1
|
Gamal R, Shreadah MA. Marine microalgae and their industrial biotechnological applications: A review. J Genet Eng Biotechnol 2024; 22:100407. [PMID: 39674656 PMCID: PMC11387356 DOI: 10.1016/j.jgeb.2024.100407] [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: 03/08/2024] [Revised: 07/03/2024] [Accepted: 07/31/2024] [Indexed: 12/16/2024]
Abstract
BACKGROUND For use in specialized programs in the food, pharmaceutical, nutraceutical, cosmetic, and animal feed sectors, micro-algal biomass has been generated industrially. They can be grown in closed buildings, such as photobioreactors, or open structures. The utilization of biomass from microalgae for energy production is another crucial topic. Because of the world's diminishing petroleum sources and the greenhouse gas emissions from gasoline lines, it is now obvious that fuels generated from petroleum are not sustainable. RESULTS Microalgae can produce a variety of unique, sustainable biofuels. These include biodiesel made from trans-esterification of microalgal lipids, bioethanol from fermentation of carbohydrates, methane created by anaerobic digestion of algal biomass, and biohydrogen produced by photobiological processes. The idea of using microalgae as a fuel source is not entirely novel. CONCLUSION This analysis emphasizes the significance of recent and noteworthy advancements in the industrial usage of microalgae, with an emphasis on their biotechnological applications.
Collapse
Affiliation(s)
- Reham Gamal
- National Institute of Oceanography and Fisheries, Cairo, Egypt.
| | | |
Collapse
|
2
|
Pannerchelvan S, Rios-Solis L, Wasoh H, Sobri MZM, Faizal Wong FW, Mohamed MS, Mohamad R, Halim M. Functional yogurt: a comprehensive review of its nutritional composition and health benefits. Food Funct 2024; 15:10927-10955. [PMID: 39446126 DOI: 10.1039/d4fo03671a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2024]
Abstract
Functional yogurt, renowned for its enhanced nutritional profile and potential health benefits, has emerged as a promising functional food. This review meticulously examines the nutritional composition of functional yogurt, highlighting its enriched content of probiotics, prebiotics, synbiotics, antioxidants, vitamins, minerals, proteins, and other bioactive compounds, which contribute to its health-promoting properties. Functional yogurt has positively affected digestive health, immune function, metabolic health, and mental well-being. It benefits digestive health by alleviating diarrhoeal symptoms, constipation, colon cancer, irritable bowel syndrome (IBS), Helicobacter pylori infection, and digestive-related allergies. Moreover, the immune-boosting properties of functional yogurt play a pivotal role in reducing the risk of infections and inflammation. In addition, functional yogurt has the potential to improve metabolic health, leading to decreased cholesterol levels and enhanced blood sugar regulation. Emerging research also suggests functional yogurt may positively influence mood, behavior, and cognitive function. Functional yogurt is a valuable addition to the human diet, holding significant implications for public health. In addition to its numerous health benefits, functional yogurt also faces limitations, such as the stability of functional compounds, sensory alterations, potential digestive discomfort, and inconsistent efficacy across populations, highlighting the need for further research and optimization.
Collapse
Affiliation(s)
- Sangkaran Pannerchelvan
- Department of Bioprocess Technology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia.
| | - Leonardo Rios-Solis
- Department of Biochemical Engineering, University College London, Gower Street, Bernard Katz Building, 6.07, WC1E 6BT, United Kingdom
| | - Helmi Wasoh
- Department of Bioprocess Technology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia.
- Bioprocessing and Biomanufacturing Complex, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia
| | - Mohamad Zulfazli Mohd Sobri
- Department of Bioprocess Technology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia.
- Bioprocessing and Biomanufacturing Complex, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia
| | - Fadzlie Wong Faizal Wong
- Department of Bioprocess Technology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia.
- Bioprocessing and Biomanufacturing Complex, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia
| | - Mohd Shamzi Mohamed
- Department of Bioprocess Technology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia.
- Bioprocessing and Biomanufacturing Complex, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia
| | - Rosfarizan Mohamad
- Department of Bioprocess Technology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia.
- Bioprocessing and Biomanufacturing Complex, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia
| | - Murni Halim
- Department of Bioprocess Technology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia.
- Bioprocessing and Biomanufacturing Complex, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia
| |
Collapse
|
3
|
Vijaya AK, Kuras S, Šimoliūnas E, Mingaila J, Makovskytė K, Buišas R, Daliri EBM, Meškys R, Baltriukienė D, Burokas A. Prebiotics Mitigate the Detrimental Effects of High-Fat Diet on memory, anxiety and microglia functionality in Ageing Mice. Brain Behav Immun 2024; 122:167-184. [PMID: 39142421 DOI: 10.1016/j.bbi.2024.08.022] [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: 04/03/2024] [Revised: 08/07/2024] [Accepted: 08/10/2024] [Indexed: 08/16/2024] Open
Abstract
Ageing is characterised by a progressive increase in systemic inflammation and especially neuroinflammation. Neuroinflammation is associated with altered brain states that affect behaviour, such as an increased level of anxiety with a concomitant decline in cognitive abilities. Although multiple factors play a role in the development of neuroinflammation, microglia have emerged as a crucial target. Microglia are the only macrophage population in the CNS parenchyma that plays a crucial role in maintaining homeostasis and in the immune response, which depends on the activation and subsequent deactivation of microglia. Therefore, microglial dysfunction has a major impact on neuroinflammation. The gut microbiota has been shown to significantly influence microglia from birth to adulthood in terms of development, proliferation, and function. Diet is a key modulating factor that influences the composition of the gut microbiota, along with prebiotics that support the growth of beneficial gut bacteria. Although the role of diet in neuroinflammation and behaviour has been well established, its relationship with microglia functionality is less explored. This article establishes a link between diet, animal behaviour and the functionality of microglia. The results of this research stem from experiments on mouse behaviour, i.e., memory, anxiety, and studies on microglia functionality, i.e., cytochemistry (phagocytosis, cellular senescence, and ROS assays), gene expression and protein quantification. In addition, shotgun sequencing was performed to identify specific bacterial families that may play a crucial role in the brain function. The results showed negative effects of long-term consumption of a high fat diet on ageing mice, epitomised by increased body weight, glucose intolerance, anxiety, cognitive impairment and microglia dysfunction compared to ageing mice on a control diet. These effects were a consequence of the changes in gut microbiota modulated by the diet. However, by adding the prebiotics fructo- and galacto-oligosaccharides, we were able to mitigate the deleterious effects of a long-term high-fat diet.
Collapse
Affiliation(s)
- Akshay Kumar Vijaya
- Department of Biological Models, Institute of Biochemistry, Life Sciences Center, Vilnius University, Sauletekio Ave. 7, LT-10257 Vilnius, Lithuania
| | - Simonas Kuras
- Department of Biological Models, Institute of Biochemistry, Life Sciences Center, Vilnius University, Sauletekio Ave. 7, LT-10257 Vilnius, Lithuania
| | - Egidijus Šimoliūnas
- Department of Biological Models, Institute of Biochemistry, Life Sciences Center, Vilnius University, Sauletekio Ave. 7, LT-10257 Vilnius, Lithuania
| | - Jonas Mingaila
- Department of Biological Models, Institute of Biochemistry, Life Sciences Center, Vilnius University, Sauletekio Ave. 7, LT-10257 Vilnius, Lithuania
| | - Karolina Makovskytė
- Department of Biological Models, Institute of Biochemistry, Life Sciences Center, Vilnius University, Sauletekio Ave. 7, LT-10257 Vilnius, Lithuania
| | - Rokas Buišas
- Department of Neurobiology and Biophysics, Institute of Bioscience, Life Sciences Center, Vilnius University, Sauletekio Ave. 7, LT-10257 Vilnius, Lithuania
| | - Eric Banan-Mwine Daliri
- Department of Biological Models, Institute of Biochemistry, Life Sciences Center, Vilnius University, Sauletekio Ave. 7, LT-10257 Vilnius, Lithuania
| | - Rolandas Meškys
- Department of Molecular Microbiology and Biotechnology, Institute of Biochemistry, Life Sciences Center, Vilnius University, Sauletekio Ave. 7, LT-10257 Vilnius, Lithuania
| | - Daiva Baltriukienė
- Department of Biological Models, Institute of Biochemistry, Life Sciences Center, Vilnius University, Sauletekio Ave. 7, LT-10257 Vilnius, Lithuania.
| | - Aurelijus Burokas
- Department of Biological Models, Institute of Biochemistry, Life Sciences Center, Vilnius University, Sauletekio Ave. 7, LT-10257 Vilnius, Lithuania.
| |
Collapse
|
4
|
Liu X, Du P, Xu J, Wang W, Zhang C. Therapeutic Effects of Intermittent Fasting Combined with SLBZS and Prebiotics on STZ-HFD-Induced Type 2 Diabetic Mice. Diabetes Metab Syndr Obes 2024; 17:4013-4030. [PMID: 39492963 PMCID: PMC11531242 DOI: 10.2147/dmso.s474196] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Accepted: 10/16/2024] [Indexed: 11/05/2024] Open
Abstract
Purpose This study aims to assess the therapeutic potential of combining Shen-Ling-Bai-Zhu-San (SLBZS) or prebiotics with intermittent fasting (IF) in type 2 diabetes mellitus (T2DM) mice and to investigate the synergistic effects and underlying mechanisms. Methods Type 2 diabetic mouse models were induced using high-fat diet (HFD) and streptozotocin (STZ), followed by IF treatment. Mice were then grouped for combined therapy with different doses of SLBZS and prebiotics. Fasting blood glucose (FBG) levels, body weight variations, and oral glucose tolerance tests were assessed to elucidate metabolic alterations. The hepatic and renal parameters were evaluated to determine systemic changes in T2DM mice, while the insulin levels were quantified by ELISA to assess glucose homeostasis. Gut microbiota alterations were examined via 16S rRNA sequencing. Alterations of the genes in relevant signaling pathways were analyzed using RT-qPCR. Results IF improved FBG, body weight, insulin levels, and other diabetes indicators. Combined IF with SLBZS or prebiotics yielded similar effects. Furthermore, it ameliorated dyslipidemia and mitigated hepatic and renal parameters in T2DM mice. Pancreatic tissue histopathology showed islet cell restoration post-intervention. IF therapy reduced the abnormally elevated GSK-3β gene expression and increased the abnormally reduced GLUT2 genes. Further analysis indicated that the combination of IF with prebiotics and high doses of SLBZS upregulated the expression of the INSR and IRS1 genes. Gut microbiota analysis revealed restored diversity and structure, with notable changes in specific bacterial families. At the family level, the contents of Akkermansiaceae and Bifidobacteriaceae were restored. Phylogenetic investigation of communities by reconstruction of unobserved states (PICRUSt2) analysis suggested metabolic pathway alterations. Conclusion IF improved type 2 diabetic symptoms, with combined SLBZS and prebiotics showing similar effects. IF with high concentration of SLBZS and prebiotics doses upregulated the INSR and IRS1 genes and had superior effects on gut microbiota compared to IF alone.
Collapse
Affiliation(s)
- Xiaoyu Liu
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, People’s Republic of China
| | - Pengyun Du
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, People’s Republic of China
| | - Jianing Xu
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, People’s Republic of China
| | - Wei Wang
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, People’s Republic of China
| | - Chenggang Zhang
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, People’s Republic of China
| |
Collapse
|
5
|
Xu J, Xie L, Fan R, Shi X, Xu W, Dong K, Ma D, Yan Y, Zhang S, Sun N, Huang G, Gao M, Yu X, Wang M, Wang F, Chen J, Tao J, Yang Y. The role of dietary inflammatory index in metabolic diseases: the associations, mechanisms, and treatments. Eur J Clin Nutr 2024:10.1038/s41430-024-01525-6. [PMID: 39433856 DOI: 10.1038/s41430-024-01525-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 10/08/2024] [Accepted: 10/10/2024] [Indexed: 10/23/2024]
Abstract
In recent years, the prevalence of metabolic diseases has increased significantly, posing a serious threat to global health. Chronic low-grade inflammation is implicated in the development of most metabolic diseases, such as type 2 diabetes mellitus (T2DM), obesity, dyslipidemia, and cardiovascular disease, serving as a link between diet and these conditions. Increasing attention has been directly toward dietary inflammatory patterns that may prevent or ameliorate metabolic diseases. The Dietary Inflammatory Index (DII) was developed to assess the inflammatory potential of dietary intake. Consequently, a growing body of research has examined the associations between the DII and the risk of several metabolic diseases. In this review, we explore the current scientific literature on the relationships between the DII, T2DM, obesity, and dyslipidemia. It summarizes recent findings and explore potential underlying mechanisms from two aspects: the interaction between diet and inflammation, and the link between inflammation and metabolic diseases. Furthermore, this review discusses the therapeutic strategies, including dietary modifications, prebiotics, and probiotics, and discusses the application of the DII in metabolic diseases, as well as future research directions.
