adjuvant to scaling and root planning, and on whom the analyses of clinical periodontal, immunological, microbiological, or glycaemic parameters were performed. RESULTS The electronic search yielded a total of 1165 articles. After removing duplicate titles and performing systematic screening, 6 studies were included in the qualitative summary. Probiotic administration improved clinical periodontal parameters (bleeding on probing and probing depth), oxidative stress markers, and inflammatory cytokines (IL-8, IL-10, and TNF-α) in relation to control groups. Experimental groups were also more advantageous in reducing the frequency of periodontopathogenic bacteria. However, the evidence of probiotics in decreasing glycated hemoglobin is still uncertain. CONCLUSION Probiotics may provide safe additional benefits to periodontal parameters of patients with type 2 diabetes and periodontal disease.
Collapse
Affiliation(s)
| | | | | | - Agnes Andrade Martins
- Department of Dentistry, Federal University of Rio Grande do Norte, Natal, RN, Brazil
| | | |
Collapse
|
31
|
K VK, Bhat RG, Rao BK, R AP. The Gut Microbiota: a Novel Player in the Pathogenesis of Uterine Fibroids. Reprod Sci 2023; 30:3443-3455. [PMID: 37418220 PMCID: PMC10691976 DOI: 10.1007/s43032-023-01289-7] [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: 03/07/2023] [Accepted: 06/25/2023] [Indexed: 07/08/2023]
Abstract
Uterine fibroid is a common gynecological disorder that affects women of reproductive age and has emerged as a major public health concern. The symptoms have a negative influence on both their physical health and quality of life. The cost of treatment has a significant impact on the disease's burden. Even though its origin is uncertain, estrogen is thought to be a key player in fibroid pathophysiology. Many theories, including those based on genetic and environmental factors, explain what causes hyper-estrogenic condition in fibroid patients. One such possibility that is currently being explored is the hypothesis that an altered gut microbiome can contribute to the development of diseases characterized by estrogen dominance. Gut dysbiosis is often a "hot area" in the health sciences. According to a recent study, uterine fibroid patients have altered gut microbiome. A variety of risk factors influence both fibroid development and gut homeostasis. Diet, lifestyle, physical activity, and environmental contaminants have an impact on estrogen and the gut flora. A better understanding of uterine fibroids' pathophysiology is required to develop effective preventative and treatment options. A few ways by which the gut microbiota contributes to UF include estrogen, impaired immune function, inflammation, and altered gut metabolites. Therefore, in the future, while treating fibroid patients, various strategies to deal with changes in the gut flora may be advantageous. For developing suggestions for clinical diagnosis and therapy, we reviewed the literature on the relationship between uterine fibroids and the gut microbiota.
Collapse
Affiliation(s)
- Vineetha K K
- Department of Obstetrics and Gynecology, Melaka Manipal Medical College, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Rajeshwari G Bhat
- Department of Obstetrics and Gynecology, Melaka Manipal Medical College, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Bhamini Krishna Rao
- Department of Physiotherapy, Manipal College of Health Professions, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Archana P R
- Department of Basic Medical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India.
| |
Collapse
|
32
|
Tang J, Wei Y, Pi C, Zheng W, Zuo Y, Shi P, Chen J, Xiong L, Chen T, Liu H, Zhao Q, Yin S, Ren W, Cao P, Zeng N, Zhao L. The therapeutic value of bifidobacteria in cardiovascular disease. NPJ Biofilms Microbiomes 2023; 9:82. [PMID: 37903770 PMCID: PMC10616273 DOI: 10.1038/s41522-023-00448-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Accepted: 10/03/2023] [Indexed: 11/01/2023] Open
Abstract
There has been an increase in cardiovascular morbidity and mortality over the past few decades, making cardiovascular disease (CVD) the leading cause of death worldwide. However, the pathogenesis of CVD is multi-factorial, complex, and not fully understood. The gut microbiome has long been recognized to play a critical role in maintaining the physiological and metabolic health of the host. Recent scientific advances have provided evidence that alterations in the gut microbiome and its metabolites have a profound influence on the development and progression of CVD. Among the trillions of microorganisms in the gut, bifidobacteria, which, interestingly, were found through the literature to play a key role not only in regulating gut microbiota function and metabolism, but also in reducing classical risk factors for CVD (e.g., obesity, hyperlipidemia, diabetes) by suppressing oxidative stress, improving immunomodulation, and correcting lipid, glucose, and cholesterol metabolism. This review explores the direct and indirect effects of bifidobacteria on the development of CVD and highlights its potential therapeutic value in hypertension, atherosclerosis, myocardial infarction, and heart failure. By describing the key role of Bifidobacterium in the link between gut microbiology and CVD, we aim to provide a theoretical basis for improving the subsequent clinical applications of Bifidobacterium and for the development of Bifidobacterium nutritional products.
