1
|
Clementino JR, de Oliveira LIG, Salgaço MK, de Oliveira FL, Mesa V, Tavares JF, Silva-Pereira L, Raimundo BVB, Oliveira KC, Medeiros AI, Silva FA, Sivieri K, Magnani M. β-Glucan Alone or Combined with Lactobacillus acidophilus Positively Influences the Bacterial Diversity and Metabolites in the Colonic Microbiota of Type II Diabetic Patients. Probiotics Antimicrob Proteins 2025:10.1007/s12602-025-10491-9. [PMID: 40011383 DOI: 10.1007/s12602-025-10491-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/14/2025] [Indexed: 02/28/2025]
Abstract
β-Glucan is a fermentable polysaccharide with prebiotic properties that has been shown to improve metabolic indicators. This study evaluated the effects of spent brewer's yeast β-glucan (BGL) and Lactobacillus acidophilus LA-5 (106 CFU/g) (LA5) alone and in combination (LA5-BGL) on the composition of the fecal microbiome of adults with type 2 diabetes mellitus (T2DM) using the Human Gut Microbial Ecosystem Simulator (SHIME®). Short-chain fatty acids (SCFAs), ammonium ions, and cytokines (IL-6 and IL-10) were measured. BGL, LA5, and LA5-BGL increased (p < 0.05) the richness and diversity of microbial communities in the gut microbiome of individuals with T2DM. All treatments increased (p < 0.05) the abundance of Bacteroides, Alistipes, Lactobacillus, Subdoligranulum, and Acidaminococcus, along with increased (p < 0.05) production of SCFAs and anti-inflammatory cytokine (IL-10) compared to the control group. BGL treatments showed a greater increase in microbial diversity, SCFAs levels (butyric, propionic, and acetic acid), and the anti-inflammatory cytokine (IL-10). LA5 showed the highest decrease in ammonium ion levels. Results indicate that BGL may have a prebiotic and immunomodulatory effect on the fecal microbial community and metabolic indicators in adults with type 2 diabetes mellitus (T2DM). Findings underscore the role of BGL as a prebiotic food.
Collapse
Affiliation(s)
| | | | | | | | - Victoria Mesa
- School of Nutrition and Dietetics, Universidad de Antioquia, Medellín, Colombia
- Faculty of Pharmacy, Université Paris Cité, Paris, France
| | - Josean Fechine Tavares
- Department of Pharmaceutical Sciences, Health Science Center, Federal University of Paraíba, João Pessoa, Brazil
| | - Ludmilla Silva-Pereira
- Department of Biological Sciences, São Paulo State University, Araraquara, São Paulo, Brazil
| | | | - Karen Cristina Oliveira
- Department of Biological Sciences, São Paulo State University, Araraquara, São Paulo, Brazil
| | - Alexandra Ivo Medeiros
- Department of Biological Sciences, São Paulo State University, Araraquara, São Paulo, Brazil
| | - Francyeli Araújo Silva
- Laboratory of Microbial Processes in Foods, Federal University of Paraíba, João Pessoa, Brazil
| | - Katia Sivieri
- Department of Food and Nutrition, São Paulo State University, Araraquara, Brazil
| | - Marciane Magnani
- Laboratory of Microbial Processes in Foods, Federal University of Paraíba, João Pessoa, Brazil.