Collapse
Affiliation(s)
- Jialu Xu
- Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Branch of National Clinical Research Center for Metabolic Diseases, Wuhan, China
| | - Lei Xie
- Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Branch of National Clinical Research Center for Metabolic Diseases, Wuhan, China
| | - Rongping Fan
- Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Branch of National Clinical Research Center for Metabolic Diseases, Wuhan, China
| | - Xiaoli Shi
- Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Branch of National Clinical Research Center for Metabolic Diseases, Wuhan, China
| | - Weijie Xu
- Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Branch of National Clinical Research Center for Metabolic Diseases, Wuhan, China
| | - Kun Dong
- Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Branch of National Clinical Research Center for Metabolic Diseases, Wuhan, China
| | - Delin Ma
- Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Branch of National Clinical Research Center for Metabolic Diseases, Wuhan, China
| | - Yongli Yan
- Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Branch of National Clinical Research Center for Metabolic Diseases, Wuhan, China
| | - Shujun Zhang
- Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Branch of National Clinical Research Center for Metabolic Diseases, Wuhan, China
| | - Nan Sun
- Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Branch of National Clinical Research Center for Metabolic Diseases, Wuhan, China
- Department of Nursing, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Guomin Huang
- Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Branch of National Clinical Research Center for Metabolic Diseases, Wuhan, China
- Department of Nursing, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Min Gao
- Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Branch of National Clinical Research Center for Metabolic Diseases, Wuhan, China
- Department of Nursing, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xuefeng Yu
- Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Branch of National Clinical Research Center for Metabolic Diseases, Wuhan, China
| | - Mei Wang
- Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Branch of National Clinical Research Center for Metabolic Diseases, Wuhan, China
| | - Fen Wang
- Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Branch of National Clinical Research Center for Metabolic Diseases, Wuhan, China
| | - Juan Chen
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Jing Tao
- Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- Branch of National Clinical Research Center for Metabolic Diseases, Wuhan, China.
- Department of Nursing, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Yan Yang
- Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- Branch of National Clinical Research Center for Metabolic Diseases, Wuhan, China.
| |
Collapse
|
6
|
Wang J, Wei HJ, Mao RF, Chang X. Gut microbiota modulating therapy for diabetes mellitus should be individualized. World J Diabetes 2024; 15:2152-2156. [PMID: 39493555 PMCID: PMC11525721 DOI: 10.4239/wjd.v15.i10.2152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 09/14/2024] [Accepted: 09/18/2024] [Indexed: 09/26/2024] Open
Abstract
In this editorial, we commented on two articles published online in August and September 2024 in the World Journal of Diabetes, which focused on modifying the gut microbiota (GM) to prevent or delay the progression of diabetes mellitus (DM) and DM-related complications. Numerous studies, many of which are animal studies, have indicated the potential role of GM in the pathogenesis of DM. However, the detailed causality and mechanisms between GM and DM have not been fully clarified. Although there have been some reports of a potential role of modifying the GM in treating DM, most lack long-term observations and are not mechanistic. Additionally, the GM and its role in DM might vary among individuals; therefore, GM-targeted interventions should be individualized to realize their therapeutic potential.
Collapse
Affiliation(s)
- Jin Wang
- Department of Ultrasound Medicine, Nanjing Lishui People’s Hospital, Zhongda Hospital Lishui Branch, Southeast University, Nanjing 211200, Jiangsu Province, China
| | - Hong-Juan Wei
- Neonatal Intensive Care Unit, Nanjing Lishui People’s Hospital, Zhongda Hospital Lishui Branch, Southeast University, Nanjing 211200, Jiangsu Province, China
| | - Rui-Feng Mao
- School of Life Science, Huaiyin Normal University, Huai'an 223300, Jiangsu Province, China
| | - Xin Chang
- Department of Ultrasound Medicine, Nanjing Lishui People’s Hospital, Zhongda Hospital Lishui Branch, Southeast University, Nanjing 211200, Jiangsu Province, China
| |
Collapse
|
7
|
Iatcu OC, Hamamah S, Covasa M. Harnessing Prebiotics to Improve Type 2 Diabetes Outcomes. Nutrients 2024; 16:3447. [PMID: 39458444 PMCID: PMC11510484 DOI: 10.3390/nu16203447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 10/08/2024] [Accepted: 10/09/2024] [Indexed: 10/28/2024] Open
Abstract
The gut microbiota, a complex ecosystem of microorganisms in the human gastrointestinal tract (GI), plays a crucial role in maintaining metabolic health and influencing disease susceptibility. Dysbiosis, or an imbalance in gut microbiota, has been linked to the development of type 2 diabetes mellitus (T2DM) through mechanisms such as reduced glucose tolerance and increased insulin resistance. A balanced gut microbiota, or eubiosis, is associated with improved glucose metabolism and insulin sensitivity, potentially reducing the risk of diabetes-related complications. Various strategies, including the use of prebiotics like inulin, fructooligosaccharides, galactooligosaccharides, resistant starch, pectic oligosaccharides, polyphenols, β-glucan, and Dendrobium officinale have been shown to improve gut microbial composition and support glycemic control in T2DM patients. These prebiotics can directly impact blood sugar levels while promoting the growth of beneficial bacteria, thus enhancing glycemic control. Studies have shown that T2DM patients often exhibit a decrease in beneficial butyrate-producing bacteria, like Roseburia and Faecalibacterium, and an increase in harmful bacteria, such as Escherichia and Prevotella. This review aims to explore the effects of different prebiotics on T2DM, their impact on gut microbiota composition, and the potential for personalized dietary interventions to optimize diabetes management and improve overall health outcomes.
Collapse
Affiliation(s)
- Oana C. Iatcu
- Department of Biomedical Sciences, College of Medicine and Biological Science, University of Suceava, 720229 Suceava, Romania;
| | - Sevag Hamamah
- Department of Basic Medical Sciences, College of Osteopathic Medicine, Western University of Health Sciences, Pomona, CA 91766, USA;
- Department of Internal Medicine, Scripps Mercy Hospital, San Diego, CA 92103, USA
| | - Mihai Covasa
- Department of Biomedical Sciences, College of Medicine and Biological Science, University of Suceava, 720229 Suceava, Romania;
- Department of Basic Medical Sciences, College of Osteopathic Medicine, Western University of Health Sciences, Pomona, CA 91766, USA;
| |
Collapse
|
8
|
Samulėnaitė S, García-Blanco A, Mayneris-Perxachs J, Domingo-Rodríguez L, Cabana-Domínguez J, Fernàndez-Castillo N, Gago-García E, Pineda-Cirera L, Burokas A, Espinosa-Carrasco J, Arboleya S, Latorre J, Stanton C, Hosomi K, Kunisawa J, Cormand B, Fernández-Real JM, Maldonado R, Martín-García E. Gut microbiota signatures of vulnerability to food addiction in mice and humans. Gut 2024; 73:1799-1815. [PMID: 38926079 PMCID: PMC11503113 DOI: 10.1136/gutjnl-2023-331445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 04/01/2024] [Indexed: 06/28/2024]
Abstract
OBJECTIVE Food addiction is a multifactorial disorder characterised by a loss of control over food intake that may promote obesity and alter gut microbiota composition. We have investigated the potential involvement of the gut microbiota in the mechanisms underlying food addiction. DESIGN We used the Yale Food Addiction Scale (YFAS) 2.0 criteria to classify extreme food addiction in mouse and human subpopulations to identify gut microbiota signatures associated with vulnerability to this disorder. RESULTS Both animal and human cohorts showed important similarities in the gut microbiota signatures linked to food addiction. The signatures suggested possible non-beneficial effects of bacteria belonging to the Proteobacteria phylum and potential protective effects of Actinobacteria against the development of food addiction in both cohorts of humans and mice. A decreased relative abundance of the species Blautia wexlerae was observed in addicted humans and of Blautia genus in addicted mice. Administration of the non-digestible carbohydrates, lactulose and rhamnose, known to favour Blautia growth, led to increased relative abundance of Blautia in mice faeces in parallel with dramatic improvements in food addiction. A similar improvement was revealed after oral administration of Blautia wexlerae as a beneficial microbe. CONCLUSION By understanding the crosstalk between this behavioural alteration and gut microbiota, these findings constitute a step forward to future treatments for food addiction and related eating disorders.
Collapse
Affiliation(s)
- Solveiga Samulėnaitė
- Laboratory of Neuropharmacology-Neurophar, Department of Medicine and Life Sciences, Pompeu Fabra University, Barcelona, Spain
- Department of Biological Models, Institute of Biochemistry, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Alejandra García-Blanco
- Laboratory of Neuropharmacology-Neurophar, Department of Medicine and Life Sciences, Pompeu Fabra University, Barcelona, Spain
| | - Jordi Mayneris-Perxachs
- Nutrition, Eumetabolism and Health Group, Girona Biomedical Research Institute (IdibGi), Girona, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, Madrid, Spain
- Department of Diabetes, Endocrinology and Nutrition, Dr Josep Trueta University Hospital, Girona, Spain
| | - Laura Domingo-Rodríguez
- Laboratory of Neuropharmacology-Neurophar, Department of Medicine and Life Sciences, Pompeu Fabra University, Barcelona, Spain
| | - Judit Cabana-Domínguez
- Departament de Genètica, Microbiologia i Estadística, Facultat de Biologia, Universitat de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras, (CIBERER), Madrid, Spain
- Institut de Biomedicina de la Universitat de Barcelona, (IBUB), Barcelona, Spain
- Institut de Recerca Sant Joan de Déu (IR-SJD), Esplugues de Llobregat, Barcelona, Spain
| | - Noèlia Fernàndez-Castillo
- Departament de Genètica, Microbiologia i Estadística, Facultat de Biologia, Universitat de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras, (CIBERER), Madrid, Spain
- Institut de Biomedicina de la Universitat de Barcelona, (IBUB), Barcelona, Spain
- Institut de Recerca Sant Joan de Déu (IR-SJD), Esplugues de Llobregat, Barcelona, Spain
| | - Edurne Gago-García
- Departament de Genètica, Microbiologia i Estadística, Facultat de Biologia, Universitat de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras, (CIBERER), Madrid, Spain
- Institut de Biomedicina de la Universitat de Barcelona, (IBUB), Barcelona, Spain
- Institut de Recerca Sant Joan de Déu (IR-SJD), Esplugues de Llobregat, Barcelona, Spain
| | - Laura Pineda-Cirera
- Departament de Genètica, Microbiologia i Estadística, Facultat de Biologia, Universitat de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras, (CIBERER), Madrid, Spain
- Institut de Biomedicina de la Universitat de Barcelona, (IBUB), Barcelona, Spain
- Institut de Recerca Sant Joan de Déu (IR-SJD), Esplugues de Llobregat, Barcelona, Spain
| | - Aurelijus Burokas
- Department of Biological Models, Institute of Biochemistry, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | | | - Silvia Arboleya
- APC Microbiome Institute, University College Cork, Cork, Ireland
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias, Consejo Superior de Investigaciones Científicas (IPLA-CSIC), Villaviciosa, Asturias, Spain
| | - Jessica Latorre
- Nutrition, Eumetabolism and Health Group, Girona Biomedical Research Institute (IdibGi), Girona, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, Madrid, Spain
- Department of Diabetes, Endocrinology and Nutrition, Dr Josep Trueta University Hospital, Girona, Spain
| | - Catherine Stanton
- APC Microbiome Institute, University College Cork, Cork, Ireland
- Teagasc Food Research Centre, Moorepark, Fermoy, Co, Cork, Ireland
| | - Koji Hosomi
- Laboratory of Vaccine Materials and Laboratory of Gut Environmental System, Microbial Research Center for Health and Medicine, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Ibaraki, Osaka, Japan. (NIBIOHN), Ibaraki, Osaka, Japan
| | - Jun Kunisawa
- Laboratory of Vaccine Materials and Laboratory of Gut Environmental System, Microbial Research Center for Health and Medicine, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Ibaraki, Osaka, Japan. (NIBIOHN), Ibaraki, Osaka, Japan
| | - Bru Cormand
- Departament de Genètica, Microbiologia i Estadística, Facultat de Biologia, Universitat de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras, (CIBERER), Madrid, Spain
- Institut de Biomedicina de la Universitat de Barcelona, (IBUB), Barcelona, Spain
- Institut de Recerca Sant Joan de Déu (IR-SJD), Esplugues de Llobregat, Barcelona, Spain
| | - Jose Manuel Fernández-Real
- Nutrition, Eumetabolism and Health Group, Girona Biomedical Research Institute (IdibGi), Girona, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, Madrid, Spain
- Department of Diabetes, Endocrinology and Nutrition, Dr Josep Trueta University Hospital, Girona, Spain
- Department of Medical Sciences, Faculty of Medicine, University of Girona, Girona, Spain
| | - Rafael Maldonado
- Laboratory of Neuropharmacology-Neurophar, Department of Medicine and Life Sciences, Pompeu Fabra University, Barcelona, Spain
- Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain
| | - Elena Martín-García
- Laboratory of Neuropharmacology-Neurophar, Department of Medicine and Life Sciences, Pompeu Fabra University, Barcelona, Spain
- Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain
- Departament de Psicobiologia i Metodologia de les Ciències de la Salut, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Barcelona, Spain
- Institut de Neurociències, Universitat Autònoma de Barcelona, Barcelona, Spain
| |
Collapse
|
9
|
Shi K, Huang K, Li L, Liu Q, Zhang Y, Zheng H. Predicting microbe-disease association based on graph autoencoder and inductive matrix completion with multi-similarities fusion. Front Microbiol 2024; 15:1438942. [PMID: 39355422 PMCID: PMC11443509 DOI: 10.3389/fmicb.2024.1438942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 08/02/2024] [Indexed: 10/03/2024] Open
Abstract
Background Clinical studies have demonstrated that microbes play a crucial role in human health and disease. The identification of microbe-disease interactions can provide insights into the pathogenesis and promote the diagnosis, treatment, and prevention of disease. Although a large number of computational methods are designed to screen novel microbe-disease associations, the accurate and efficient methods are still lacking due to data inconsistence, underutilization of prior information, and model performance. Methods In this study, we proposed an improved deep learning-based framework, named GIMMDA, to identify latent microbe-disease associations, which is based on graph autoencoder and inductive matrix completion. By co-training the information from microbe and disease space, the new representations of microbes and diseases are used to reconstruct microbe-disease association in the end-to-end framework. In particular, a similarity fusion strategy is conducted to improve prediction performance. Results The experimental results show that the performance of GIMMDA is competitive with that of existing state-of-the-art methods on 3 datasets (i.e., HMDAD, Disbiome, and multiMDA). In particular, it performs best with the area under the receiver operating characteristic curve (AUC) of 0.9735, 0.9156, 0.9396 on abovementioned 3 datasets, respectively. And the result also confirms that different similarity fusions can improve the prediction performance. Furthermore, case studies on two diseases, i.e., asthma and obesity, validate the effectiveness and reliability of our proposed model. Conclusion The proposed GIMMDA model show a strong capability in predicting microbe-disease associations. We expect that GPUDMDA will help identify potential microbe-related diseases in the future.