Collapse
Affiliation(s)
- Jia Tang
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy of Southwest Medical University, Luzhou, 646000, P.R. China
- Luzhou Key Laboratory of Traditional Chinese Medicine for Chronic Diseases Jointly Built by Sichuan and Chongqing, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, P.R. China
- Chengdu University of Traditional Chinese Medicine State Key Laboratory of Southwestern Chinese Medicine Resources, 1166 Liutai Avenue, Wenjiang District, Chengdu, Sichuan, 611137, P.R. China
- Key Laboratory of Medical Electrophysiology, Ministry of Education, Development Planning Department of Southwest Medical University, Luzhou, Sichuan, 646000, P.R. China
| | - Yumeng Wei
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy of Southwest Medical University, Luzhou, 646000, P.R. China
- Central Nervous System Drug Key Laboratory of Sichuan Province, School of Pharmacy of Southwest Medical University, Luzhou, Sichuan, 646000, P.R. China
| | - Chao Pi
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy of Southwest Medical University, Luzhou, 646000, P.R. China
- Central Nervous System Drug Key Laboratory of Sichuan Province, School of Pharmacy of Southwest Medical University, Luzhou, Sichuan, 646000, P.R. China
| | - Wenwu Zheng
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, P.R. China
| | - Ying Zuo
- Department of Comprehensive Medicine, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, P.R. China
| | - Peng Shi
- Key Laboratory of Medical Electrophysiology, Ministry of Education, Development Planning Department of Southwest Medical University, Luzhou, Sichuan, 646000, P.R. China
| | - Jinglin Chen
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy of Southwest Medical University, Luzhou, 646000, P.R. China
- Luzhou Key Laboratory of Traditional Chinese Medicine for Chronic Diseases Jointly Built by Sichuan and Chongqing, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, P.R. China
- Chengdu University of Traditional Chinese Medicine State Key Laboratory of Southwestern Chinese Medicine Resources, 1166 Liutai Avenue, Wenjiang District, Chengdu, Sichuan, 611137, P.R. China
- Key Laboratory of Medical Electrophysiology, Ministry of Education, Development Planning Department of Southwest Medical University, Luzhou, Sichuan, 646000, P.R. China
| | - Linjin Xiong
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy of Southwest Medical University, Luzhou, 646000, P.R. China
- Luzhou Key Laboratory of Traditional Chinese Medicine for Chronic Diseases Jointly Built by Sichuan and Chongqing, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, P.R. China
- Chengdu University of Traditional Chinese Medicine State Key Laboratory of Southwestern Chinese Medicine Resources, 1166 Liutai Avenue, Wenjiang District, Chengdu, Sichuan, 611137, P.R. China
- Key Laboratory of Medical Electrophysiology, Ministry of Education, Development Planning Department of Southwest Medical University, Luzhou, Sichuan, 646000, P.R. China
| | - Tao Chen
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy of Southwest Medical University, Luzhou, 646000, P.R. China
- Luzhou Key Laboratory of Traditional Chinese Medicine for Chronic Diseases Jointly Built by Sichuan and Chongqing, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, P.R. China
- Chengdu University of Traditional Chinese Medicine State Key Laboratory of Southwestern Chinese Medicine Resources, 1166 Liutai Avenue, Wenjiang District, Chengdu, Sichuan, 611137, P.R. China
- Key Laboratory of Medical Electrophysiology, Ministry of Education, Development Planning Department of Southwest Medical University, Luzhou, Sichuan, 646000, P.R. China
| | - Huiyang Liu
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy of Southwest Medical University, Luzhou, 646000, P.R. China
- Luzhou Key Laboratory of Traditional Chinese Medicine for Chronic Diseases Jointly Built by Sichuan and Chongqing, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, P.R. China
- Chengdu University of Traditional Chinese Medicine State Key Laboratory of Southwestern Chinese Medicine Resources, 1166 Liutai Avenue, Wenjiang District, Chengdu, Sichuan, 611137, P.R. China
- Key Laboratory of Medical Electrophysiology, Ministry of Education, Development Planning Department of Southwest Medical University, Luzhou, Sichuan, 646000, P.R. China
| | - Qianjiao Zhao
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy of Southwest Medical University, Luzhou, 646000, P.R. China
- Luzhou Key Laboratory of Traditional Chinese Medicine for Chronic Diseases Jointly Built by Sichuan and Chongqing, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, P.R. China
- Chengdu University of Traditional Chinese Medicine State Key Laboratory of Southwestern Chinese Medicine Resources, 1166 Liutai Avenue, Wenjiang District, Chengdu, Sichuan, 611137, P.R. China
- Key Laboratory of Medical Electrophysiology, Ministry of Education, Development Planning Department of Southwest Medical University, Luzhou, Sichuan, 646000, P.R. China
| | - Suyu Yin
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy of Southwest Medical University, Luzhou, 646000, P.R. China
- Luzhou Key Laboratory of Traditional Chinese Medicine for Chronic Diseases Jointly Built by Sichuan and Chongqing, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, P.R. China
- Chengdu University of Traditional Chinese Medicine State Key Laboratory of Southwestern Chinese Medicine Resources, 1166 Liutai Avenue, Wenjiang District, Chengdu, Sichuan, 611137, P.R. China
- Key Laboratory of Medical Electrophysiology, Ministry of Education, Development Planning Department of Southwest Medical University, Luzhou, Sichuan, 646000, P.R. China
| | - Wei Ren
- National Traditional Chinese Medicine Clinical Research Base and Drug Research Center of the Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, P.R. China
| | - Peng Cao
- The Affiliated Hospital of Traditional Chinese and Western Medicine Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210028, P.R. China.
| | - Nan Zeng
- Chengdu University of Traditional Chinese Medicine State Key Laboratory of Southwestern Chinese Medicine Resources, 1166 Liutai Avenue, Wenjiang District, Chengdu, Sichuan, 611137, P.R. China.
| | - Ling Zhao
- Luzhou Key Laboratory of Traditional Chinese Medicine for Chronic Diseases Jointly Built by Sichuan and Chongqing, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, P.R. China.
- Chengdu University of Traditional Chinese Medicine State Key Laboratory of Southwestern Chinese Medicine Resources, 1166 Liutai Avenue, Wenjiang District, Chengdu, Sichuan, 611137, P.R. China.
- Key Laboratory of Medical Electrophysiology, Ministry of Education, Development Planning Department of Southwest Medical University, Luzhou, Sichuan, 646000, P.R. China.
| |
Collapse
|
33
|
Moreira ALG, Silva GA, Silva PHF, Salvador SL, Vicente RM, Ferreira GC, Tanus-Santos JE, Mayer MPA, Ishikawa KH, de Souza SLS, Furlaneto FAC, Messora MR. Bifidobacterium animalis subspecies lactis HN019 can reduce the sequelae of experimental periodontitis in rats modulating intestinal parameters, expression of lipogenic genes, and levels of hepatic steatosis. J Periodontal Res 2023; 58:1006-1019. [PMID: 37482954 DOI: 10.1111/jre.13163] [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: 12/09/2022] [Revised: 05/15/2023] [Accepted: 07/07/2023] [Indexed: 07/25/2023]
Abstract
OBJECTIVE To determine whether Bifidobacterium animalis subspecies lactis HN019 (B. lactis HN019) can reduce the sequelae of experimental periodontitis (EP) in rats modulating systemic parameters. BACKGROUND This study evaluated the effects of probiotic therapy (PROB) in the prevention of local and systemic damage resulting from EP. METHODS Forty-eight rats were allocated into four groups: C (control), PROB, EP, and EP-PROB. PROB (1 × 1010 CFU/mL) administration lasted 8 weeks and PE was induced on the 7th week by placing ligature on the animals' lower first molars. All animals were euthanized in the 9th week of the experiment. Biomolecular analyses, RT-PCR, and histomorphometric analyses were performed. The data obtained were analyzed statistically (ANOVA, Tukey, p < .05). RESULTS The EP group had higher dyslipidemia when compared to the C group, as well as higher levels of insulin resistance, proteinuria levels, percentages of systolic blood pressure, percentage of fatty hepatocytes in the liver, and expression of adipokines was up-regulated (LEPR, NAMPT, and FABP4). All these parameters (except insulin resistance, systolic blood pressure, LEPR and FABP4 gene expression) were reduced in the EP-PROB group when compared to the EP group. The EP group had lower villus height and crypt depth, as well as a greater reduction in Bacteroidetes and a greater increase in Firmicutes when compared to the EP-PROB group. Greater alveolar bone loss was observed in the EP group when compared to the EP-PROB group. CONCLUSION Bifidobacterium lactis HN019 can reduce the sequelae of EP in rats modulating intestinal parameters, attenuating expression of lipogenic genes and hepatic steatosis.