| |
Collapse
|
2
|
Gao Y, Zhao T, Lv N, Liu S, Yuan T, Fu Y, Zhao W, Zhu B. Metformin-induced changes of the gut microbiota in patients with type 2 diabetes mellitus: results from a prospective cohort study. Endocrine 2024; 85:1178-1192. [PMID: 38761345 DOI: 10.1007/s12020-024-03828-x] [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: 01/30/2024] [Accepted: 04/09/2024] [Indexed: 05/20/2024]
Abstract
BACKGROUND The influence of the microbiota on hypoglycemic agents is becoming more apparent. The effects of metformin, a primary anti-diabetes drug, on gut microbiota are still not fully understood. RESEARCH DESIGN AND METHODS This prospective cohort study aims to investigate the longitudinal effects of metformin on the gut microbiota of 25 treatment-naïve diabetes patients, each receiving a daily dose of 1500 mg. Microbiota compositions were analyzed at baseline, and at 1, 3, and 6 months of medication using 16S rRNA gene sequencing. RESULTS Prior to the 3-month period of metformin treatment, significant improvements were noted in body mass index (BMI) and glycemic-related parameters, such as fasting blood glucose (FPG) and hemoglobin A1c (HbA1c), alongside homeostasis model assessment indices of insulin resistance (HOMA-IR). At the 3-month mark of medication, a significant reduction in the α-diversity of the gut microbiota was noted, while β-diversity exhibited no marked variances throughout the treatment duration. The Firmicutes to Bacteroidetes ratio. markedly decreased. Metformin treatment consistently increased Escherichia-Shigella and decreased Romboutsia, while Pseudomonas decreased at 3 months. Fuzzy c-means clustering identified three longitudinal trajectory clusters for microbial fluctuations: (i) genera temporarily changing, (ii) genera continuing to decrease (Bacteroides), and (iii) genera continuing to increase(Lachnospiraceae ND3007 group, [Eubacterium] xylanophilum group, Romboutsia, Faecalibacterium and Ruminococcaceae UCG-014). The correlation matrix revealed associations between specific fecal taxa and metformin-related clinical parameters HbA1c, FPG, Uric Acid (UA), high-density lipoproteincholesterol (HDL-C), alanine aminotransferase (ALT), hypersensitive C-reactive protein (hs-CRP), triglyceride (TG) (P < 0.05). Metacyc database showed that metformin significantly altered 17 functional pathways. Amino acid metabolism pathways such as isoleucine biosynthesis predominated in the post-treatment group. CONCLUSIONS Metformin's role in glucose metabolism regulation may primarily involve specific alterations in certain gut microbial species rather than an overall increase in microbial species diversity. This may suggest gut microbiota targets in future studies on metabolic abnormalities caused by metformin.
Collapse
Affiliation(s)
- Yuting Gao
- Key Laboratory of Endocrinology of National Health Commission, Department of Endocrinology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Tianyi Zhao
- Department of Physical Examination Center, China-Japan Friendship Hospital, Beijing, China
| | - Na Lv
- Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Shixuan Liu
- Department of Pathology, China-Japan Friendship Hospital, Beijing, China
| | - Tao Yuan
- Key Laboratory of Endocrinology of National Health Commission, Department of Endocrinology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Yong Fu
- Key Laboratory of Endocrinology of National Health Commission, Department of Endocrinology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Weigang Zhao
- Key Laboratory of Endocrinology of National Health Commission, Department of Endocrinology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China.