Collapse
Affiliation(s)
- Kai Shi
- College of Computer Science and Engineering, Guilin University of Technology, Guilin, China
- Guangxi Key Laboratory of Embedded Technology and Intelligent Systems, Guilin University of Technology, Guilin, China
| | - Kai Huang
- College of Computer Science and Engineering, Guilin University of Technology, Guilin, China
| | - Lin Li
- College of Computer Science and Engineering, Guilin University of Technology, Guilin, China
| | - Qiaohui Liu
- College of Computer Science and Engineering, Guilin University of Technology, Guilin, China
| | - Yi Zhang
- College of Computer Science and Engineering, Guilin University of Technology, Guilin, China
| | - Huilin Zheng
- College of Computer Science and Engineering, Guilin University of Technology, Guilin, China
| |
Collapse
|
10
|
Nikdasti A, Khodadadi ES, Ferdosi F, Dadgostar E, Yahyazadeh S, Heidari P, Ehtiati S, Vakili O, Khatami SH. Nutritional Strategies in Major Depression Disorder: From Ketogenic Diet to Modulation of the Microbiota-Gut-Brain Axis. Mol Neurobiol 2024:10.1007/s12035-024-04446-4. [PMID: 39192045 DOI: 10.1007/s12035-024-04446-4] [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: 03/31/2024] [Accepted: 08/15/2024] [Indexed: 08/29/2024]
Abstract
Major depressive disorder (MDD) is a leading cause of disability worldwide. While traditional pharmacological treatments are effective for many cases, a significant proportion of patients do not achieve full remission or experience side effects. Nutritional interventions hold promise as an alternative or adjunctive approach, especially for treatment-resistant depression. This review examines the potential role of nutrition in managing MDD through addressing biological deficits and modulating pathways relevant to its pathophysiology. Specifically, it explores the ketogenic diet and gut microbiome modulation through various methods, including probiotics, prebiotics, synbiotics, postbiotics, and fecal microbiota transplantation. Numerous studies link dietary inadequacies to increased MDD risk and deficiencies in nutrients like omega-3 s, vitamins D and B, magnesium, and zinc. These deficiencies impact neurotransmitters, inflammation, and other biological factors in MDD. The gut-brain axis also regulates mood, stress response, and immunity, and disruptions are implicated in MDD. While medications aid acute symptoms, nutritional strategies may improve long-term outcomes by preventing relapse and promoting sustained remission. This comprehensive review aims to provide insights into nutrition's multifaceted relationship with MDD and its potential for developing more effective integrated treatment approaches.
Collapse
Affiliation(s)
- Ali Nikdasti
- Department of Comparative Biomedicine and Food Science, University of Padova, Padova, Italy
| | - Elaheh Sadat Khodadadi
- Department of Comparative Biomedicine and Food Science, University of Padova, Padova, Italy
| | - Felora Ferdosi
- Department of Radiology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Ehsan Dadgostar
- Behavioral Sciences Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
- Student Research Committee, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Sheida Yahyazadeh
- Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Parasta Heidari
- School of Medicine, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Sajad Ehtiati
- Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Omid Vakili
- Department of Clinical Biochemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran.
| | - Seyyed Hossein Khatami
- Student Research Committee, Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
11
|
Beteri B, Barone M, Turroni S, Brigidi P, Tzortzis G, Vulevic J, Sekulic K, Motei DE, Costabile A. Impact of Combined Prebiotic Galacto-Oligosaccharides and Bifidobacterium breve-Derived Postbiotic on Gut Microbiota and HbA1c in Prediabetic Adults: A Double-Blind, Randomized, Placebo-Controlled Study. Nutrients 2024; 16:2205. [PMID: 39064648 PMCID: PMC11280236 DOI: 10.3390/nu16142205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 07/03/2024] [Accepted: 07/07/2024] [Indexed: 07/28/2024] Open
Abstract
The complex interactions between intestinal microbiota and metabolic disorders are well-documented, with implications for glucose metabolism, energy expenditure, and intestinal permeability. Prebiotics induce beneficial changes in gut microbiota composition in prediabetes, while postbiotics can enhance gut barrier function, complementing each other to improve glucose metabolism and insulin sensitivity. This study investigated the effects of a 12-week dietary fibre (DF) supplement on gut health, metabolic function, and diet. The supplement contained konjac glucomannan (KGM), galacto-oligosaccharides (GOSs), and exopolysaccharides (EPSs) from Bifidobacterium breve. In a randomised, double-blind, placebo-controlled, parallel-group clinical trial, 53 prediabetic volunteers were randomly assigned to either a daily DF supplement (YMETA) or a placebo (cellulose microcrystalline) for 12 weeks, followed by a 4-week follow-up. Measurements included gut microbiota composition, glycated haemoglobin (HbA1c), fasting plasma glucose (FPG), plasma lipids, anthropometry, body composition, blood pressure, and dietary intake. The intervention group showed a significant increase in alpha diversity and butyrate-producing bacteria, with reductions in HbA1c and FPG levels below prediabetes thresholds. No significant changes were observed in the placebo group. This study suggests that manipulating the human gut microbiome through dietary interventions could be a promising therapeutic approach to managing prediabetes and preventing or delaying diabetes.
Collapse
Affiliation(s)
- Beyda Beteri
- School of Life and Health Sciences, University of Roehampton, London SW15 4JD, UK; (B.B.); (D.-E.M.)
| | - Monica Barone
- Human Microbiomics Unit, Department of Medical and Surgical Sciences, University of Bologna, 40138 Bologna, Italy; (M.B.); (P.B.)
| | - Silvia Turroni
- Unit of Microbiome Science and Biotechnology, Department of Pharmacy and Biotechnology, University of Bologna, 40126 Bologna, Italy;
| | - Patrizia Brigidi
- Human Microbiomics Unit, Department of Medical and Surgical Sciences, University of Bologna, 40138 Bologna, Italy; (M.B.); (P.B.)
| | - George Tzortzis
- veMico Ltd., Amelia House, Crescent Road, Worthing BN11 1RL, UK; (G.T.); (J.V.)
| | - Jelena Vulevic
- veMico Ltd., Amelia House, Crescent Road, Worthing BN11 1RL, UK; (G.T.); (J.V.)
| | - Karol Sekulic
- Alberta Health Services, Edmonton, AB T5J 3E4, Canada;
| | - Diana-Elena Motei
- School of Life and Health Sciences, University of Roehampton, London SW15 4JD, UK; (B.B.); (D.-E.M.)
| | - Adele Costabile
- School of Life and Health Sciences, University of Roehampton, London SW15 4JD, UK; (B.B.); (D.-E.M.)
| |
Collapse
|
12
|
Park M, Lee HB, Kim HR, Kang MC, Jeong D, Choi HD, Hong JS, Park HY. Resistant starch-enriched brown rice exhibits prebiotic properties and enhances gut health in obese mice. Food Res Int 2024; 187:114417. [PMID: 38763667 DOI: 10.1016/j.foodres.2024.114417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 04/20/2024] [Accepted: 04/22/2024] [Indexed: 05/21/2024]
Abstract
Resistant starch serves as a prebiotic in the large intestine, aiding in the maintenance of a healthy intestinal environment and mitigating associated chronic illnesses. This study aimed to investigate the impact of resistant starch-enriched brown rice (RBR) on intestinal health and functionality. We assessed changes in resistant starch concentration, structural alterations, and branch chain length distribution throughout the digestion process using an in vitro model. The efficacy of RBR in the intestinal environment was evaluated through analyses of its prebiotic potential, effects on intestinal microbiota, and intestinal function-related proteins in obese animals fed a high-fat diet. RBR exhibited a higher yield of insoluble fraction in both the small and large intestines compared to white and brown rice. The total digestible starch content decreased, while the resistant starch content significantly increased during in vitro digestion. Furthermore, RBR notably enhanced the growth of four probiotic strains compared to white and brown rice, displaying higher proliferation activity than the positive control, FOS. Notably, consumption of RBR by high-fat diet-induced obese mice suppressed colon shortening, increased Bifidobacteria growth, and improved intestinal permeability. These findings underscore the potential prebiotic and gut health-promoting attributes of RBR, offering insights for the development of functional foods aimed at preventing gastrointestinal diseases.
Collapse
Affiliation(s)
- Miri Park
- Food Functionality Research Division, Korea Food Research Institute, Wanju-gun 55365, Republic of Korea
| | - Hye-Bin Lee
- Food Functionality Research Division, Korea Food Research Institute, Wanju-gun 55365, Republic of Korea
| | - Ha Ram Kim
- Food Convergence Research Division, Korea Food Research Institute, Wanju-gun 55365, Republic of Korea
| | - Min-Cheol Kang
- Food Convergence Research Division, Korea Food Research Institute, Wanju-gun 55365, Republic of Korea
| | - Duyun Jeong
- Department of Food and Food Service Industry, Kyungpook National University, Sangju 37224, Republic of Korea
| | - Hee-Don Choi
- Food Convergence Research Division, Korea Food Research Institute, Wanju-gun 55365, Republic of Korea
| | - Jung Sun Hong
- Food Convergence Research Division, Korea Food Research Institute, Wanju-gun 55365, Republic of Korea
| | - Ho-Young Park
- Food Functionality Research Division, Korea Food Research Institute, Wanju-gun 55365, Republic of Korea; Department of Food Biotechnology, University of Science and Technology, Daejeon 34113, Republic of Korea.
| |
Collapse
|
13
|
Bock PM, Martins AF, Schaan BD. Understanding how pre- and probiotics affect the gut microbiome and metabolic health. Am J Physiol Endocrinol Metab 2024; 327:E89-E102. [PMID: 38809510 DOI: 10.1152/ajpendo.00054.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 05/22/2024] [Accepted: 05/23/2024] [Indexed: 05/30/2024]
Abstract
The gut microbiome, a complex assembly of microorganisms, significantly impacts human health by influencing nutrient absorption, the immune system, and disease response. These microorganisms form a dynamic ecosystem that is critical to maintaining overall well-being. Prebiotics and probiotics are pivotal in regulating gut microbiota composition. Prebiotics nourish beneficial bacteria and promote their growth, whereas probiotics help maintain balance within the microbiome. This intricate balance extends to several aspects of health, including maintaining the integrity of the gut barrier, regulating immune responses, and producing metabolites crucial for metabolic health. Dysbiosis, or an imbalance in the gut microbiota, has been linked to metabolic disorders such as type 2 diabetes, obesity, and cardiovascular disease. Impaired gut barrier function, endotoxemia, and low-grade inflammation are associated with toll-like receptors influencing proinflammatory pathways. Short-chain fatty acids derived from microbial fermentation modulate anti-inflammatory and immune system pathways. Prebiotics positively influence gut microbiota, whereas probiotics, especially Lactobacillus and Bifidobacterium strains, may improve metabolic outcomes, such as glycemic control in diabetes. It is important to consider strain-specific effects and study variability when interpreting these findings, highlighting the need for further research to optimize their therapeutic potential. The aim of this report is therefore to review the role of the gut microbiota in metabolic health and disease and the effects of prebiotics and probiotics on the gut microbiome and their therapeutic role, integrating a broad understanding of physiological mechanisms with a clinical perspective.
Collapse
Affiliation(s)
- Patricia M Bock
- Pharmacology, Institute of Basic Science, Universidade Federal do Rio Grande, Rio Grande, Brazil
| | - Andreza F Martins
- Microbiology, Department of Microbiology, Immunology, and Parasitology, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
- Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Beatriz D Schaan
- Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
- Faculty of Medicine, Department of Internal Medicine, Graduate Program in Medical Sciences: Endocrinology, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| |
Collapse
|
14
|
Basuray N, Deehan EC, Vieira FT, Avedzi HM, Duke RL, Colín-Ramírez E, Tun HM, Zhang Z, Wine E, Madsen KL, Field CJ, Haqq AM. Dichotomous effect of dietary fiber in pediatrics: a narrative review of the health benefits and tolerance of fiber. Eur J Clin Nutr 2024; 78:557-568. [PMID: 38480843 DOI: 10.1038/s41430-024-01429-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Revised: 02/27/2024] [Accepted: 03/01/2024] [Indexed: 03/18/2024]
Abstract
Dietary fibers are associated with favorable gastrointestinal, immune, and metabolic health outcomes when consumed at sufficient levels. Despite the well-described benefits of dietary fibers, children and adolescents continue to fall short of daily recommended levels. This gap in fiber intake (i.e., "fiber gap") might increase the risk of developing early-onset pediatric obesity and obesity-related comorbidities such as type 2 diabetes mellitus into adulthood. The structure-dependent physicochemical properties of dietary fiber are diverse. Differences in solubility, viscosity, water-holding capacity, binding capability, bulking effect, and fermentability influence the physiological effects of dietary fibers that aid in regulating appetite, glycemic and lipidemic responses, and inflammation. Of growing interest is the fermentation of fibers by the gut microbiota, which yields both beneficial and less favorable end-products such as short-chain fatty acids (e.g., acetate, propionate, and butyrate) that impart metabolic and immunomodulatory properties, and gases (e.g., hydrogen, carbon dioxide, and methane) that cause gastrointestinal symptoms, respectively. This narrative review summarizes (1) the implications of fibers on the gut microbiota and the pathophysiology of pediatric obesity, (2) some factors that potentially contribute to the fiber gap with an emphasis on undesirable gastrointestinal symptoms, (3) some methods to alleviate fiber-induced symptoms, and (4) the therapeutic potential of whole foods and commonly marketed fiber supplements for improved health in pediatric obesity.