Collapse
Affiliation(s)
- André L G Moreira
- Department of Oral and Maxillofacial Surgery and Periodontology, School of Dentistry of Ribeirão Preto, University of São Paulo - USP, Ribeirão Preto, São Paulo, Brazil
| | - Giselle A Silva
- Department of Oral and Maxillofacial Surgery and Periodontology, School of Dentistry of Ribeirão Preto, University of São Paulo - USP, Ribeirão Preto, São Paulo, Brazil
| | - Pedro H F Silva
- Department of Oral and Maxillofacial Surgery and Periodontology, School of Dentistry of Ribeirão Preto, University of São Paulo - USP, Ribeirão Preto, São Paulo, Brazil
| | - Sérgio L Salvador
- Department of Clinical Analyses, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo - USP, Ribeirão Preto, São Paulo, Brazil
| | - Raphael M Vicente
- Department of Oral and Maxillofacial Surgery and Periodontology, School of Dentistry of Ribeirão Preto, University of São Paulo - USP, Ribeirão Preto, São Paulo, Brazil
| | - Graziele C Ferreira
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo - USP, Ribeirão Preto, São Paulo, Brazil
| | - Jose E Tanus-Santos
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo - USP, Ribeirão Preto, São Paulo, Brazil
| | - Marcia P A Mayer
- Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Karin H Ishikawa
- Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Sérgio Luís Scombatti de Souza
- Department of Oral and Maxillofacial Surgery and Periodontology, School of Dentistry of Ribeirão Preto, University of São Paulo - USP, Ribeirão Preto, São Paulo, Brazil
| | - Flávia A C Furlaneto
- Department of Oral and Maxillofacial Surgery and Periodontology, School of Dentistry of Ribeirão Preto, University of São Paulo - USP, Ribeirão Preto, São Paulo, Brazil
| | - Michel R Messora
- Department of Oral and Maxillofacial Surgery and Periodontology, School of Dentistry of Ribeirão Preto, University of São Paulo - USP, Ribeirão Preto, São Paulo, Brazil
| |
Collapse
|
34
|
Song X, Liu Y, Zhang X, Weng P, Zhang R, Wu Z. Role of intestinal probiotics in the modulation of lipid metabolism: implications for therapeutic treatments. FOOD SCIENCE AND HUMAN WELLNESS 2023. [DOI: 10.1016/j.fshw.2023.02.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/28/2023]
|
35
|
Cuesta-Marti C, Uhlig F, Muguerza B, Hyland N, Clarke G, Schellekens H. Microbes, oxytocin and stress: Converging players regulating eating behavior. J Neuroendocrinol 2023; 35:e13243. [PMID: 36872624 DOI: 10.1111/jne.13243] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 01/26/2023] [Accepted: 02/02/2023] [Indexed: 02/17/2023]
Abstract
Oxytocin is a peptide-hormone extensively studied for its multifaceted biological functions and has recently gained attention for its role in eating behavior, through its action as an anorexigenic neuropeptide. Moreover, the gut microbiota is involved in oxytocinergic signaling through the brain-gut axis, specifically in the regulation of social behavior. The gut microbiota is also implicated in appetite regulation and is postulated to play a role in central regulation of hedonic eating. In this review, we provide an overview on oxytocin and its individual links with the microbiome, the homeostatic and non-homeostatic regulation of eating behavior as well as social behavior and stress.
Collapse
Affiliation(s)
- Cristina Cuesta-Marti
- Department of Anatomy & Neuroscience, University College Cork, Cork, Ireland
- APC Microbiome Ireland, Cork, Ireland
| | - Friederike Uhlig
- APC Microbiome Ireland, Cork, Ireland
- Department of Physiology, University College Cork, Ireland
| | - Begoña Muguerza
- Department of Anatomy & Neuroscience, University College Cork, Cork, Ireland
- APC Microbiome Ireland, Cork, Ireland
- Universitat Rovira i Virgili, Department of Biochemistry & Biotechnology, Nutrigenomics Research Group, Tarragona, Spain
| | - Niall Hyland
- APC Microbiome Ireland, Cork, Ireland
- Department of Physiology, University College Cork, Ireland
| | - Gerard Clarke
- APC Microbiome Ireland, Cork, Ireland
- Department of Psychiatry & Neurobehavioural Science, University College Cork, Cork, Ireland
| | - Harriët Schellekens
- Department of Anatomy & Neuroscience, University College Cork, Cork, Ireland
- APC Microbiome Ireland, Cork, Ireland
| |
Collapse
|
36
|
Kim JS, Williams KC, Kirkland RA, Schade R, Freeman KG, Cawthon CR, Rautmann AW, Smith JM, Edwards GL, Glenn TC, Holmes PV, de Lartigue G, de La Serre CB. The gut-brain axis mediates bacterial driven modulation of reward signaling. Mol Metab 2023; 75:101764. [PMID: 37380023 PMCID: PMC10372379 DOI: 10.1016/j.molmet.2023.101764] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 06/18/2023] [Accepted: 06/23/2023] [Indexed: 06/30/2023] Open
Abstract
OBJECTIVE Our goal is to investigate if microbiota composition modulates reward signaling and assess the role of the vagus in mediating microbiota to brain communication. METHODS Male germ-free Fisher rats were colonized with gastrointestinal contents from chow (low fat (LF) ConvLF) or HF (ConvHF) fed rats. RESULTS Following colonization, ConvHF rats consumed significantly more food than ConvLF animals. ConvHF rats displayed lower feeding-induced extracellular DOPAC levels (a metabolite of dopamine) in the Nucleus Accumbens (NAc) as well as reduced motivation for HF foods compared to ConvLF rats. Dopamine receptor 2 (DDR2) expression levels in the NAc were also significantly lower in ConvHF animals. Similar deficits were observed in conventionally raised HF fed rats, showing that diet-driven alteration in reward can be initiated via microbiota. Selective gut to brain deafferentation restored DOPAC levels, DRD2 expression, and motivational drive in ConvHF rats. CONCLUSIONS We concluded from these data that a HF-type microbiota is sufficient to alter appetitive feeding behavior and that bacteria to reward communication is mediated by the vagus nerve.