| | - Baoli Zhu
- Institute of Microbiology, Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
3
|
Li DZ, Ding J, Fan JB. Establishment of a risk prediction model for gastrointestinal motility disorder in patients with type 2 diabetes mellitus. WORLD CHINESE JOURNAL OF DIGESTOLOGY 2024; 32:591-599. [DOI: 10.11569/wcjd.v32.i8.591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2024]
|
4
|
Tao M, Cao K, Pu X, Hou Y, He L, Liu W, Ren Y, Yang X. Cadmium exposure induces changes in gut microbial composition and metabolic function in long-tailed dwarf hamsters, Cricetulus longicaudatus. Ecol Evol 2024; 14:e11682. [PMID: 38966245 PMCID: PMC11222731 DOI: 10.1002/ece3.11682] [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: 04/01/2024] [Revised: 05/30/2024] [Accepted: 06/19/2024] [Indexed: 07/06/2024] Open
Abstract
Numerous studies have demonstrated that exposure to cadmium disrupts the diversity and composition of the gut microbiota, resulting in damage to organ tissue. However, there remains a lack of comprehensive understanding regarding the broader ecological reality associated with this phenomenon. In this study, we conducted a thorough evaluation of the effects of different concentrations of Cd (6, 12, 24, and 48 mg/L) over a period of 35 consecutive days on the organ viscera and the gut microbiota of long-tailed dwarf hamsters, Cricetulus longicaudatus (Rodentia: Cricetidae), using histopathological analysis, 16S rDNA, and metagenome sequencing. Our findings revealed that the results suggest that Cd exposure induced liver, spleen, and kidney damage, potentially leading to increased intestinal permeability and inflammation. These alterations were accompanied by significant perturbations in the gut microbiota composition, particularly affecting potentially pathogenic bacteria such as Prevotella and Treponema within the gut ecosystem. Consequently, host susceptibility to underlying diseases was heightened due to these changes. Notably though, Cd exposure did not significantly impact the overall structure of the gut microbiota itself. Additionally, Cd exposure induced significant changes in the metabolic functions, with the pathways related to disease and environmental information processing notably enhanced, possibly indicating stronger innate defense mechanisms against external injuries among wild mammals exposed to Cd. This study offers a novel approach to comprehensively evaluate the significant impact of Cd pollution on ecosystems by investigating both structural and functional alterations in the digestive system, as well as disruptions in intestinal flora among wild mammals.
Collapse
Affiliation(s)
- Mengfan Tao
- Shanxi Key Laboratory of Integrated Pest Management in Agriculture, College of Plant ProtectionShanxi Agricultural UniversityTaiyuanChina
| | - Kanglin Cao
- Shanxi Key Laboratory of Integrated Pest Management in Agriculture, College of Plant ProtectionShanxi Agricultural UniversityTaiyuanChina
| | - Xinsheng Pu
- Shanxi Key Laboratory of Integrated Pest Management in Agriculture, College of Plant ProtectionShanxi Agricultural UniversityTaiyuanChina
| | - Yu Hou
- Shanxi Key Laboratory of Integrated Pest Management in Agriculture, College of Plant ProtectionShanxi Agricultural UniversityTaiyuanChina
| | - Lei He
- Shanxi Forestry and Grassland General Engineering StationTaiyuanChina
| | - Wei Liu
- Shanxi Forestry and Grassland General Engineering StationTaiyuanChina
| | - Yue Ren
- Shanxi Key Laboratory of Integrated Pest Management in Agriculture, College of Plant ProtectionShanxi Agricultural UniversityTaiyuanChina
| | - Xin'gen Yang
- Shanxi Key Laboratory of Integrated Pest Management in Agriculture, College of Plant ProtectionShanxi Agricultural UniversityTaiyuanChina
| |
Collapse
|
5
|
Rathi A, Pagare R. Efficacy and Safety of Bacillus coagulans LBSC in Drug Induced Constipation Associated With Functional Gastrointestinal Disorder: A Double-Blind, Randomized, Interventional, Parallel, Controlled Trial a Clinical Study on Bacillus coagulans LBSC for Drug Induced Constipation Associated With FGIDs. GLOBAL ADVANCES IN INTEGRATIVE MEDICINE AND HEALTH 2024; 13:27536130241286511. [PMID: 39295947 PMCID: PMC11409293 DOI: 10.1177/27536130241286511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 09/02/2024] [Accepted: 09/08/2024] [Indexed: 09/21/2024]
Abstract