Collapse
Affiliation(s)
- Nandini Basuray
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB, Canada
| | - Edward C Deehan
- Department of Food Science and Technology, University of Nebraska, Lincoln, NE, USA
- Nebraska Food for Health Center, Lincoln, NE, USA
| | - Flávio T Vieira
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB, Canada
- Department of Pediatrics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Hayford M Avedzi
- Department of Pediatrics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Reena L Duke
- Department of Pediatrics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | | | - Hein M Tun
- JC School of Public Health and Primary Care, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, SAR, China
| | - Zhengxiao Zhang
- College of Ocean Food and Biological Engineering, Jimei University, Xiamen, Fujian, China
| | - Eytan Wine
- Department of Pediatrics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Karen L Madsen
- Department of Medicine, University of Alberta, Edmonton, AB, Canada
| | - Catherine J Field
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB, Canada
| | - Andrea M Haqq
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB, Canada.
- Department of Pediatrics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada.
| |
Collapse
|
15
|
Bavaro AR, Di Biase M, Linsalata V, D’Antuono I, Di Stefano V, Lonigro SL, Garbetta A, Valerio F, Melilli MG, Cardinali A. Potential Prebiotic Effect of Inulin-Enriched Pasta after In Vitro Gastrointestinal Digestion and Simulated Gut Fermentation. Foods 2024; 13:1815. [PMID: 38928756 PMCID: PMC11202534 DOI: 10.3390/foods13121815] [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: 04/22/2024] [Revised: 05/26/2024] [Accepted: 06/06/2024] [Indexed: 06/28/2024] Open
Abstract
In the current study, the prebiotic potential of an innovative functional pasta enriched with 12% (w/w) inulin was investigated. To this aim, pasta was subjected to in vitro gastrointestinal digestion followed by simulated gut fermentation compared to the control pasta (CTRL) not containing inulin. The incorporation of inulin positively (p < 0.05) affected some organoleptic traits and the cooking quality of the final product, giving an overall score significantly higher than CTRL. The resultant essential amino acid content was similar in both pasta samples while the total protein content was lower in inulin-enriched pasta for the polymer substitution to durum wheat flour. The prebiotic potential of chicory inulin was preliminarily tested in in vitro experiments using seven probiotic strains and among them Lacticaseibacillus paracasei IMPC2.1 was selected for the simulated gut fermentation studies. The positive prebiotic activity score registered with the probiotic strain suggested the suitability of the inulin-enriched pasta with respect to acting as a prebiotic source favoring the growth of the probiotic strain and short chain fatty acid (SCFA) production. The present study contributes to broadening knowledge on the prebiotic efficacy of inulin when incorporated into a complex food matrix.
Collapse
Affiliation(s)
- Anna Rita Bavaro
- Institute of Sciences of Food Productions (ISPA), National Research Council (CNR), 70126 Bari, Italy; (A.R.B.); (M.D.B.); (V.L.); (I.D.); (S.L.L.); (A.G.); (A.C.)
| | - Mariaelena Di Biase
- Institute of Sciences of Food Productions (ISPA), National Research Council (CNR), 70126 Bari, Italy; (A.R.B.); (M.D.B.); (V.L.); (I.D.); (S.L.L.); (A.G.); (A.C.)
| | - Vito Linsalata
- Institute of Sciences of Food Productions (ISPA), National Research Council (CNR), 70126 Bari, Italy; (A.R.B.); (M.D.B.); (V.L.); (I.D.); (S.L.L.); (A.G.); (A.C.)
| | - Isabella D’Antuono
- Institute of Sciences of Food Productions (ISPA), National Research Council (CNR), 70126 Bari, Italy; (A.R.B.); (M.D.B.); (V.L.); (I.D.); (S.L.L.); (A.G.); (A.C.)
| | - Vita Di Stefano
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, 90123 Palermo, Italy;
| | - Stella Lisa Lonigro
- Institute of Sciences of Food Productions (ISPA), National Research Council (CNR), 70126 Bari, Italy; (A.R.B.); (M.D.B.); (V.L.); (I.D.); (S.L.L.); (A.G.); (A.C.)
| | - Antonella Garbetta
- Institute of Sciences of Food Productions (ISPA), National Research Council (CNR), 70126 Bari, Italy; (A.R.B.); (M.D.B.); (V.L.); (I.D.); (S.L.L.); (A.G.); (A.C.)
| | - Francesca Valerio
- Institute of Sciences of Food Productions (ISPA), National Research Council (CNR), 70126 Bari, Italy; (A.R.B.); (M.D.B.); (V.L.); (I.D.); (S.L.L.); (A.G.); (A.C.)
| | - Maria Grazia Melilli
- Institute of Biomolecular Chemistry (ICB), National Research Council (CNR), 95126 Catania, Italy
| | - Angela Cardinali
- Institute of Sciences of Food Productions (ISPA), National Research Council (CNR), 70126 Bari, Italy; (A.R.B.); (M.D.B.); (V.L.); (I.D.); (S.L.L.); (A.G.); (A.C.)
| |
Collapse
|
16
|
Xu YW, Tian J, Song Y, Zhang BC, Wang J. Metabolic syndrome's new therapy: Supplement the gut microbiome. World J Diabetes 2024; 15:793-796. [PMID: 38680700 PMCID: PMC11045428 DOI: 10.4239/wjd.v15.i4.793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 01/18/2024] [Accepted: 02/27/2024] [Indexed: 04/11/2024] Open
Abstract
This letter to the editor discusses the publication on gut microbiome supplementation as therapy for metabolic syndrome. Gut microbiome dysbiosis disrupts intestinal bacterial homeostasis and is related to chronic inflammation, insulin resistance, cardiovascular diseases, type 2 diabetes mellitus, and obesity. Previous research has found that increasing the abundance of beneficial microbiota in the gut modulates metabolic syndrome by reducing chronic inflammation and insulin resistance. Prebiotics, probiotics, synbiotics, and postbiotics are often used as supplements to increase the number of beneficial microbes and thus the production of short-chain fatty acids, which have positive effects on the gut microbiome and metabolic syndrome. In this review article, the author summarizes the available supplements to increase the abundance of beneficial gut microbiota and reduce the abundance of harmful microbiota in patients with metabolic disorders. Our group is also researching the role of the gut microbiota in chronic liver disease. This article will be of great help to our research. At the end of the letter, the mechanism of the gut microbiota in chronic liver disease is discussed.
Collapse
Affiliation(s)
- Yong-Wei Xu
- Department of Gastroenterology, Songjiang Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 201600, China
| | - Jun Tian
- Department of Gastroenterology, Songjiang Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 201600, China
| | - Yan Song
- Department of Gastroenterology, Songjiang Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 201600, China
| | - Ba-Cui Zhang
- Department of Gastroenterology, Songjiang Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 201600, China
| | - Jing Wang
- Department of Gastroenterology, Songjiang Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 201600, China
| |
Collapse
|
17
|
Gościniak A, Rosiak N, Szymanowska D, Miklaszewski A, Cielecka-Piontek J. Prebiotic Systems Containing Anthocyanin-Rich Pomegranate Flower Extracts with Antioxidant and Antidiabetic Effects. Pharmaceutics 2024; 16:526. [PMID: 38675187 PMCID: PMC11054034 DOI: 10.3390/pharmaceutics16040526] [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: 03/04/2024] [Revised: 04/04/2024] [Accepted: 04/07/2024] [Indexed: 04/28/2024] Open
Abstract
Pomegranate flower extract, rich in anthocyanins, demonstrates beneficial health-promoting properties such as an anti-diabetic and antioxidant effect, among others. However, the potential health-promoting properties may be hindered by the low stability of anthocyanins. Therefore, the aim of our study was to assess whether stabilizing carriers, namely HP-γ-cyclodextrin (HP-γ-CD), α-cyclodextrin (α-CD), Methyl-β-cyclodextrin (Me-β-CD), Inulin (Inu) and Arabic gum (AGu) affect the antioxidant and antidiabetic activity of lyophilized pomegranate flower extract, how they influence stability, release profile, and whether the systems exhibit prebiotic activity. Interactions between pomegranate flower extract and these factors were analyzed using FT-IR. The structures were examined through microscopic imaging while for the prepared prebiotic systems, antidiabetic activity was determined and confirmed by the inhibition of α-amylase and α-glucosidase; antioxidant activity was expressed by DPPH and CUPRAC assays. The content of pelargonidin-3,5-glucoside in these systems was assessed using the HPLC method. The release profiles of pelargonidin-3,5-glucoside were examined in a medium at pH = 6.8 and pH = 1.2, and the stability was assessed after subjecting the systems to high temperatures (T = 90 °C). The prebiotic potential was evaluated for 10 prebiotic bacterial strains (Lactobacillus acidophilus, Lactobacillus casei, Lactobacillus plantarum, Lactobacillus brevis Lactobacillus rhamnosus gg, Lactobacillus reuteri, Pediococcus pentosaceus, Lactococcus lactis, Lactobacillus fermentum lf, Streptococcus thermophilus). As a result of the conducted research, better functionalities of the obtained systems containing Pomegranate flower extract were proven in terms of prebiotic and antidiabetic effects. The obtained delivery systems for pelargonidin-3,5-glucoside allow for better use of its health-promoting effects.
Collapse
Affiliation(s)
- Anna Gościniak
- Department of Pharmacognosy and Biomaterials, Poznan University of Medical Sciences, Rokietnicka 3, 60-806 Poznan, Poland; (A.G.); (N.R.); (D.S.)
| | - Natalia Rosiak
- Department of Pharmacognosy and Biomaterials, Poznan University of Medical Sciences, Rokietnicka 3, 60-806 Poznan, Poland; (A.G.); (N.R.); (D.S.)
| | - Daria Szymanowska
- Department of Pharmacognosy and Biomaterials, Poznan University of Medical Sciences, Rokietnicka 3, 60-806 Poznan, Poland; (A.G.); (N.R.); (D.S.)
- Department of Biotechnology and Food Microbiology, Poznan University of Life Sciences, Wojska Polskiego 48, 60-627 Poznan, Poland
| | - Andrzej Miklaszewski
- Institute of Materials Science and Engineering, Poznan University of Technology, Jana Pawła II 24, 61-138 Poznan, Poland;
| | - Judyta Cielecka-Piontek
- Department of Pharmacognosy and Biomaterials, Poznan University of Medical Sciences, Rokietnicka 3, 60-806 Poznan, Poland; (A.G.); (N.R.); (D.S.)
| |
Collapse
|
18
|
Manrique P, Montero I, Fernandez-Gosende M, Martinez N, Cantabrana CH, Rios-Covian D. Past, present, and future of microbiome-based therapies. MICROBIOME RESEARCH REPORTS 2024; 3:23. [PMID: 38841413 PMCID: PMC11149097 DOI: 10.20517/mrr.2023.80] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 03/07/2024] [Accepted: 03/12/2024] [Indexed: 06/07/2024]
Abstract
Technological advances in studying the human microbiome in depth have enabled the identification of microbial signatures associated with health and disease. This confirms the crucial role of microbiota in maintaining homeostasis and the host health status. Nowadays, there are several ways to modulate the microbiota composition to effectively improve host health; therefore, the development of therapeutic treatments based on the gut microbiota is experiencing rapid growth. In this review, we summarize the influence of the gut microbiota on the development of infectious disease and cancer, which are two of the main targets of microbiome-based therapies currently being developed. We analyze the two-way interaction between the gut microbiota and traditional drugs in order to emphasize the influence of gut microbial composition on drug effectivity and treatment response. We explore the different strategies currently available for modulating this ecosystem to our benefit, ranging from 1st generation intervention strategies to more complex 2nd generation microbiome-based therapies and their regulatory framework. Lastly, we finish with a quick overview of what we believe is the future of these strategies, that is 3rd generation microbiome-based therapies developed with the use of artificial intelligence (AI) algorithms.