Collapse
Affiliation(s)
- Jiyoung S Kim
- Department of Nutritional Sciences, University of Georgia, USA
| | | | | | - Ruth Schade
- Department of Nutritional Sciences, University of Georgia, USA
| | | | | | | | | | - Gaylen L Edwards
- Department of Physiology and Pharmacology, University of Georgia, USA
| | - Travis C Glenn
- Department of Environmental Health Science, University of Georgia, USA
| | | | - Guillaume de Lartigue
- Monell Chemical Senses Center and Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, USA
| | | |
Collapse
|
37
|
Jain S, Marotta F, Haghshenas L, Yadav H. Treating Leaky Syndrome in the Over 65s: Progress and Challenges. Clin Interv Aging 2023; 18:1447-1451. [PMID: 37671072 PMCID: PMC10476862 DOI: 10.2147/cia.s409801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Accepted: 08/03/2023] [Indexed: 09/07/2023] Open
Abstract
As we age, our organ functions gradually decline. Circulating factors in the blood and the integrity of organ barriers can become dysfunctional, resulting in a condition known as leaky syndrome. This condition involves the unregulated exchange or leakage of components between organs. However, the triggers of leaky syndrome, as well as its role in aging-related disorders and illnesses, remain largely unknown. In this editorial, we discuss potential mechanisms that originate from the gut and resident microbes (microbiome) to contribute in leaky syndrome. Furthermore, we explore how the food we consume can impact the development of leaky syndrome, potentially influencing the biology of aging and challenges to diagnose the leaky gut condition accurately and clinically.
Collapse
Affiliation(s)
- Shalini Jain
- Department of Neurosurgery and Brain Repair, University of South Florida, Tampa, FL, USA
- USF Center for Microbiome Research, Microbiomes Institute, University of South Florida, Tampa, FL, USA
| | | | - Leila Haghshenas
- Department of Clinical Bioinformatics, Harvard Medical School, Boston, MA, USA
| | - Hariom Yadav
- Department of Neurosurgery and Brain Repair, University of South Florida, Tampa, FL, USA
- USF Center for Microbiome Research, Microbiomes Institute, University of South Florida, Tampa, FL, USA
- Department of Internal Medicine- Digestive Diseases and Nutrition, University of South Florida, Tampa, FL, USA
| |
Collapse
|
38
|
Chen J, Li Y, Chen M, Liu X, Chen J, Li X, Wang C, Wan G, Tian J. Pantethine Ameliorates Recognition Impairment in a Mouse Model of Alzheimer's Disease by Modulating Cholesterol Content and Intestinal Flora Species. Mol Nutr Food Res 2023; 67:e2200799. [PMID: 37194410 DOI: 10.1002/mnfr.202200799] [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: 11/15/2022] [Revised: 05/02/2023] [Indexed: 05/18/2023]
Abstract
SCOPE As a natural dietary low-molecular-weight thiol, pantethine helps maintain brain homeostasis and function in Alzheimer's disease (AD) mice. The current study aims to investigate the protective effects and underlying mechanisms of pantethine on the mitigation of cognitive deficits and pathology in a triple transgenic AD mouse model. METHODS AND RESULTS Compared to control mice, oral administration of pantethine improve spatial learning and memory ability, relieve anxiety, and reduce the production of amyloid-β (Aβ), neuronal damage, and inflammation in 3×Tg-AD mice. Pantethine reduces body weight, body fat, and the production of cholesterol in 3×Tg-AD mice by inhibiting sterol regulatory element-binding protein (SREBP2) signal pathway and apolipoprotein E (APOE) expression; lipid rafts in the brain, which are necessary for the processing of the Aβ precursor protein (APP), are also decreased. In addition, pantethine regulates the composition, distribution, and abundance of characteristic flora in the intestine; these floras are considered protective and anti-inflammatory in the gastrointestinal tract, suggesting a possible improvement in the gut flora of 3×Tg-AD mice. CONCLUSION This study highlights the potential therapeutic effect of pantethine in AD by reducing cholesterol and lipid raft formation and regulating intestinal flora, suggesting a new option for the development of clinical drugs for AD.
Collapse
Affiliation(s)
- Jianfeng Chen
- Shenzhen Key Laboratory of Marine Biotechnology and Ecology, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, Guangdong, 518055, China
| | - Yongsui Li
- Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, 518055, China
| | - Minyu Chen
- Shenzhen Key Laboratory of Marine Biotechnology and Ecology, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, Guangdong, 518055, China
| | - Xinwei Liu
- Shenzhen Key Laboratory of Marine Biotechnology and Ecology, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, Guangdong, 518055, China
| | - Jinghong Chen
- Shenzhen Key Laboratory of Marine Biotechnology and Ecology, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, Guangdong, 518055, China
- Shenzhen Bay Laboratory, Shenzhen, Guangdong, 518055, China
| | - Xinlu Li
- Shenzhen Key Laboratory of Marine Biotechnology and Ecology, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, Guangdong, 518055, China
| | - Chao Wang
- Chemical Analysis & Physical Testing Institute, Shenzhen Center for Disease Control and Prevention, Shenzhen, Guangdong, 518055, China
| | - Guohui Wan
- National-Local Joint Engineering Laboratory of Druggability and New Drug Evaluation, National Engineering Research Center for New Drug and Druggability (cultivation), Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, Guangdong, 510000, China
| | - Jing Tian
- Shenzhen Key Laboratory of Marine Biotechnology and Ecology, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, Guangdong, 518055, China
| |
Collapse
|
39
|
Cui M, Yu P, Liu TC, Liu J, Li K, Zhou P, Liu X. Dose-dependent effects of xylooligosaccharides on glycemic regulation with L. rhamnosus CCFM1060 in diabetic mice. Food Funct 2023. [PMID: 37325857 DOI: 10.1039/d3fo00162h] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Dietary intervention with the probiotic Lactobacillus rhamnosus CCFM1060 has been proved to be effective on glycemic regulation in diabetic mice. Therefore characterization of the potential symbiotic effect of prebiotic xylooligosaccharides (XOS) with L. rhamnosus CCFM1060 would be desirable. In this study, we evaluated any dose-dependent relationship between XOS and L. rhamnosus CCFM1060, and the potential impact on glycemic regulation. Diabetic mice were randomly assigned to receive 5 × 109 CFU mL-1L. rhamnosus CCFM1060, 5 × 109 CFU mL-1L. rhamnosus CCFM1060 with 250 mg kg-1 XOS (L-LXOS), or 5 × 109 CFU mL-1L. rhamnosus CCFM1060 with 500 mg kg-1 XOS (L-HXOS) for 7 weeks. In addition to characterization of the host metabolism, the intestinal microbiota were analyzed using 16S rRNA gene sequencing. The results showed that L. rhamnosus alone and L-LXOS intervention significantly alleviated diabetes symptoms and increased the populations of short-chain fatty acid (SCFA)-producing bacteria. The intake of L-HXOS had an adverse effect on glucose metabolism, causing increased insulin resistance and inflammation. Although a significant increase in the relative abundance of Bifidobacterium was observed in the L-HXOS group, the abundance of SCFA-producing bacteria, such as Romboutsia and Clostrudium sensu stricto 1, decreased. KEGG pathway analysis revealed that the adverse effects of L-HXOS intervention might be attributed to the metabolic pathways involved in amino acid, cofactor, and vitamin metabolism. This study revealed that L. rhamnosus CCFM1060 combined with different doses of XOS exerted dose-dependent effects on glucose metabolism. Therefore, the type and dose of prebiotics should be carefully evaluated when developing individualized symbiotic formula.