Background Active drugs and nutraceutical supplements commonly induce various gastrointestinal illnesses, and constipation is a major gastrointestinal symptom accompanied with functional gastrointestinal disorders. Drug-induced imbalance in gut microbiota may play critical role in such physiological disturbances. Probiotics have been known for resuming normal and healthy gut microbiome. Objective To investigate the clinical efficacy and safety of Bacillus coagulans LBSC in the treatment of drug induced constipation associated with functional gastrointestinal disorder (FGID) symptoms. Methods A prospective, interventional, randomized, double-blind, parallel, multi-arm, controlled trial with 168 patients experiencing drug induced constipation associated with FGID symptoms (DICAWFGID) screened through Rome IV criteria were randomized into 2 arms, i.e. placebo arm (n = 28) and atorvastatin, atenolol, metformin, amitriptyline, and calcium in test arm (n = 28/arm). Patients in both arms received similar dosages (1 g sachet, 3 times a day) for 35 days. The occurrence of constipation using Bristol Stool Form Scale, assessment of degree of constipation on 4-point Likert scale, occurrence of hard stool and degree of stool expulsion on 3-point scale, and defecation frequency were primary endpoints. While, secondary outcomes consisted of the changes in severity of FGID symptoms, visual analogue scale and tolerance to IP, along with reports of adverse events (AEs) and severe adverse events (SAEs). Results There was a significant reduction in occurrence of constipation (≥98.6% and P-value <0.05) in test arm over the placebo arm. Assessment of co-primary endpoints showed significant improvements in degree of stool consistency (P-value 0.0232; CI: 0.1870, 1.1629), borderline significantly superior in degree of stool expulsion (P-value 0.0553; CI: 0.0378, -0.4939), while the other co-primary efficacy endpoints displayed considerably improved advancement (non-significant, P-value ≥0.05). The intra group analysis of symptoms at start of treatment (SOT) and end of treatment (EOT) revealed a significant reduction in scores for occurrence of constipation and degree of constipation, whereas significant improvement in the scores for degree of stool consistency and degree of stool expulsion (P-value <0.001) after the intervention period. In secondary endpoints, the processed responses clearly signified a considerable positive improvement (non-significant, P-value ≥0.05) in other symptoms of constipation associated with FGIDs as determined by the changes in the EOT-SOT score. The study data also highlighted the safety of Bacillus coagulans LBSC at the studied dose. No AEs and/or SAEs were documented during the investigation. Conclusion At the studied dose, Bacillus coagulans LBSC was safe for oral consumption and effective in the management of the drug induced constipation associated with FGIDs symptoms.
Collapse
Affiliation(s)
- Ankit Rathi
- Department of Biological Sciences, School of Science, Sandip University, Nashik, Maharashtra, India
| | - Ravikiran Pagare
- Department of Biological Sciences, School of Science, Sandip University, Nashik, Maharashtra, India
| |
Collapse
|
6
|
Foretz M, Guigas B, Viollet B. Metformin: update on mechanisms of action and repurposing potential. Nat Rev Endocrinol 2023; 19:460-476. [PMID: 37130947 PMCID: PMC10153049 DOI: 10.1038/s41574-023-00833-4] [Citation(s) in RCA: 265] [Impact Index Per Article: 132.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/24/2023] [Indexed: 05/04/2023]
Abstract
Currently, metformin is the first-line medication to treat type 2 diabetes mellitus (T2DM) in most guidelines and is used daily by >200 million patients. Surprisingly, the mechanisms underlying its therapeutic action are complex and are still not fully understood. Early evidence highlighted the liver as the major organ involved in the effect of metformin on reducing blood levels of glucose. However, increasing evidence points towards other sites of action that might also have an important role, including the gastrointestinal tract, the gut microbial communities and the tissue-resident immune cells. At the molecular level, it seems that the mechanisms of action vary depending on the dose of metformin used and duration of treatment. Initial studies have shown that metformin targets hepatic mitochondria; however, the identification of a novel target at low concentrations of metformin at the lysosome surface might reveal a new mechanism of action. Based on the efficacy and safety records in T2DM, attention has been given to the repurposing of metformin as part of adjunct therapy for the treatment of cancer, age-related diseases, inflammatory diseases and COVID-19. In this Review, we highlight the latest advances in our understanding of the mechanisms of action of metformin and discuss potential emerging novel therapeutic uses.