Collapse
|
19
|
Apalowo OE, Adegoye GA, Obuotor TM. Microbial-Based Bioactive Compounds to Alleviate Inflammation in Obesity. Curr Issues Mol Biol 2024; 46:1810-1831. [PMID: 38534735 DOI: 10.3390/cimb46030119] [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: 02/07/2024] [Revised: 02/20/2024] [Accepted: 02/23/2024] [Indexed: 03/28/2024] Open
Abstract
The increased prevalence of obesity with several other metabolic disorders, including diabetes and non-alcoholic fatty liver disease, has reached global pandemic proportions. Lifestyle changes may result in a persistent positive energy balance, hastening the onset of these age-related disorders and consequently leading to a diminished lifespan. Although suggestions have been raised on the possible link between obesity and the gut microbiota, progress has been hampered due to the extensive diversity and complexities of the gut microbiota. Being recognized as a potential biomarker owing to its pivotal role in metabolic activities, the dysregulation of the gut microbiota can give rise to a persistent low-grade inflammatory state associated with chronic diseases during aging. This chronic inflammatory state, also known as inflammaging, induced by the chronic activation of the innate immune system via the macrophage, is controlled by the gut microbiota, which links nutrition, metabolism, and the innate immune response. Here, we present the functional roles of prebiotics, probiotics, synbiotics, and postbiotics as bioactive compounds by underscoring their putative contributions to (1) the reduction in gut hyperpermeability due to lipopolysaccharide (LPS) inactivation, (2) increased intestinal barrier function as a consequence of the upregulation of tight junction proteins, and (3) inhibition of proinflammatory pathways, overall leading to the alleviation of chronic inflammation in the management of obesity.
Collapse
Affiliation(s)
- Oladayo Emmanuel Apalowo
- Department of Food Science, Nutrition and Health Promotion, Mississippi State University, Starkville, MS 39762, USA
| | - Grace Adeola Adegoye
- Department of Nutrition and Health Science, Ball State University, Muncie, IN 47306, USA
| | | |
Collapse
|
20
|
Kaul R, Paul P, Harfouche M, Saliba R, Chaari A. Microbiome-modulating nutraceuticals ameliorate dyslipidemia in type 2 diabetes: A systematic review, meta-analysis, and meta-regression of clinical trials. Diabetes Metab Res Rev 2024; 40:e3675. [PMID: 37381688 DOI: 10.1002/dmrr.3675] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 03/26/2023] [Accepted: 05/19/2023] [Indexed: 06/30/2023]
Abstract
AIMS Type 2 Diabetes is intrinsically linked to cardiovascular disease (CVD) via diabetic dyslipidemia, both of which remain global health concerns with annually increasing prevalence. Given the established links between gut microbiome dysbiosis and metabolic diseases, its modulation is an attractive target to ameliorate metabolic imbalances in such patients. There is a need to quantitively summarise, analyse, and describe future directions in this field. METHODS We conducted a systematic review, meta-analysis, and meta-regression following searches in major scientific databases for clinical trials investigating the effect of pro/pre/synbiotics on lipid profile published until April 2022. Data were pooled using random-effects meta-analysis and reported as mean differences with 95% confidence intervals (CIs). PROSPERO No. CRD42022348525. RESULTS Data from 47 trial comparisons across 42 studies (n = 2692) revealed that, compared to placebo/control groups, the administration of pro/pre/synbiotics was associated with statistically significant changes in total cholesterol (-9.97 mg/dL [95% CI: -15.08; -4.87], p < 0.0001), low-density lipoprotein (-6.29 mg/dL [95% CI: -9.25; -3.33], p < 0.0001), high-density lipoprotein (+3.21 mg/dL [95% CI: 2.20; 4.22], p < 0.0001), very-low-density lipoprotein (-4.52 mg/dL [95% CI: -6.36; -2.67], p < 0.0001) and triglyceride (-22.93 mg/dL [95% CI: -33.99; -11.87], p < 0.001). These results are influenced by patient characteristics such as age or baseline BMI, and intervention characteristics such as dosage and duration. CONCLUSIONS Our study shows that adjunct supplementation with a subset of pro/pre/synbiotics ameliorates dyslipidemia in diabetic individuals and has the potential to reduce CVD risk. However, widespread inter-study heterogeneity and the presence of several unknown confounders limit their adoption in clinical practice; future trials should be designed with these in mind.
Collapse
Affiliation(s)
- Ridhima Kaul
- Medical Education Division, Weill Cornell Medicine-Qatar, Cornell University, Qatar Foundation - Education City, Doha, Qatar
| | - Pradipta Paul
- Medical Education Division, Weill Cornell Medicine-Qatar, Cornell University, Qatar Foundation - Education City, Doha, Qatar
| | - Manale Harfouche
- Infectious Disease Epidemiology Group, Weill Cornell Medicine - Qatar, Cornell University, Qatar Foundation - Education City, Doha, Qatar
- World Health Organization Collaborating Centre for Disease Epidemiology Analytics on HIV/AIDS, Sexually Transmitted Infections, and Viral Hepatitis, Weill Cornell Medicine - Qatar, Cornell University, Qatar Foundation - Education City, Doha, Qatar
| | - Reya Saliba
- Health Sciences Library, Weill Cornell Medicine - Qatar, Cornell University, Qatar Foundation - Education City, Doha, Qatar
| | - Ali Chaari
- Premedical Division, Weill Cornell Medicine - Qatar, Qatar Foundation - Education City, Doha, Qatar
| |
Collapse
|
21
|
Garcia-Gutierrez E, O’Mahony AK, Dos Santos RS, Marroquí L, Cotter PD. Gut microbial metabolic signatures in diabetes mellitus and potential preventive and therapeutic applications. Gut Microbes 2024; 16:2401654. [PMID: 39420751 PMCID: PMC11492678 DOI: 10.1080/19490976.2024.2401654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 07/08/2024] [Accepted: 09/03/2024] [Indexed: 10/19/2024] Open
Abstract
Diabetes mellitus can be subdivided into several categories based on origin and clinical characteristics. The most common forms of diabetes are type 1 (T1D), type 2 diabetes (T2D) and gestational diabetes mellitus (GDM). T1D and T2D are chronic diseases affecting around 537 million adults worldwide and it is projected that these numbers will increase by 12% over the next two decades, while GDM affects up to 30% of women during pregnancy, depending on diagnosis methods. These forms of diabetes have varied origins: T1D is an autoimmune disease, while T2D is commonly associated with, but not limited to, certain lifestyle patterns and GDM can result of a combination of genetic predisposition and pregnancy factors. Despite some pathogenic differences among these forms of diabetes, there are some common markers associated with their development. For instance, gut barrier impairment and inflammation associated with an unbalanced gut microbiota and their metabolites may be common factors in diabetes development and progression. Here, we summarize the microbial signatures that have been linked to diabetes, how they are connected to diet and, ultimately, the impact on metabolite profiles resulting from host-gut microbiota-diet interactions. Additionally, we summarize recent advances relating to promising preventive and therapeutic interventions focusing on the targeted modulation of the gut microbiota to alleviate T1D, T2D and GDM.
Collapse
Affiliation(s)
- Enriqueta Garcia-Gutierrez
- Food Biosciences Department, Teagasc Food Research Centre, Fermoy, Co. Cork, Ireland
- APC Microbiome Ireland, University College Cork, Co. Cork, Ireland
- VistaMilk SFI Research Centre, Fermoy, Co. Cork, Ireland
- Departamento de Ingeniería Agronómica, Instituto de Biotecnología Vegetal, ETSIA-Universidad Politécnica de Cartagena, Cartagena, Spain
| | - A. Kate O’Mahony
- Food Biosciences Department, Teagasc Food Research Centre, Fermoy, Co. Cork, Ireland
- APC Microbiome Ireland, University College Cork, Co. Cork, Ireland
- School of Microbiology, University College Cork, Co. Cork, Ireland
| | - Reinaldo Sousa Dos Santos
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universidad Miguel Hernández de Elche, Elche, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
| | - Laura Marroquí
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universidad Miguel Hernández de Elche, Elche, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
| | - Paul D. Cotter
- Food Biosciences Department, Teagasc Food Research Centre, Fermoy, Co. Cork, Ireland
- APC Microbiome Ireland, University College Cork, Co. Cork, Ireland
- VistaMilk SFI Research Centre, Fermoy, Co. Cork, Ireland
| |
Collapse
|
22
|
Antony MA, Chowdhury A, Edem D, Raj R, Nain P, Joglekar M, Verma V, Kant R. Gut microbiome supplementation as therapy for metabolic syndrome. World J Diabetes 2023; 14:1502-1513. [PMID: 37970133 PMCID: PMC10642415 DOI: 10.4239/wjd.v14.i10.1502] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 07/31/2023] [Accepted: 08/28/2023] [Indexed: 10/09/2023] Open
Abstract
The gut microbiome is defined as an ecological community of commensal symbiotic and pathogenic microorganisms that exist in our body. Gut microbiome dysbiosis is a condition of dysregulated and disrupted intestinal bacterial homeostasis, and recent evidence has shown that dysbiosis is related to chronic inflammation, insulin resistance, cardiovascular diseases (CVD), type 2 diabetes mellitus (T2DM), and obesity. It is well known that obesity, T2DM and CVD are caused or worsened by multiple factors like genetic predisposition, environmental factors, unhealthy high calorie diets, and sedentary lifestyle. However, recent evidence from human and mouse models suggest that the gut microbiome is also an active player in the modulation of metabolic syndrome, a set of risk factors including obesity, hyperglycemia, and dyslipidemia that increase the risk for CVD, T2DM, and other diseases. Current research aims to identify treatments to increase the number of beneficial microbiota in the gut microbiome in order to modulate metabolic syndrome by reducing chronic inflammation and insulin resistance. There is increasing interest in supplements, classified as prebiotics, probiotics, synbiotics, or postbiotics, and their effect on the gut microbiome and metabolic syndrome. In this review article, we have summarized current research on these supplements that are available to improve the abundance of beneficial gut microbiota and to reduce the harmful ones in patients with metabolic syndrome.
Collapse
Affiliation(s)
- Mc Anto Antony
- Department of Endocrinology, Diabetes and Metabolism, Medical University of South Carolina/AnMed Campus, Anderson, SC 29621, United States
| | - Aniqa Chowdhury
- Department of Endocrinology, Diabetes and Metabolism, Medical University of South Carolina/AnMed Campus, Anderson, SC 29621, United States
| | - Dinesh Edem
- Department of Endocrinology, Diabetes and Metabolism, University of Arkansas for Medical Sciences, Little Rock, AK 72205, United States
| | - Rishi Raj
- Department of Endocrinology, Diabetes and Metabolism, Pikeville Medical Center, Pikeville, KY 41501, United States
| | - Priyanshu Nain
- Department of Graduate Medical Education, Maulana Azad Medical College, Delhi 110002, India
| | - Mansi Joglekar
- Department of Endocrinology, Diabetes and Metabolism, Medical University of South Carolina/AnMed Campus, Anderson, SC 29621, United States
| | - Vipin Verma
- Department of Internal Medicine, Medical University of South Carolina/AnMed Campus, Anderson, SC 29621, United States
| | - Ravi Kant
- Department of Endocrinology, Diabetes and Metabolism, Medical University of South Carolina/AnMed Campus, Anderson, SC 29621, United States
| |
Collapse
|
23
|
Townsend JR, Kirby TO, Sapp PA, Gonzalez AM, Marshall TM, Esposito R. Nutrient synergy: definition, evidence, and future directions. Front Nutr 2023; 10:1279925. [PMID: 37899823 PMCID: PMC10600480 DOI: 10.3389/fnut.2023.1279925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Accepted: 09/28/2023] [Indexed: 10/31/2023] Open
Abstract
Nutrient synergy refers to the concept that the combined effects of two or more nutrients working together have a greater physiological impact on the body than when each nutrient is consumed individually. While nutrition science traditionally focuses on isolating single nutrients to study their effects, it is recognized that nutrients interact in complex ways, and their combined consumption can lead to additive effects. Additionally, the Dietary Reference Intakes (DRIs) provide guidelines to prevent nutrient deficiencies and excessive intake but are not designed to assess the potential synergistic effects of consuming nutrients together. Even the term synergy is often applied in different manners depending on the scientific discipline. Considering these issues, the aim of this narrative review is to investigate the potential health benefits of consuming different nutrients and nutrient supplements in combination, a concept we define as nutrient synergy, which has gained considerable attention for its impact on overall well-being. We will examine how nutrient synergy affects major bodily systems, influencing systemic health. Additionally, we will address the challenges associated with promoting and conducting research on this topic, while proposing potential solutions to enhance the quality and quantity of scientific literature on nutrient synergy.
Collapse
Affiliation(s)
- Jeremy R. Townsend
- Research, Nutrition, and Innovation, Athletic Greens International, Carson City, NV, United States
- Health & Human Performance, Concordia University Chicago, River Forest, IL, United States
| | - Trevor O. Kirby
- Research, Nutrition, and Innovation, Athletic Greens International, Carson City, NV, United States
| | - Philip A. Sapp
- Research, Nutrition, and Innovation, Athletic Greens International, Carson City, NV, United States
| | - Adam M. Gonzalez
- Department of Allied Health and Kinesiology, Hofstra University, Hempstead, NY, United States
| | - Tess M. Marshall
- Research, Nutrition, and Innovation, Athletic Greens International, Carson City, NV, United States
| | - Ralph Esposito
- Research, Nutrition, and Innovation, Athletic Greens International, Carson City, NV, United States
- Department of Nutrition, Food Studies, and Public Health, New York University-Steinhardt, New York, NY, United States
| |
Collapse
|
24
|
Antony MA, Patel S, Verma V, Kant R. The Role of Gut Microbiome Supplementation in COVID-19 Management. Cureus 2023; 15:e46960. [PMID: 38021562 PMCID: PMC10640765 DOI: 10.7759/cureus.46960] [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] [Accepted: 10/13/2023] [Indexed: 12/01/2023] Open
Abstract
COVID-19, which is caused by the RNA virus, SARS-CoV-2, mainly affects the respiratory system and has a varied clinical presentation. However, several studies have shown that COVID-19 can also affect the gastrointestinal (GI) system. Patients can experience various GI symptoms, such as vomiting and diarrhea, and the virus has been detected in the stool samples of patients hospitalized with COVID-19. There have also been rare reports of COVID-19 presenting with isolated GI symptoms and lack of respiratory symptoms, and the virus has also been detected for prolonged periods in the fecal samples of COVID-19 patients. Major alterations in the gut microbiome in the form of depletion of beneficial organisms and an abundance of pathogenic organisms have been reported in the fecal samples of hospitalized COVID-19 patients. Although the US FDA has approved several drugs to manage COVID-19, their efficacy remains modest. So, there is a constant ongoing effort to investigate novel treatment options for COVID-19. Health supplements like probiotics, prebiotics, postbiotics, and synbiotics have been popularly known for their various health benefits. In this review, we have summarized the current literature, which shows the potential benefit of these health supplements to mitigate and/or prevent the clinical presentation of COVID-19.