Collapse
Affiliation(s)
- Mengjun Cui
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, Jiangsu, China.
| | - Peng Yu
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, Jiangsu, China.
| | - Tristan C Liu
- Taicang Port Economic and Technological Development Zone New Zone, Standard Foods (China) Co., Ltd, No. 88 Dalian West Road, Suzhou, Jiangsu, P. R. China
| | - Jianguo Liu
- Taicang Port Economic and Technological Development Zone New Zone, Standard Foods (China) Co., Ltd, No. 88 Dalian West Road, Suzhou, Jiangsu, P. R. China
| | - Kexin Li
- Le Bonta Wellness Co., Ltd, Room 5, 4th Floor, Building 1, No. 39, Jiatai Road, Pilot Free Trade Zone, Shanghai, China
| | - Peng Zhou
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, Jiangsu, China.
| | - Xiaoming Liu
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China.
| |
Collapse
|
40
|
Jawhara S. Healthy Diet and Lifestyle Improve the Gut Microbiota and Help Combat Fungal Infection. Microorganisms 2023; 11:1556. [PMID: 37375058 DOI: 10.3390/microorganisms11061556] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 06/08/2023] [Accepted: 06/09/2023] [Indexed: 06/29/2023] Open
Abstract
Western diets are rapidly spreading due to globalization, causing an increase in obesity and diseases of civilization. These Western diets are associated with changes in the gut microbiota related to intestinal inflammation. This review discusses the adverse effects of Western diets, which are high in fat and sugar and low in vegetable fiber, on the gut microbiota. This leads to gut dysbiosis and overgrowth of Candida albicans, which is a major cause of fungal infection worldwide. In addition to an unhealthy Western diet, other factors related to disease development and gut dysbiosis include smoking, excessive alcohol consumption, lack of physical activity, prolonged use of antibiotics, and chronic psychological stress. This review suggests that a diversified diet containing vegetable fiber, omega-3 polyunsaturated fatty acids, vitamins D and E, as well as micronutrients associated with probiotic or prebiotic supplements can improve the biodiversity of the microbiota, lead to short-chain fatty acid production, and reduce the abundance of fungal species in the gut. The review also discusses a variety of foods and plants that are effective against fungal overgrowth and gut dysbiosis in traditional medicine. Overall, healthy diets and lifestyle factors contribute to human well-being and increase the biodiversity of the gut microbiota, which positively modulates the brain and central nervous system.
Collapse
Affiliation(s)
- Samir Jawhara
- UMR 8576-UGSF-Unité de Glycobiologie Structurale et Fonctionnelle, Centre National de la Recherche Scientifique, F-59000 Lille, France
- Institut National de la Santé et de la Recherche Médicale U1285, University of Lille, F-59000 Lille, France
- Medicine Faculty, University of Lille, F-59000 Lille, France
| |
Collapse
|
41
|
Li S, Zheng J, He J, Liu H, Huang Y, Huang L, Wang K, Zhao X, Feng B, Che L, Fang Z, Li J, Xu S, Lin Y, Jiang X, Hua L, Zhuo Y, Wu D. Dietary fiber during gestation improves lactational feed intake of sows by modulating gut microbiota. J Anim Sci Biotechnol 2023; 14:65. [PMID: 37143119 PMCID: PMC10161572 DOI: 10.1186/s40104-023-00870-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 03/14/2023] [Indexed: 05/06/2023] Open
Abstract
BACKGROUND The feed intake of sows during lactation is often lower than their needs. High-fiber feed is usually used during gestation to increase the voluntary feed intake of sows during lactation. However, the mechanism underlying the effect of bulky diets on the appetites of sows during lactation have not been fully clarified. The current study was conducted to determine whether a high-fiber diet during gestation improves lactational feed intake (LFI) of sows by modulating gut microbiota. METHODS We selected an appropriate high-fiber diet during gestation and utilized the fecal microbial transplantation (FMT) method to conduct research on the role of the gut microbiota in feed intake regulation of sows during lactation, as follows: high-fiber (HF) diet during gestation (n = 23), low-fiber (LF) diet during gestation (n = 23), and low-fiber diet + HF-FMT (LFM) during gestation (n = 23). RESULTS Compared with the LF, sows in the HF and LFM groups had a higher LFI, while the sows also had higher peptide tyrosine tyrosine and glucagon-like peptide 1 on d 110 of gestation (G110 d). The litter weight gain of piglets during lactation and weaning weight of piglets from LFM group were higher than LF group. Sows given a HF diet had lower Proteobacteria, especially Escherichia-Shigella, on G110 d and higher Lactobacillus, especially Lactobacillus_mucosae_LM1 and Lactobacillus_amylovorus, on d 7 of lactation (L7 d). The abundance of Escherichia-Shigella was reduced by HF-FMT in numerically compared with the LF. In addition, HF and HF-FMT both decreased the perinatal concentrations of proinflammatory factors, such as endotoxin (ET), lipocalin-2 (LCN-2), tumor necrosis factor-α (TNF-α), and interleukin-1β (IL-1β). The concentration of ET and LCN-2 and the abundance of Proteobacteria and Escherichia-Shigella were negatively correlated with the LFI of sows. CONCLUSION The high abundance of Proteobacteria, especially Escherichia-Shigella of LF sows in late gestation, led to increased endotoxin levels, which result in inflammatory responses and adverse effects on the LFI of sows. Adding HF during gestation reverses this process by increasing the abundance of Lactobacillus, especially Lactobacillus_mucosae_LM1 and Lactobacillus_amylovorus.