Collapse
Affiliation(s)
- Marc Foretz
- Université Paris Cité, CNRS, Inserm, Institut Cochin, Paris, France
| | - Bruno Guigas
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands
| | - Benoit Viollet
- Université Paris Cité, CNRS, Inserm, Institut Cochin, Paris, France.
| |
Collapse
|
7
|
Li R, Shokri F, Rincon AL, Rivadeneira F, Medina-Gomez C, Ahmadizar F. Bi-Directional Interactions between Glucose-Lowering Medications and Gut Microbiome in Patients with Type 2 Diabetes Mellitus: A Systematic Review. Genes (Basel) 2023; 14:1572. [PMID: 37628624 PMCID: PMC10454120 DOI: 10.3390/genes14081572] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 07/24/2023] [Accepted: 07/26/2023] [Indexed: 08/27/2023] Open
Abstract
Background: Although common drugs for treating type 2 diabetes (T2D) are widely used, their therapeutic effects vary greatly. The interaction between the gut microbiome and glucose-lowering drugs is one of the main contributors to the variability in T2D progression and response to therapy. On the one hand, glucose-lowering drugs can alter gut microbiome components. On the other hand, specific gut microbiota can influence glycemic control as the therapeutic effects of these drugs. Therefore, this systematic review assesses the bi-directional relationships between common glucose-lowering drugs and gut microbiome profiles. Methods: A systematic search of Embase, Web of Science, PubMed, and Google Scholar databases was performed. Observational studies and randomised controlled trials (RCTs), published from inception to July 2023, comprising T2D patients and investigating bi-directional interactions between glucose-lowering drugs and gut microbiome, were included. Results: Summarised findings indicated that glucose-lowering drugs could increase metabolic-healthy promoting taxa (e.g., Bifidobacterium) and decrease harmful taxa (e.g., Bacteroides and Intestinibacter). Our findings also showed a significantly different abundance of gut microbiome taxa (e.g., Enterococcus faecium (i.e., E. faecium)) in T2D patients with poor compared to optimal glycemic control. Conclusions: This review provides evidence for glucose-lowering drug and gut microbiome interactions, highlighting the potential of gut microbiome modulators as co-adjuvants for T2D treatment.
Collapse
Affiliation(s)
- Ruolin Li
- Department of Internal Medicine, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands; (R.L.); (F.R.); (C.M.-G.)
| | - Fereshteh Shokri
- Department of Epidemiology, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands;
| | - Alejandro Lopez Rincon
- Department of Data Science & Biostatistics, Julius Global Health, University Medical Center Utrecht, 3508 GA Utrecht, The Netherlands;
| | - Fernando Rivadeneira
- Department of Internal Medicine, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands; (R.L.); (F.R.); (C.M.-G.)
| | - Carolina Medina-Gomez
- Department of Internal Medicine, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands; (R.L.); (F.R.); (C.M.-G.)
| | - Fariba Ahmadizar
- Department of Epidemiology, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands;
- Department of Data Science & Biostatistics, Julius Global Health, University Medical Center Utrecht, 3508 GA Utrecht, The Netherlands;
| |
Collapse
|
8
|
Carrizales-Sánchez AK, Tamez-Rivera O, García-Gamboa R, García-Cayuela T, Rodríguez-Gutiérrez NA, Elizondo-Montemayor L, García-Rivas G, Pacheco A, Hernández-Brenes C, Senés-Guerrero C. Gut microbial composition and functionality of school-age Mexican population with metabolic syndrome and type-2 diabetes mellitus using shotgun metagenomic sequencing. Front Pediatr 2023; 11:1193832. [PMID: 37342535 PMCID: PMC10277889 DOI: 10.3389/fped.2023.1193832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Accepted: 05/05/2023] [Indexed: 06/23/2023] Open
Abstract