Collapse
Affiliation(s)
- Mc Anto Antony
- Department of Endocrinology, Diabetes and Metabolism, Medical University of South Carolina, Anderson, USA
| | - Siddharth Patel
- Department of Internal Medicine, Decatur Morgan Hospital, Decatur, USA
| | - Vipin Verma
- Department of Internal Medicine, Medical University of South Carolina, Anderson, USA
| | - Ravi Kant
- Department of Endocrinology, Diabetes and Metabolism, Medical University of South Carolina, Anderson, USA
| |
Collapse
|
25
|
Patra D, Banerjee D, Ramprasad P, Roy S, Pal D, Dasgupta S. Recent insights of obesity-induced gut and adipose tissue dysbiosis in type 2 diabetes. Front Mol Biosci 2023; 10:1224982. [PMID: 37842639 PMCID: PMC10575740 DOI: 10.3389/fmolb.2023.1224982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Accepted: 09/14/2023] [Indexed: 10/17/2023] Open
Abstract
An imbalance in microbial homeostasis, referred to as dysbiosis, is critically associated with the progression of obesity-induced metabolic disorders including type 2 diabetes (T2D). Alteration in gut microbial diversity and the abundance of pathogenic bacteria disrupt metabolic homeostasis and potentiate chronic inflammation, due to intestinal leakage or release of a diverse range of microbial metabolites. The obesity-associated shifts in gut microbial diversity worsen the triglyceride and cholesterol level that regulates adipogenesis, lipolysis, and fatty acid oxidation. Moreover, an intricate interaction of the gut-brain axis coupled with the altered microbiome profile and microbiome-derived metabolites disrupt bidirectional communication for instigating insulin resistance. Furthermore, a distinct microbial community within visceral adipose tissue is associated with its dysfunction in obese T2D individuals. The specific bacterial signature was found in the mesenteric adipose tissue of T2D patients. Recently, it has been shown that in Crohn's disease, the gut-derived bacterium Clostridium innocuum translocated to the mesenteric adipose tissue and modulates its function by inducing M2 macrophage polarization, increasing adipogenesis, and promoting microbial surveillance. Considering these facts, modulation of microbiota in the gut and adipose tissue could serve as one of the contemporary approaches to manage T2D by using prebiotics, probiotics, or faecal microbial transplantation. Altogether, this review consolidates the current knowledge on gut and adipose tissue dysbiosis and its role in the development and progression of obesity-induced T2D. It emphasizes the significance of the gut microbiota and its metabolites as well as the alteration of adipose tissue microbiome profile for promoting adipose tissue dysfunction, and identifying novel therapeutic strategies, providing valuable insights and directions for future research and potential clinical interventions.
Collapse
Affiliation(s)
- Debarun Patra
- Department of Biomedical Engineering, Indian Institute of Technology Ropar, Punjab, Punjab, India
| | - Dipanjan Banerjee
- Department of Molecular Biology and Biotechnology, Tezpur University, Napaam, Assam, India
| | - Palla Ramprasad
- Department of Biomedical Engineering, Indian Institute of Technology Ropar, Punjab, Punjab, India
| | - Soumyajit Roy
- Department of Biomedical Engineering, Indian Institute of Technology Ropar, Punjab, Punjab, India
| | - Durba Pal
- Department of Biomedical Engineering, Indian Institute of Technology Ropar, Punjab, Punjab, India
| | - Suman Dasgupta
- Department of Molecular Biology and Biotechnology, Tezpur University, Napaam, Assam, India
| |
Collapse
|
26
|
Li D, Cheng Y, Zeng X, Li Y, Xia Z, Yang X, Ren D. Polysaccharide from Ziyang Selenium-Enriched Green Tea Prevents Obesity and Promotes Adipose Thermogenesis via Modulating the Gut Microbiota. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:13363-13375. [PMID: 37647585 DOI: 10.1021/acs.jafc.3c04193] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/01/2023]
Abstract
The objective of the current study was to explore the potential mechanism of Ziyang selenium-enriched green tea polysaccharide (Se-GTP) against obesity. The results showed that Se-GTP significantly alleviated obesity and related metabolic disorders caused by high-fat diet (HFD) in mice. 16S rRNA gene sequencing results revealed that Se-GTP improved gut microbiota disturbance of obese mice and facilitated proliferation of probiotics such as Bacteroides, Bifidobacterium, Lactobacillus, and Akkermansia. In addition, the colonic content of succinate, a product of microbial metabolite in connection with adipocyte thermogenesis, was significantly enhanced by Se-GTP treatment. Therefore, Se-GTP facilitated brown adipose tissue (BAT) thermogenesis and inguinal white adipose tissue (iWAT) browning in obese mice, which could be revealed by increased expressions of thermogenic marker proteins UCP1, PGC-1α, and CIDEA in BAT and iWAT. Interestingly, Se-GTP intervention also observably increased the content of M2-like macrophages in iWAT of obese mice. To summarize, the results of this study are the first to show that Se-GTP can stimulate the browning of iWAT and BAT thermogenesis to counteract obesity, which may be pertinent with the alteration of gut microbiota in obese mice.
Collapse
Affiliation(s)
- Donglu Li
- Shaanxi Engineering Laboratory for Food Green Processing and Safety Control, and Shaanxi Key Laboratory for Hazard Factors Assessment in Processing and Storage of Agricultural Products, College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710119, China
| | - Yukun Cheng
- Shaanxi Engineering Laboratory for Food Green Processing and Safety Control, and Shaanxi Key Laboratory for Hazard Factors Assessment in Processing and Storage of Agricultural Products, College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710119, China
| | - Xiaoqian Zeng
- Shaanxi Engineering Laboratory for Food Green Processing and Safety Control, and Shaanxi Key Laboratory for Hazard Factors Assessment in Processing and Storage of Agricultural Products, College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710119, China
| | - Yixiao Li
- Shaanxi Engineering Laboratory for Food Green Processing and Safety Control, and Shaanxi Key Laboratory for Hazard Factors Assessment in Processing and Storage of Agricultural Products, College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710119, China
| | - Zengrun Xia
- Key Laboratory of Se-enriched Products Development and Quality Control, Ministry of Agriculture and Rural Affairs/National-Local Joint Engineering Laboratory of Se-enriched Food Development, Ankang 725000, China
| | - Xingbin Yang
- Shaanxi Engineering Laboratory for Food Green Processing and Safety Control, and Shaanxi Key Laboratory for Hazard Factors Assessment in Processing and Storage of Agricultural Products, College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710119, China
| | - Daoyuan Ren
- Shaanxi Engineering Laboratory for Food Green Processing and Safety Control, and Shaanxi Key Laboratory for Hazard Factors Assessment in Processing and Storage of Agricultural Products, College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710119, China
| |
Collapse
|
27
|
Megur A, Daliri EBM, Balnionytė T, Stankevičiūtė J, Lastauskienė E, Burokas A. In vitro screening and characterization of lactic acid bacteria from Lithuanian fermented food with potential probiotic properties. Front Microbiol 2023; 14:1213370. [PMID: 37744916 PMCID: PMC10516296 DOI: 10.3389/fmicb.2023.1213370] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 08/14/2023] [Indexed: 09/26/2023] Open
Abstract
The present work aimed to identify probiotic candidates from Lithuanian homemade fermented food samples. A total of 23 lactic acid bacteria were isolated from different fermented food samples. Among these, only 12 showed resistance to low pH, tolerance to pepsin, bile salts, and pancreatin. The 12 strains also exhibited antimicrobial activity against Staphylococcus aureus ATCC 29213, Salmonella Typhimurium ATCC 14028, Streptococcus pyogenes ATCC 12384, Streptococcus pyogenes ATCC 19615, and Klebsiella pneumoniae ATCC 13883. Cell-free supernatants of isolate 3A and 55w showed the strongest antioxidant activity of 26.37 μg/mL and 26.06 μg/mL, respectively. Isolate 11w exhibited the strongest auto-aggregation ability of 79.96% as well as the strongest adhesion to HCT116 colon cells (25.671 ± 0.43%). The selected strains were tested for their synbiotic relation in the presence of a prebiotic. The selected candidates showed high proliferation in the presence of 4% as compared to 2% galactooligosaccharides. Among the strains tested for tryptophan production ability, isolate 11w produced the highest L-tryptophan levels of 16.63 ± 2.25 μm, exhibiting psychobiotic ability in the presence of a prebiotic. The safety of these strains was studied by ascertaining their antibiotic susceptibility, mucin degradation, gelatin hydrolysis, and hemolytic activity. In all, isolates 40C and 11w demonstrated the most desirable probiotic potentials and were identified by 16S RNA and later confirmed by whole genome sequencing as Lacticaseibacillus paracasei 11w, and Lactiplantibacillus plantarum 40C: following with the harboring plasmid investigation. Out of all the 23 selected strains, only Lacticaseibacillus paracasei 11w showed the potential and desirable probiotic properties.
Collapse
Affiliation(s)
- Ashwinipriyadarshini Megur
- Department of Biological Models, Institute of Biochemistry, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Eric Banan-Mwine Daliri
- Department of Biological Models, Institute of Biochemistry, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Toma Balnionytė
- Department of Biological Models, Institute of Biochemistry, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Jonita Stankevičiūtė
- Department of Molecular Microbiology and Biotechnology, Institute of Biochemistry, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Eglė Lastauskienė
- Department of Microbiology and Biotechnology, Institute of Biosciences, Life Science Center, Vilnius University, Vilnius, Lithuania
| | - Aurelijus Burokas
- Department of Biological Models, Institute of Biochemistry, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| |
Collapse
|
28
|
Mishima M, Takeda S, Nagane M, Suzuki T, Ogata M, Shima A, Aihara N, Kamiie J, Suzuki R, Mizugaki H, Okamatsu-Ogura Y, Satoh T, Yamashita T. Prebiotic effect of poly-D-3-hydroxybutyrate prevents dyslipidemia in obese mice. FASEB J 2023; 37:e23121. [PMID: 37548278 DOI: 10.1096/fj.202301191r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Revised: 07/16/2023] [Accepted: 07/20/2023] [Indexed: 08/08/2023]
Abstract
Obesity is a global health problem caused by genetic, environmental, and psychological factors and is associated with various health disorders. As such, there is a growing focus on the prevention of obesity and related diseases. The gut microbiota plays a crucial role in these diseases and has become a therapeutic target. Prebiotics, such as poly-d-3-hydroxybutyric acid (PHB), have gained attention for their potential to alter the gut microbiota, promote beneficial bacterial growth, and alleviate obesity. In this study, we examined the prebiotic effects of PHB in obese mice. We found that, in C57BL/6N mice, PHB reduced blood lipid levels. Analysis of the intestinal microflora also revealed an increase in short-chain fatty acid-producing bacteria. When PHB was administered to obese mice, subcutaneous fat and dyslipidemia were reduced, and the number of beneficial bacteria in the intestinal microflora increased. Furthermore, fatty degradation and oxidative stress were suppressed in the liver. PHB regulates gut bacterial changes related to obesity and effectively inhibits dyslipidemia, suggesting that it could be a prebiotic agent for curing various obesity-related diseases. In summary, PHB increases the beneficial gut microbiota, leading to an alleviation of obesity-associated dyslipidemia.