Collapse
Affiliation(s)
- Shuang Li
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, No. 211, Huimin Road, Wenjiang District, Chengdu, 611130, Sichuan, People's Republic of China
| | - Jie Zheng
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, No. 211, Huimin Road, Wenjiang District, Chengdu, 611130, Sichuan, People's Republic of China
| | - Jiaqi He
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, No. 211, Huimin Road, Wenjiang District, Chengdu, 611130, Sichuan, People's Republic of China
| | - Hao Liu
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, No. 211, Huimin Road, Wenjiang District, Chengdu, 611130, Sichuan, People's Republic of China
| | - Yingyan Huang
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, No. 211, Huimin Road, Wenjiang District, Chengdu, 611130, Sichuan, People's Republic of China
| | - Liansu Huang
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, No. 211, Huimin Road, Wenjiang District, Chengdu, 611130, Sichuan, People's Republic of China
| | - Ke Wang
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, No. 211, Huimin Road, Wenjiang District, Chengdu, 611130, Sichuan, People's Republic of China
| | - Xilun Zhao
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, No. 211, Huimin Road, Wenjiang District, Chengdu, 611130, Sichuan, People's Republic of China
| | - Bin Feng
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, No. 211, Huimin Road, Wenjiang District, Chengdu, 611130, Sichuan, People's Republic of China
| | - Lianqiang Che
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, No. 211, Huimin Road, Wenjiang District, Chengdu, 611130, Sichuan, People's Republic of China
| | - Zhengfeng Fang
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, No. 211, Huimin Road, Wenjiang District, Chengdu, 611130, Sichuan, People's Republic of China
| | - Jian Li
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, No. 211, Huimin Road, Wenjiang District, Chengdu, 611130, Sichuan, People's Republic of China
| | - Shengyu Xu
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, No. 211, Huimin Road, Wenjiang District, Chengdu, 611130, Sichuan, People's Republic of China
| | - Yan Lin
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, No. 211, Huimin Road, Wenjiang District, Chengdu, 611130, Sichuan, People's Republic of China
| | - Xuemei Jiang
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, No. 211, Huimin Road, Wenjiang District, Chengdu, 611130, Sichuan, People's Republic of China
| | - Lun Hua
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, No. 211, Huimin Road, Wenjiang District, Chengdu, 611130, Sichuan, People's Republic of China
| | - Yong Zhuo
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, No. 211, Huimin Road, Wenjiang District, Chengdu, 611130, Sichuan, People's Republic of China.
| | - De Wu
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, No. 211, Huimin Road, Wenjiang District, Chengdu, 611130, Sichuan, People's Republic of China.
| |
Collapse
|
42
|
Clemente-Suárez VJ, Redondo-Flórez L, Beltrán-Velasco AI, Martín-Rodríguez A, Martínez-Guardado I, Navarro-Jiménez E, Laborde-Cárdenas CC, Tornero-Aguilera JF. The Role of Adipokines in Health and Disease. Biomedicines 2023; 11:biomedicines11051290. [PMID: 37238961 DOI: 10.3390/biomedicines11051290] [Citation(s) in RCA: 152] [Impact Index Per Article: 76.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 04/21/2023] [Accepted: 04/25/2023] [Indexed: 05/28/2023] Open
Abstract
Adipokines are cell-signaling proteins secreted by adipose tissue that has been related to a low-grade state of inflammation and different pathologies. The present review aims to analyze the role of adipokines in health and disease in order to understand the important functions and effects of these cytokines. For this aim, the present review delves into the type of adipocytes and the cytokines produced, as well as their functions; the relations of adipokines in inflammation and different diseases such as cardiovascular, atherosclerosis, mental diseases, metabolic disorders, cancer, and eating behaviors; and finally, the role of microbiota, nutrition, and physical activity in adipokines is discussed. This information would allow for a better understanding of these important cytokines and their effects on body organisms.
Collapse
Affiliation(s)
| | - Laura Redondo-Flórez
- Department of Health Sciences, Faculty of Biomedical and Health Sciences, Universidad Europea de Madrid, C/Tajo s/n, 28670 Madrid, Spain
| | - Ana Isabel Beltrán-Velasco
- Department of Psychology, Faculty of Life and Natural Sciences, University of Nebrija, C/del Hostal, 28248 Madrid, Spain
| | | | - Ismael Martínez-Guardado
- BRABE Group, Department of Psychology, Faculty of Life and Natural Sciences, University of Nebrija, C/del Hostal, 28248 Madrid, Spain
| | | | | | | |
Collapse
|
43
|
Xie Z, Zhang G, Liu R, Wang Y, Tsapieva AN, Zhang L, Han J. Heat-Killed Lacticaseibacillus paracasei Repairs Lipopolysaccharide-Induced Intestinal Epithelial Barrier Damage via MLCK/MLC Pathway Activation. Nutrients 2023; 15:nu15071758. [PMID: 37049598 PMCID: PMC10097264 DOI: 10.3390/nu15071758] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 03/30/2023] [Accepted: 03/31/2023] [Indexed: 04/14/2023] Open
Abstract
Intestinal epithelial barrier function is closely associated with the development of many intestinal diseases. Heat-killed Lacticaseibacillus paracasei (HK-LP) has been shown to improve intestinal health and enhance immunity. However, the function of HK-LP in the intestinal barrier is still unclear. This study characterized the inflammatory effects of seven HK-LP (1 μg/mL) on the intestinal barrier using lipopolysaccharide (LPS) (100 μg/mL)-induced Caco-2 cells. In this study, HK-LP 6105, 6115, and 6235 were selected, and their effects on the modulation of inflammatory factors and tight junction protein expression (claudin-1, zona occludens-1, and occludin) were compared. The effect of different cultivation times (18 and 48 h) was investigated in response to LPS-induced intestinal epithelial barrier dysfunction. Our results showed that HK-LP 6105, 6115, and 6235 improved LPS-induced intestinal barrier permeability reduction and transepithelial resistance. Furthermore, HK-LP 6105, 6115, and 6235 inhibited the pro-inflammatory factors (TNF-α, IL-1β, IL-6) and increased the expression of the anti-inflammatory factors (IL-4, IL-10, and TGF-β). HK-LP 6105, 6115, and 6235 ameliorated the inflammatory response. It inhibited the nuclear factor kappa B (NF-κB) signaling pathway-mediated myosin light chain (MLC)/MLC kinase signaling pathway by downregulating the Toll-like receptor 4 (TLR4)/NF-κB pathway. Thus, the results suggest that HK-LP 6150, 6115, and 6235 may improve intestinal health by regulating inflammation and TJ proteins. Postbiotics produced by these strains exhibit anti-inflammatory properties that can protect the intestinal barrier.