Gut metagenome in pediatric subjects with metabolic syndrome (MetS) and type-2 diabetes mellitus (T2DM) has been poorly studied, despite an alarming worldwide increase in the prevalence and incidence of obesity and MetS within this population. The objective of this study was to characterize the gut microbiome taxonomic composition of Mexican pediatric subjects with MetS and T2DM using shotgun metagenomics and analyze the potential relationship with metabolic changes and proinflammatory effects. Paired-end reads of fecal DNA samples were obtained through the Illumina HiSeq X Platform. Statistical analyses and correlational studies were conducted using gut microbiome data and metadata from all individuals. Gut microbial dysbiosis was observed in MetS and T2DM children compared to healthy subjects, which was characterized by an increase in facultative anaerobes (i.e., enteric and lactic acid bacteria) and a decrease in strict anaerobes (i.e., Erysipelatoclostridium, Shaalia, and Actinomyces genera). This may cause a loss of gut hypoxic environment, increased gut microbial nitrogen metabolism, and higher production of pathogen-associated molecular patterns. These metabolic changes may trigger the activation of proinflammatory activity and impair the host's intermediate metabolism, leading to a possible progression of the characteristic risk factors of MetS and T2DM, such as insulin resistance, dyslipidemia, and an increased abdominal circumference. Furthermore, specific viruses (Jiaodavirus genus and Inoviridae family) showed positive correlations with proinflammatory cytokines involved in these metabolic diseases. This study provides novel evidence for the characterization of MetS and T2DM pediatric subjects in which the whole gut microbial composition has been characterized. Additionally, it describes specific gut microorganisms with functional changes that may influence the onset of relevant health risk factors.
Collapse
Affiliation(s)
| | - Oscar Tamez-Rivera
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Monterrey, Nuevo Leon, Mexico
| | - Ricardo García-Gamboa
- Tecnologico de Monterrey, Escuela de Ingenieria y Ciencias, Zapopan, Jalisco, Mexico
- Tecnologico de Monterrey, Escuela de Medicina, Colonia Nuevo México, Zapopan, Jalisco, México
| | - Tomás García-Cayuela
- Tecnologico de Monterrey, Escuela de Ingenieria y Ciencias, Zapopan, Jalisco, Mexico
| | - Nora A Rodríguez-Gutiérrez
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Monterrey, Nuevo Leon, Mexico
- Hospital Regional Materno Infantil de Alta Especialidad, Guadalupe, Nuevo Leon, Mexico
| | | | - Gerardo García-Rivas
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Monterrey, Nuevo Leon, Mexico
- Tecnologico de Monterrey, Institute for Obesity Research, Monterrey, Nuevo Leon, Mexico
| | - Adriana Pacheco
- Tecnologico de Monterrey, Escuela de Ingenieria y Ciencias, Monterrey, Nuevo Leon, Mexico
| | - Carmen Hernández-Brenes
- Tecnologico de Monterrey, Institute for Obesity Research, Monterrey, Nuevo Leon, Mexico
- Tecnologico de Monterrey, Escuela de Ingenieria y Ciencias, Monterrey, Nuevo Leon, Mexico
| | | |
Collapse
|
9
|
Deng Y, Jiang S, Huang Y, Tan X, Huang Y, Chen L, Xu J, Xiong X, Zhou J, Xu Y. Metformin Contributes to the Therapeutic Effects of Acne Vulgaris by Modifying the Gut Microbiome. Dermatol Ther 2023. [DOI: 10.1155/2023/9336867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/03/2023]
Abstract
Background. Considering the increasing side effects of the first-line treatment for acne vulgaris, metformin was developed to be an effective adjunct therapy, but its mechanism of action is poorly defined. Recent evidence shows that the gut microbiota is a site of metformin action. The aim of this study was to evaluate the effects and mechanism of action for metformin in the adjuvant treatment of acne vulgaris by regulating gut microbiota. Methods. First, untreated acne patients were randomly allocated into two treatment groups. Both groups were treated with isotretinoin, but only one was additionally treated with metformin, for three months. Sprague Dawley (SD) rats were used as acne models, and they were also separated into groups that received isotretinoin, metformin, a combination of isotretinoin and metformin, and the vehicle, respectively. Then, the fecal samples from drug-intervention rats were transferred to germ-free rats with acne. The severity of the disease was evaluated using the Global Acne Grading System (GAGS) scoring for patients, and the number of comedones and mononuclear cells in pathological sections was used for rats. The composition of the gut microbiota was detected using gene sequencing for 16S rDNA. Results. Metformin had strong effects on the composition and function of the gut microbiota, and this correlated with the reduction in the severity of acne in both humans and rats. The fecal transfer to pseudo-germ-free rats improved both the inflammatory phenotype and comedones of acne in recipients of metformin-altered microbiota. Conclusion. The results suggest that metformin improves the symptoms of acne vulgaris by modulating the gut microbiota.