Collapse
Affiliation(s)
- Mayuko Mishima
- School of Veterinary Medicine, Azabu University, Sagamihara, Japan
| | - Shiro Takeda
- School of Veterinary Medicine, Azabu University, Sagamihara, Japan
- Center for Human and Animal Symbiosis Science, Azabu University, Sagamihara, Japan
| | - Masaki Nagane
- School of Veterinary Medicine, Azabu University, Sagamihara, Japan
- Center for Human and Animal Symbiosis Science, Azabu University, Sagamihara, Japan
| | - Takehito Suzuki
- School of Veterinary Medicine, Azabu University, Sagamihara, Japan
- Center for Human and Animal Symbiosis Science, Azabu University, Sagamihara, Japan
| | - Masaya Ogata
- School of Veterinary Medicine, Azabu University, Sagamihara, Japan
| | - Ayaka Shima
- Anicom Specialty Medical Institute Inc., Tokyo, Japan
| | - Naoyuki Aihara
- School of Veterinary Medicine, Azabu University, Sagamihara, Japan
| | - Junichi Kamiie
- School of Veterinary Medicine, Azabu University, Sagamihara, Japan
| | - Rimina Suzuki
- School of Veterinary Medicine, Azabu University, Sagamihara, Japan
| | - Hinano Mizugaki
- School of Veterinary Medicine, Azabu University, Sagamihara, Japan
| | | | - Takumi Satoh
- Department of Antiaging Food Research, School of Bioscience and Biotechnology, Tokyo University of Technology, Hachioji, Japan
| | | |
Collapse
|
29
|
Devi R, Sharma E, Thakur R, Lal P, Kumar A, Altaf MA, Singh B, Tiwari RK, Lal MK, Kumar R. Non-dairy prebiotics: Conceptual relevance with nutrigenomics and mechanistic understanding of the effects on human health. Food Res Int 2023; 170:112980. [PMID: 37316060 DOI: 10.1016/j.foodres.2023.112980] [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: 01/15/2023] [Revised: 05/08/2023] [Accepted: 05/13/2023] [Indexed: 06/16/2023]
Abstract
The increasing health awareness of consumers has made a shift towards vegan and non-dairy prebiotics counterparts. Non-dairy prebiotics when fortified with vegan products have interesting properties and widely found its applications in food industry. The chief vegan products that have prebiotics added include water-soluble plant-based extracts (fermented beverages, frozen desserts), cereals (bread, cookies), and fruits (juices & jelly, ready to eat fruits). The main prebiotic components utilized are inulin, oligofructose, polydextrose, fructooligosaccharides, and xylooligosaccharides. Prebiotics' formulations, type and food matrix affect food products, host health, and technological attributes. Prebiotics from non-dairy sources have a variety of physiological effects that help to prevent and treat chronic metabolic diseases. This review focuses on mechanistic insight on non-dairy prebiotics affecting human health, how nutrigenomics is related to prebiotics development, and role of gene-microbes' interactions. The review will provide industries and researchers with important information about prebiotics, mechanism of non-dairy prebiotics and microbe interaction as well as prebiotic based vegan products.
Collapse
Affiliation(s)
- Rajni Devi
- Department of Microbiology, Punjab Agricultural University, Ludhiana 141004, India
| | - Eshita Sharma
- Department of Molecular Biology and Biochemistry, Guru Nanak Dev University, Amritsar 143005, India
| | - Richa Thakur
- Division of Silviculture and Forest Management, Himalayan Forest Research Institute, Conifer Campus, Shimla, India
| | - Priyanka Lal
- Department of Agricultural Economics and Extension, School of Agriculture, Lovely Professional University, Jalandhar GT Road (NH1), Phagwara, India
| | - Awadhesh Kumar
- Division of Crop Physiology and Biochemistry, ICAR-National Rice Research Institute, Cuttack, India
| | | | - Brajesh Singh
- ICAR-Central Potato Research Institute, Shimla 171001, India
| | | | - Milan Kumar Lal
- ICAR-Central Potato Research Institute, Shimla 171001, India.
| | - Ravinder Kumar
- ICAR-Central Potato Research Institute, Shimla 171001, India.
| |
Collapse
|
30
|
Batiha GES, Al-kuraishy HM, Al-Gareeb AI, Ashour NA, Negm WA. Potential role of tirzepatide towards Covid-19 infection in diabetic patients: a perspective approach. Inflammopharmacology 2023; 31:1683-1693. [PMID: 37208555 PMCID: PMC10198595 DOI: 10.1007/s10787-023-01239-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 04/21/2023] [Indexed: 05/21/2023]
Abstract
In Covid-19, variations in fasting blood glucose are considered a distinct risk element for a bad prognosis and outcome in Covid-19 patients. Tirazepatide (TZT), a dual glucagon-like peptide-1 (GLP-1)and glucose-dependent insulinotropic polypeptide (GIP) receptor agonist may be effective in managing Covid-19-induced hyperglycemia in diabetic and non-diabetic patients. The beneficial effect of TZT in T2DM and obesity is related to direct activation of GIP and GLP-1 receptors with subsequent improvement of insulin sensitivity and reduction of body weight. TZT improves endothelial dysfunction (ED) and associated inflammatory changes through modulation of glucose homeostasis, insulin sensitivity, and pro-inflammatory biomarkers release. TZT, through activation of the GLP-1 receptor, may produce beneficial effects against Covid-19 severity since GLP-1 receptor agonists (GLP-1RAs) have anti-inflammatory and pulmoprotective implications in Covid-19. Therefore, GLP-1RAs could effectively treat severely affected Covid-19 diabetic and non-diabetic patients. Notably, using GLP-1RAs in T2DM patients prevents glucose variability, a common finding in Covid-19 patients. Therefore, GLP-1RAs like TZT could be a therapeutic strategy in T2DM patients with Covid-19 to prevent glucose variability-induced complications. In Covid-19, the inflammatory signaling pathways are highly activated, resulting in hyperinflammation. GLP-1RAs reduce inflammatory biomarkers like IL-6, CRP, and ferritin in Covid-19 patients. Therefore, GLP-1RAs like TZ may be effective in Covid-19 patients by reducing the inflammatory burden. The anti-obesogenic effect of TZT may reduce Covid-19 severity by ameliorating body weight and adiposity. Furthermore, Covid-19 may induce substantial alterations in gut microbiota. GLP-1RA preserves gut microbiota and prevents intestinal dysbiosis. Herein, TZT, like other GLP-1RA, may attenuate Covid-19-induced gut microbiota alterations and, by this mechanism, may mitigate intestinal inflammation and systemic complications in Covid-19 patients with either T2DM or obesity. As opposed to that, glucose-dependent insulinotropic polypeptide (GIP) was reduced in obese and T2DM patients. However, activation of GIP-1R by TZT in T2DM patients improves glucose homeostasis. Thus, TZT, through activation of both GIP and GLP-1, may reduce obesity-mediated inflammation. In Covid-19, GIP response to the meal is impaired, leading to postprandial hyperglycemia and abnormal glucose homeostasis. Therefore, using TZT in severely affected Covid-19 patients may prevent the development of glucose variability and hyperglycemia-induced oxidative stress. Moreover, exaggerated inflammatory disorders in Covid-19 due to the release of pro-inflammatory cytokines like IL-1β, IL-6, and TNF-α may lead to systemic inflammation and cytokine storm development. Besides, GIP-1 inhibits expression of IL-1β, IL-6, MCP-1, chemokines and TNF-α. Therefore, using GIP-1RA like TZT may inhibit the onset of inflammatory disorders in severely affected Covid-19 patients. In conclusion, TZT, through activation of GLP-1 and GIP receptors, may prevent SARS-CoV-2-induced hyperinflammation and glucose variability in diabetic and non-diabetic patients.
Collapse
Affiliation(s)
- Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, AlBeheira, P.O. Box 22511, Damanhour, Egypt
| | - Hayder M. Al-kuraishy
- Department of Clinical Pharmacology and Medicine, College of Medicine, AL-Mustansiriyia University, P.O. Box 14132, Baghdad, Iraq
| | - Ali I. Al-Gareeb
- Department of Clinical Pharmacology and Medicine, College of Medicine, AL-Mustansiriyia University, P.O. Box 14132, Baghdad, Iraq
| | - Nada A. Ashour
- Department of Clinical Pharmacology and Toxicology, Faculty of Pharmacy, Tanta University, Tanta, 31527 Egypt
| | - Walaa A. Negm
- Department of Pharmacognosy, Faculty of Pharmacy, Tanta University, Tanta, 31527, Egypt
| |
Collapse
|
31
|
Dini I, Mancusi A. Weight Loss Supplements. Molecules 2023; 28:5357. [PMID: 37513229 PMCID: PMC10384751 DOI: 10.3390/molecules28145357] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 07/08/2023] [Accepted: 07/10/2023] [Indexed: 07/30/2023] Open
Abstract
Being overweight or obese can predispose people to chronic diseases and metabolic disorders such as cardiovascular illnesses, diabetes, Alzheimer's disease, and cancer, which are costly public health problems and leading causes of mortality worldwide. Many people hope to solve this problem by using food supplements, as they can be self-prescribed, contain molecules of natural origin considered to be incapable of causing damage to health, and the only sacrifice they require is economic. The market offers supplements containing food plant-derived molecules (e.g., primary and secondary metabolites, vitamins, and fibers), microbes (probiotics), and microbial-derived fractions (postbiotics). They can control lipid and carbohydrate metabolism, reduce appetite (interacting with the central nervous system) and adipogenesis, influence intestinal microbiota activity, and increase energy expenditure. Unfortunately, the copious choice of products and different legislation on food supplements worldwide can confuse consumers. This review summarizes the activity and toxicity of dietary supplements for weight control to clarify their potentiality and adverse reactions. A lack of research regarding commercially available supplements has been noted. Supplements containing postbiotic moieties are of particular interest. They are easier to store and transport and are safe even for people with a deficient immune system.
Collapse
Affiliation(s)
- Irene Dini
- Department of Pharmacy, University of Naples Federico II, Via Domenico Montesano 49, 80131 Napoli, Italy
| | - Andrea Mancusi
- Department of Food Microbiology, Istituto Zooprofilattico Sperimentale del Mezzogiorno, Via Salute 2, 80055 Portici, Italy
| |
Collapse
|
32
|
Bagyánszki M, Bódi N. Key elements determining the intestinal region-specific environment of enteric neurons in type 1 diabetes. World J Gastroenterol 2023; 29:2704-2716. [PMID: 37274063 PMCID: PMC10237112 DOI: 10.3748/wjg.v29.i18.2704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 03/28/2023] [Accepted: 04/17/2023] [Indexed: 05/11/2023] Open
Abstract
Diabetes, as a metabolic disorder, is accompanied with several gastrointestinal (GI) symptoms, like abdominal pain, gastroparesis, diarrhoea or constipation. Serious and complex enteric nervous system damage is confirmed in the background of these diabetic motility complaints. The anatomical length of the GI tract, as well as genetic, developmental, structural and functional differences between its segments contribute to the distinct, intestinal region-specific effects of hyperglycemia. These observations support and highlight the importance of a regional approach in diabetes-related enteric neuropathy. Intestinal large and microvessels are essential for the blood supply of enteric ganglia. Bidirectional morpho-functional linkage exists between enteric neurons and enteroglia, however, there is also a reciprocal communication between enteric neurons and immune cells on which intestinal microbial composition has crucial influence. From this point of view, it is more appropriate to say that enteric neurons partake in multidirectional communication and interact with these key players of the intestinal wall. These interplays may differ from segment to segment, thus, the microenvironment of enteric neurons could be considered strictly regional. The goal of this review is to summarize the main tissue components and molecular factors, such as enteric glia cells, interstitial cells of Cajal, gut vasculature, intestinal epithelium, gut microbiota, immune cells, enteroendocrine cells, pro-oxidants, antioxidant molecules and extracellular matrix, which create and determine a gut region-dependent neuronal environment in diabetes.
Collapse
Affiliation(s)
- Mária Bagyánszki
- Department of Physiology, Anatomy and Neuroscience, Faculty of Science and Informatics, University of Szeged, Szeged H-6726, Hungary
| | - Nikolett Bódi
- Department of Physiology, Anatomy and Neuroscience, Faculty of Science and Informatics, University of Szeged, Szeged H-6726, Hungary
| |
Collapse
|
33
|
Pezzino S, Sofia M, Mazzone C, Castorina S, Puleo S, Barchitta M, Agodi A, Gallo L, La Greca G, Latteri S. Gut Microbiome in the Progression of NAFLD, NASH and Cirrhosis, and Its Connection with Biotics: A Bibliometric Study Using Dimensions Scientific Research Database. BIOLOGY 2023; 12:biology12050662. [PMID: 37237476 DOI: 10.3390/biology12050662] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 03/30/2023] [Accepted: 04/19/2023] [Indexed: 05/28/2023]
Abstract
There is growing evidence that gut microbiota dysbiosis is linked to the etiopathogenesis of nonalcoholic fatty liver disease (NAFLD), from the initial stage of disease until the progressive stage of nonalcoholic steatohepatitis (NASH) and the final stage of cirrhosis. Conversely, probiotics, prebiotics, and synbiotics have shown promise in restoring dysbiosis and lowering clinical indicators of disease in a number of both preclinical and clinical studies. Additionally, postbiotics and parabiotics have recently garnered some attention. The purpose of this bibliometric analysis is to assess recent publishing trends concerning the role of the gut microbiome in the progression of NAFLD, NASH and cirrhosis and its connection with biotics. The free access version of the Dimensions scientific research database was used to find publications in this field from 2002 to 2022. VOSviewer and Dimensions' integrated tools were used to analyze current research trends. Research into the following topics is expected to emerge in this field: (1) evaluation of risk factors which are correlated with the progression of NAFLD, such as obesity and metabolic syndrome; (2) pathogenic mechanisms, such as liver inflammation through toll-like receptors activation, or alteration of short-chain fatty acids metabolisms, which contribute to NAFLD development and its progression in more severe forms, such as cirrhosis; (3) therapy for cirrhosis through dysbiosis reduction, and research on hepatic encephalopathy a common consequence of cirrhosis; (4) evaluation of diversity, and composition of gut microbiome under NAFLD, and as it varies under NASH and cirrhosis by rRNA gene sequencing, a tool which can also be used for the development of new probiotics and explore into the impact of biotics on the gut microbiome; (5) treatments to reduce dysbiosis with new probiotics, such as Akkermansia, or with fecal microbiome transplantation.