Collapse
Affiliation(s)
- Zhixin Xie
- College of Food Science, Northeast Agricultural University, Harbin 150030, China
| | - Gongsheng Zhang
- College of Food Science, Northeast Agricultural University, Harbin 150030, China
| | - Rongxu Liu
- Heilongjiang Green Food Science Research Institute, Harbin 150030, China
| | - Yucong Wang
- College of Food Science, Northeast Agricultural University, Harbin 150030, China
| | - Anna N Tsapieva
- Department of Molecular Microbiology, FSBSI Institute of Experimental Medicine, Acad.,197376 St. Petersburg, Russia
| | - Lili Zhang
- College of Food Science, Northeast Agricultural University, Harbin 150030, China
| | - Jianchun Han
- College of Food Science, Northeast Agricultural University, Harbin 150030, China
- Heilongjiang Green Food Science Research Institute, Harbin 150030, China
| |
Collapse
|
44
|
Youn HY, Kim HJ, Kim DH, Jang YS, Kim H, Seo KH. Gut microbiota modulation via short-term administration of potential probiotic kefir yeast Kluyveromyces marxianus A4 and A5 in BALB/c mice. Food Sci Biotechnol 2023; 32:589-598. [PMID: 36911334 PMCID: PMC9992467 DOI: 10.1007/s10068-023-01268-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 01/19/2023] [Accepted: 01/26/2023] [Indexed: 02/16/2023] Open
Abstract
Kefir yeast, Kluyveromyces marxianus, has been evaluated for its potential probiotic properties-survivability, non-pathogenicity, and antioxidant and anti-microbial activities. However, host gut microbiota modulation of kefir yeasts remains unclear. Here, we compared kefir yeast strains K. marxianus A4 (Km A4) and K. marxianus A5 (Km A5) with Saccharomyces boulardii ATCC MYA-796 (Sb MYA-796) by investigating their adherence to colorectal adenocarcinoma (Caco-2) cells and gut microbiota modulation in BALB/c mice. The kefir yeast strains exhibited higher intestinal cell adhesion than Sb MYA-796 (p < 0.05). Bacteroidetes, Bacteroidales, and Bacteroides were more abundant in the 1 × 108 CFU/mL of Km A4 treatment group than in the control group (p < 0.05). Moreover, 1 × 108 CFU/mL of Km A5 increased Corynebacteriales and Corynebacterium compared to the 1 × 108 CFU/mL of Km A4 treatment group (p < 0.01). The results showed that Km A4 and Km A5 had good Caco-2 cell adhesion ability and modulated gut microbiota upon short-term administration in healthy mice. Supplementary Information The online version contains supplementary material available at 10.1007/s10068-023-01268-3.
Collapse
Affiliation(s)
- Hye-Young Youn
- Center for One Health, Department of Veterinary Public Health, College of Veterinary Medicine, Konkuk University, Seoul, 05029 Republic of Korea
| | - Hyeon-Jin Kim
- Center for One Health, Department of Veterinary Public Health, College of Veterinary Medicine, Konkuk University, Seoul, 05029 Republic of Korea
| | - Dong-Hyeon Kim
- Center for One Health, Department of Veterinary Public Health, College of Veterinary Medicine, Konkuk University, Seoul, 05029 Republic of Korea
| | - Yong-Seok Jang
- Center for One Health, Department of Veterinary Public Health, College of Veterinary Medicine, Konkuk University, Seoul, 05029 Republic of Korea
| | - Hyunsook Kim
- Department of Food & Nutrition, College of Human Ecology, Hanyang University, Seoul, 04763 Republic of Korea
| | - Kun-Ho Seo
- Center for One Health, Department of Veterinary Public Health, College of Veterinary Medicine, Konkuk University, Seoul, 05029 Republic of Korea
| |
Collapse
|
45
|
The Role of Gut Microbiota in High-Fat-Diet-Induced Diabetes: Lessons from Animal Models and Humans. Nutrients 2023; 15:nu15040922. [PMID: 36839280 PMCID: PMC9963658 DOI: 10.3390/nu15040922] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 02/02/2023] [Accepted: 02/10/2023] [Indexed: 02/15/2023] Open
Abstract
The number of diabetes mellitus patients is increasing rapidly worldwide. Diet and nutrition are strongly believed to play a significant role in the development of diabetes mellitus. However, the specific dietary factors and detailed mechanisms of its development have not been clearly elucidated. Increasing evidence indicates the intestinal microbiota is becoming abundantly apparent in the progression and prevention of insulin resistance in diabetes. Differences in gut microbiota composition, particularly butyrate-producing bacteria, have been observed in preclinical animal models as well as human patients compared to healthy controls. Gut microbiota dysbiosis may disrupt intestinal barrier functions and alter host metabolic pathways, directly or indirectly relating to insulin resistance. In this article, we focus on dietary fat, diabetes, and gut microbiome characterization. The promising probiotic and prebiotic approaches to diabetes, by favorably modifying the composition of the gut microbial community, warrant further investigation through well-designed human clinical studies.
Collapse
|
46
|
Interaction between gut microbiota and sex hormones and their relation to sexual dimorphism in metabolic diseases. Biol Sex Differ 2023; 14:4. [PMID: 36750874 PMCID: PMC9903633 DOI: 10.1186/s13293-023-00490-2] [Citation(s) in RCA: 54] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Accepted: 01/30/2023] [Indexed: 02/09/2023] Open
Abstract
Metabolic diseases, such as obesity, metabolic syndrome (MetS) and type 2 diabetes (T2D), are now a widespread pandemic in the developed world. These pathologies show sex differences in their development and prevalence, and sex steroids, mainly estrogen and testosterone, are thought to play a prominent role in this sexual dimorphism. The influence of sex hormones on these pathologies is not only reflected in differences between men and women, but also between women themselves, depending on the hormonal changes associated with the menopause. The observed sex differences in gut microbiota composition have led to multiple studies highlighting the interaction between steroid hormones and the gut microbiota and its influence on metabolic diseases, ultimately pointing to a new therapy for these diseases based on the manipulation of the gut microbiota. This review aims to shed light on the role of sexual hormones in sex differences in the development and prevalence of metabolic diseases, focusing on obesity, MetS and T2D. We focus also the interaction between sex hormones and the gut microbiota, and in particular the role of microbiota in aspects such as gut barrier integrity, inflammatory status, and the gut-brain axis, given the relevance of these factors in the development of metabolic diseases.
Collapse
|
47
|
Liu B, Zhang L, Yang H, Zheng H, Liao X. Microbiota: A potential orchestrator of antidiabetic therapy. Front Endocrinol (Lausanne) 2023; 14:973624. [PMID: 36777348 PMCID: PMC9911464 DOI: 10.3389/fendo.2023.973624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 01/17/2023] [Indexed: 01/28/2023] Open
Abstract
The gut microbiota, as a 'new organ' of humans, has been identified to affect many biological processes, including immunity, inflammatory response, gut-brain neural circuits, and energy metabolism. Profound dysbiosis of the gut microbiome could change the metabolic pattern, aggravate systemic inflammation and insulin resistance, and exacerbate metabolic disturbance and the progression of type 2 diabetes (T2D). The aim of this review is to focus on the potential roles and functional mechanisms of gut microbiota in the antidiabetic therapy. In general, antidiabetic drugs (α-glucosidase inhibitor, biguanides, incretin-based agents, and traditional Chinese medicine) induce the alteration of microbial diversity and composition, and the levels of bacterial component and derived metabolites, such as lipopolysaccharide (LPS), short chain fatty acids (SCFAs), bile acids and indoles. The altered microbial metabolites are involved in the regulation of gut barrier, inflammation response, insulin resistance and glucose homeostasis. Furthermore, we summarize the new strategies for antidiabetic treatment based on microbial regulation, such as pro/prebiotics administration and fecal microbiota transplantation, and discuss the need for more basic and clinical researches to evaluate the feasibility and efficacy of the new therapies for diabetes.
Collapse
Affiliation(s)
| | | | | | - Hongting Zheng
- Department of Endocrinology, Chongqing Education Commission Key Laboratory of Diabetic Translational Research, the Second Affiliated Hospital of Army Medical University, Chongqing, China
| | - Xiaoyu Liao
- Department of Endocrinology, Chongqing Education Commission Key Laboratory of Diabetic Translational Research, the Second Affiliated Hospital of Army Medical University, Chongqing, China
| |
Collapse
|
48
|
Gaike AH, Kalamkar SD, Gajjar V, Divate U, Karandikar-Iyer S, Goel P, Shouche YS, Ghaskadbi SS. Effect of long-term oral glutathione supplementation on gut microbiome of type 2 diabetic individuals. FEMS Microbiol Lett 2023; 370:fnad116. [PMID: 37935462 DOI: 10.1093/femsle/fnad116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 10/23/2023] [Accepted: 11/02/2023] [Indexed: 11/09/2023] Open
Abstract
The aim of this study was to check the effect of long-term oral glutathione (GSH) supplementation on alteration in gut microbiome of Indian diabetic individuals. Early morning fresh stool sample of diabetic individuals recruited in a randomized clinical trial wherein they were given 500 mg GSH supplementation orally once a day for a period of 6 months was collected and gut microbiome was analysed using high throughput 16S rRNA metagenomic sequencing. Long-term GSH supplementation as reported in our earlier work showed significant increase in body stores of GSH and stabilized decreased glycated haemoglobin (HbA1c). Analysis of gut microbiome revealed that abundance of phylum Proteobacteria significantly decreased (P < 0.05) in individuals with GSH supplementation after 6 months compared to those without it. Beneficial dominant genera such as Megasphaera, Bacteroides, and Megamonas were found to be significantly enriched (P < 0.05), while pathogenic Escherichia/Shigella was found to be depleted (P < 0.05) after supplementation. Data clearly demonstrate that GSH supplementation along with antidiabetic treatment helps restore the gut microbiome by enriching beneficial bacteria of healthy gut and reducing significantly the load of pathogenic bacteria of diabetic gut.
Collapse
Affiliation(s)
- Akshay H Gaike
- Department of Zoology, Savitribai Phule Pune University, Pune 411007, India
- National Centre for Cell Science, Pune 411007, India
| | - Saurabh D Kalamkar
- Department of Zoology, Savitribai Phule Pune University, Pune 411007, India
| | | | | | | | - Pranay Goel
- Biology Division, Indian Institute of Science Education and Research, Pune 411008, India
| | - Yogesh S Shouche
- National Centre for Cell Science, Pune 411007, India
- SKAN Research Trust, Bengaluru 560034, India
| | - Saroj S Ghaskadbi
- Department of Zoology, Savitribai Phule Pune University, Pune 411007, India
| |
Collapse
|
49
|
Makwana S, Prajapati JB, Pipaliya R, Hati S. Effects of probiotic fermented milk on management of obesity studied in high-fat-diet induced obese rat model. FOOD PRODUCTION, PROCESSING AND NUTRITION 2023. [DOI: 10.1186/s43014-022-00112-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
AbstractThe current study aimed to explore the hypothesis that probiotic bacteria are significantly involved in the control of obesity using Wistar rats as the test group by feeding high fat diets (HFD) induced obesity. A total of four groups of rats were considered viz., normal pellet diet fed (NC), HFD fed (DC), HFD fed rats treated with probiotic fermented milk with soy protein isolate (SPI) and whey protein concentrate (WPC) (T1), HFD fed rats treated with probiotic fermented milk without WPC and SPI (T2). Body weight, abdominal fat weight, liver weight, serum Alanine aminotransferase level, and alkaline phosphatase level significantly (p < 0.05) decreased after giving daily probiotic milk product supplementation with @ 2 ml per day for continuous 4 weeks. Whereas, C-reactive protein and Aspartate aminotransferase levels were not altered to a significant extent. The histology of the liver from the disease model group showed large lipid vacuoles deposited in the parenchyma cells. Product T2 confirmed fewer micro vesicular fatty changes and the appearance of T2 was better than T1. Overall, the in vivo study results indicated that the probiotic fermented milk exerted a better anti-obesity effect.
Graphical Abstract
Collapse
|
50
|
Tu Y, Kuang X, Zhang L, Xu X. The associations of gut microbiota, endocrine system and bone metabolism. Front Microbiol 2023; 14:1124945. [PMID: 37089533 PMCID: PMC10116073 DOI: 10.3389/fmicb.2023.1124945] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 03/16/2023] [Indexed: 04/25/2023] Open
Abstract
Gut microbiota is of great importance in human health, and its roles in the maintenance of skeletal homeostasis have long been recognized as the "gut-bone axis." Recent evidence has indicated intercorrelations between gut microbiota, endocrine system and bone metabolism. This review article discussed the complex interactions between gut microbiota and bone metabolism-related hormones, including sex steroids, insulin-like growth factors, 5-hydroxytryptamine, parathyroid hormone, glucagon-like peptides, peptide YY, etc. Although the underlying mechanisms still need further investigation, the regulatory effect of gut microbiota on bone health via interplaying with endocrine system may provide a new paradigm for the better management of musculoskeletal disorders.
Collapse
Affiliation(s)
- Ye Tu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xinyi Kuang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Ling Zhang
- Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- *Correspondence: Ling Zhang,
| | - Xin Xu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Xin Xu,
| |
Collapse
|