Collapse
Affiliation(s)
- Salvatore Pezzino
- Department of Surgical Sciences and Advanced Technologies "G. F. Ingrassia", Cannizzaro Hospital, University of Catania, 95123 Catania, Italy
| | - Maria Sofia
- Department of Surgical Sciences and Advanced Technologies "G. F. Ingrassia", Cannizzaro Hospital, University of Catania, 95123 Catania, Italy
| | - Chiara Mazzone
- Department of Surgical Sciences and Advanced Technologies "G. F. Ingrassia", Cannizzaro Hospital, University of Catania, 95123 Catania, Italy
| | - Sergio Castorina
- Department of Surgical Sciences and Advanced Technologies "G. F. Ingrassia", Cannizzaro Hospital, University of Catania, 95123 Catania, Italy
| | - Stefano Puleo
- Department of Surgical Sciences and Advanced Technologies "G. F. Ingrassia", Cannizzaro Hospital, University of Catania, 95123 Catania, Italy
| | - Martina Barchitta
- Department of Surgical Sciences and Advanced Technologies "G. F. Ingrassia", Cannizzaro Hospital, University of Catania, 95123 Catania, Italy
| | - Antonella Agodi
- Department of Surgical Sciences and Advanced Technologies "G. F. Ingrassia", Cannizzaro Hospital, University of Catania, 95123 Catania, Italy
| | - Luisa Gallo
- Department of Surgical Sciences and Advanced Technologies "G. F. Ingrassia", Cannizzaro Hospital, University of Catania, 95123 Catania, Italy
| | - Gaetano La Greca
- Department of Surgical Sciences and Advanced Technologies "G. F. Ingrassia", Cannizzaro Hospital, University of Catania, 95123 Catania, Italy
| | - Saverio Latteri
- Department of Surgical Sciences and Advanced Technologies "G. F. Ingrassia", Cannizzaro Hospital, University of Catania, 95123 Catania, Italy
| |
Collapse
|
34
|
Borshchev YY, Sonin DL, Burovenko IY, Borshchev VY, Cheburkin YV, Borshcheva OV, Galagudza MM. The Effect of Probiotic Strains on Myocardial Infarction Size, Biochemical and Immunological Parameters in Rats with Systemic Inflammatory Response Syndrome and Polymorbidity. J EVOL BIOCHEM PHYS+ 2022; 58:2058-2069. [PMID: 36573161 PMCID: PMC9773675 DOI: 10.1134/s0022093022060321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2022] [Revised: 10/31/2022] [Accepted: 11/01/2022] [Indexed: 12/24/2022]
Abstract
Numerous experimental and clinical studies have shown the effectiveness of various probiotic strains in metabolic disorders, gastrointestinal and liver diseases, immune system pathology. The effects of probiotics on cardiovascular dysfunction are less well known. The development and validation of a new experimental model in rats, including obesity, acute colon inflammation and antibiotic-induced dysbiosis, with common characteristics of systemic inflammatory response syndrome (SIRS), became the basis for investigating the effects of probiotic drugs on myocardial resistance to ischemic-reperfusion injury (IRI) using an in vivo model of infarction after coronary occlusion. A 24% increase in myocardial infarction compared to intact animals (p < 0.05) and significant changes in leukogram, biochemical and immunological parameters were found in Wistar rats with SIRS modelling. Introduction of a mixture of strains of Lactobacillus acidophilus (LA-5) and Bifidobacterium animalis subsp. lactis (BB-12) to animals with SIRS reduced infarct size to a value close to the control. Rats treated with LA-5 and BB-12 also showed normalization of the leukocyte count, bile acids, transforming growth factor-β, interleukins: IL-1α, IL-2, IL-6, IL-8, tumor necrosis factor-α, lipopolysaccharide and monocyte chemoattractant protein-1 in blood in comparison with the SIRS group and with the groups treated with other probiotic strains. The obtained data convincingly show the prospects for further study of the cardiotropic potential of probiotic microorganisms in translational studies.
Collapse
Affiliation(s)
- Yu. Yu. Borshchev
- Almazov National Medical Research Center of the Ministry of Health of the Russian Federation, St. Petersburg, Russia
| | - D. L. Sonin
- Almazov National Medical Research Center of the Ministry of Health of the Russian Federation, St. Petersburg, Russia
- Pavlov First Saint Petersburg State Medical University, St. Petersburg, Russia
| | - I. Yu. Burovenko
- Almazov National Medical Research Center of the Ministry of Health of the Russian Federation, St. Petersburg, Russia
| | - V. Yu. Borshchev
- Pavlov First Saint Petersburg State Medical University, St. Petersburg, Russia
| | - Yu. V. Cheburkin
- Almazov National Medical Research Center of the Ministry of Health of the Russian Federation, St. Petersburg, Russia
| | - O. V. Borshcheva
- Almazov National Medical Research Center of the Ministry of Health of the Russian Federation, St. Petersburg, Russia
| | - M. M. Galagudza
- Almazov National Medical Research Center of the Ministry of Health of the Russian Federation, St. Petersburg, Russia
- Pavlov First Saint Petersburg State Medical University, St. Petersburg, Russia
| |
Collapse
|
35
|
In Vitro Production of Galactooligosaccharides by a Novel β-Galactosidase of Lactobacillus bulgaricus. Int J Mol Sci 2022; 23:ijms232214308. [PMID: 36430784 PMCID: PMC9697242 DOI: 10.3390/ijms232214308] [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: 10/30/2022] [Revised: 11/11/2022] [Accepted: 11/14/2022] [Indexed: 11/19/2022] Open
Abstract
β-galactosidase is an enzyme with dual activity and important industrial application. As a hydrolase, the enzyme eliminates lactose in milk, while as a trans-galactosidase it produces prebiotic galactooligosaccharides (GOS) with various degrees of polymerization (DP). The aim of the present study is the molecular characterization of β-galactosidase from a Bulgarian isolate, Lactobacillus delbrueckii subsp. bulgaricus 43. The sequencing of the β-gal gene showed that it encodes a new enzyme with 21 amino acid replacements compared to all other β-galactosidases of this species. The molecular model revealed that the new β-galactosidase acts as a tetramer. The amino acids D207, H386, N464, E465, Y510, E532, H535, W562, N593, and W980 form the catalytic center and interact with Mg2+ ions and substrate. The β-gal gene was cloned into a vector allowing heterologous expression of E. coli BL21(DE3) with high efficiency, as the crude enzyme reached 3015 U/mL of the culture or 2011 U/mg of protein. The enzyme's temperature optimum at 55 °C, a pH optimum of 6.5, and a positive influence of Mg2+, Mn2+, and Ca2+ on its activity were observed. From lactose, β-Gal produced a large amount of GOS with DP3 containing β-(1→3) and β-(1→4) linkages, as the latter bond is particularly atypical for the L. bulgaricus enzymes. DP3-GOS formation was positively affected by high lactose concentrations. The process of lactose conversion was rapid, with a 34% yield of DP3-GOS in 6 h, and complete degradation of 200 g/L of lactose for 12 h. On the other hand, the enzyme was quite stable at 55 °C and retained about 20% of its activity after 24 h of incubation at this temperature. These properties expand our horizons as regards the use of β-galactosidases in industrial processes for the production of lactose-free milk and GOS-enriched foods.
Collapse
|
36
|
Portincasa P, Bonfrate L, Wang DQH, Frühbeck G, Garruti G, Di Ciaula A. Novel insights into the pathogenic impact of diabetes on the gastrointestinal tract. Eur J Clin Invest 2022; 52:e13846. [PMID: 35904418 DOI: 10.1111/eci.13846] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Revised: 06/20/2022] [Accepted: 06/26/2022] [Indexed: 11/09/2022]
Abstract
Type 2 and type 1 diabetes are common endocrine disorders with a progressively increasing incidence worldwide. These chronic, systemic diseases have multiorgan implications, and the whole gastrointestinal (GI) tract represents a frequent target in terms of symptom appearance and interdependent pathophysiological mechanisms. Metabolic alterations linked with diabetic complications, neuropathy and disrupted hormone homeostasis can lead to upper and/or lower GI symptoms in up to 75% of diabetic patients, with multifactorial involvement of the oesophagus, stomach, upper and lower intestine, and of the gallbladder. On the other hand, altered gastrointestinal motility and/or secretions are able to affect glucose and lipid homeostasis in the short and long term. Finally, diabetes has been linked with increased cancer risk at different levels of the GI tract. The presence of GI symptoms and a comprehensive assessment of GI function should be carefully considered in the management of diabetic patients to avoid further complications and to ameliorate the quality of life. Additionally, the presence of gastrointestinal dysfunction should be adequately managed to improve metabolic homeostasis, the efficacy of antidiabetic treatments and secondary prevention strategies.
Collapse
Affiliation(s)
- Piero Portincasa
- Clinica Medica "A. Murri", Department of Biomedical Sciences and Human Oncology, University of Bari Aldo Moro, Bari, Italy
| | - Leonilde Bonfrate
- Clinica Medica "A. Murri", Department of Biomedical Sciences and Human Oncology, University of Bari Aldo Moro, Bari, Italy
| | - David Q-H Wang
- Department of Medicine and Genetics, Division of Gastroenterology and Liver Diseases, Marion Bessin Liver Research Center, Einstein-Mount Sinai Diabetes Research Center, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Gema Frühbeck
- Department of Endocrinology & Nutrition, Clínica Universidad de Navarra, Pamplona, Spain Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain.,CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), ISCIII, Pamplona, Spain.,Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Gabriella Garruti
- Department of Emergency and Organ Transplants, Unit of Endocrinology, University of Bari Medical School, Bari, Italy
| | - Agostino Di Ciaula
- Clinica Medica "A. Murri", Department of Biomedical Sciences and Human Oncology, University of Bari Aldo Moro, Bari, Italy
| |
Collapse
|
37
|
Paul P, Kaul R, Harfouche M, Arabi M, Al-Najjar Y, Sarkar A, Saliba R, Chaari A. The effect of microbiome-modulating probiotics, prebiotics and synbiotics on glucose homeostasis in type 2 diabetes: A systematic review, meta-analysis, and meta-regression of clinical trials. Pharmacol Res 2022; 185:106520. [DOI: 10.1016/j.phrs.2022.106520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 10/10/2022] [Accepted: 10/17/2022] [Indexed: 11/29/2022]
|
38
|
Zhao Q, Shi J, Chen S, Hao D, Wan S, Niu H, Zhang Y. Salidroside Affects Gut Microbiota Structure in db/db Mice by Affecting Insulin, Blood Glucose and Body Weight. Diabetes Metab Syndr Obes 2022; 15:2619-2631. [PMID: 36060789 PMCID: PMC9438798 DOI: 10.2147/dmso.s372192] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 08/05/2022] [Indexed: 11/23/2022] Open
Abstract
PURPOSE The purpose of this study was to investigate the regulatory effect of salidroside on the intestinal flora of mice with type 2 diabetes (T2DM) and its protective effect in the body. PATIENTS AND METHODS We acclimated 8-week-old mice for 7 days, divided them into 4 groups, and continued dosing for 8 weeks. We recorded weekly blood glucose levels and body weight for each mouse. After the completion of the feeding cycle, the 16S rRNA of the intestinal flora in the mice was sequenced, and the insulin and C-peptide levels in each group of mice were measured. Four samples were taken from each group for liver and kidney section staining. RESULTS Our results showed that gut microbiota diversity and function were significantly different between the diabetic mice and healthy mice and that insulin levels, body weight, and blood glucose levels could significantly influence gut microbiota changes at the genus level. The gut microbiota diversity and function of db/db mice were also altered after salidroside administration. Salidroside could attenuate inflammatory damage, lipid accumulation and inflammatory changes in the diabetic liver, as well as diabetic kidney damage. Candidatus arthromitus and Odoribacter are important species of the microbiota during diabetes and may serve as potential therapeutic targets. CONCLUSION Our investigation of the associated pathological conditions and fecal microbiota in db/db mice provides new insights into the pathogenesis of T2DM and provides implications for the diagnosis and treatment of T2DM.
Collapse
Affiliation(s)
- Qin Zhao
- Biobank, Hospital of Chengdu Office of People’s Government of Tibetan Autonomous Region (Hospital.C.T.), Chengdu, Sichuan Povince, People’s Republic of China
| | - Jing Shi
- Biobank, Hospital of Chengdu Office of People’s Government of Tibetan Autonomous Region (Hospital.C.T.), Chengdu, Sichuan Povince, People’s Republic of China
| | - Siyuan Chen
- Biobank, Hospital of Chengdu Office of People’s Government of Tibetan Autonomous Region (Hospital.C.T.), Chengdu, Sichuan Povince, People’s Republic of China
| | - Doudou Hao
- Biobank, Hospital of Chengdu Office of People’s Government of Tibetan Autonomous Region (Hospital.C.T.), Chengdu, Sichuan Povince, People’s Republic of China
| | - Sha Wan
- Biobank, Hospital of Chengdu Office of People’s Government of Tibetan Autonomous Region (Hospital.C.T.), Chengdu, Sichuan Povince, People’s Republic of China
| | - Haomeng Niu
- Medical School, Tibet University, Lhasa, Tibet Autonomous Region, People’s Republic of China
| | - Yongqun Zhang
- Biobank, Hospital of Chengdu Office of People’s Government of Tibetan Autonomous Region (Hospital.C.T.), Chengdu, Sichuan Povince, People’s Republic of China
- Correspondence: Yongqun Zhang, Biobank, Hospital of Chengdu Office of People’s Government of Tibetan Autonomous Region (Hospital.C.T.), No. 20 Ximianqiao Road, Chengdu, Sichuan Povince, 610041, People’s Republic of China, Tel +86-28-85593218, Fax +86 28-85558071, Email
| |
Collapse
|