Collapse
|
10
|
Jia L, Huang S, Sun B, Shang Y, Zhu C. Pharmacomicrobiomics and type 2 diabetes mellitus: A novel perspective towards possible treatment. Front Endocrinol (Lausanne) 2023; 14:1149256. [PMID: 37033254 PMCID: PMC10076675 DOI: 10.3389/fendo.2023.1149256] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Accepted: 03/14/2023] [Indexed: 04/11/2023] Open
Abstract
Type 2 diabetes mellitus (T2DM), a major driver of mortality worldwide, is more likely to develop other cardiometabolic risk factors, ultimately leading to diabetes-related mortality. Although a set of measures including lifestyle intervention and antidiabetic drugs have been proposed to manage T2DM, problems associated with potential side-effects and drug resistance are still unresolved. Pharmacomicrobiomics is an emerging field that investigates the interactions between the gut microbiome and drug response variability or drug toxicity. In recent years, increasing evidence supports that the gut microbiome, as the second genome, can serve as an attractive target for improving drug efficacy and safety by manipulating its composition. In this review, we outline the different composition of gut microbiome in T2DM and highlight how these microbiomes actually play a vital role in its development. Furthermore, we also investigate current state-of-the-art knowledge on pharmacomicrobiomics and microbiome's role in modulating the response to antidiabetic drugs, as well as provide innovative potential personalized treatments, including approaches for predicting response to treatment and for modulating the microbiome to improve drug efficacy or reduce drug toxicity.
Collapse
Affiliation(s)
- Liyang Jia
- Department of Traditional Chinese Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Shiqiong Huang
- Department of Pharmacy, The First Hospital of Changsha, Changsha, China
| | - Boyu Sun
- Department of Pharmacy, The Third People’s Hospital of Qingdao, Qingdao, China
| | - Yongguang Shang
- Department of Pharmacy, China-Japan Friendship Hospital, Beijing, China
- *Correspondence: Yongguang Shang, ; Chunsheng Zhu,
| | - Chunsheng Zhu
- Department of Traditional Chinese Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- *Correspondence: Yongguang Shang, ; Chunsheng Zhu,
| |
Collapse
|
11
|
Liu W, Luo Z, Zhou J, Sun B. Gut Microbiota and Antidiabetic Drugs: Perspectives of Personalized Treatment in Type 2 Diabetes Mellitus. Front Cell Infect Microbiol 2022; 12:853771. [PMID: 35711668 PMCID: PMC9194476 DOI: 10.3389/fcimb.2022.853771] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 05/04/2022] [Indexed: 12/23/2022] Open
Abstract
Alterations in the composition and function of the gut microbiota have been reported in patients with type 2 diabetes mellitus (T2DM). Emerging studies show that prescribed antidiabetic drugs distort the gut microbiota signature associated with T2DM. Even more importantly, accumulated evidence provides support for the notion that gut microbiota, in turn, mediates the efficacy and safety of antidiabetic drugs. In this review, we highlight the current state-of-the-art knowledge on the crosstalk and interactions between gut microbiota and antidiabetic drugs, including metformin, α-glucosidase inhibitors, glucagon-like peptide-1 receptor agonists, dipeptidyl peptidase-4 inhibitors, sodium-glucose cotransporter 2 inhibitors, traditional Chinese medicines and other antidiabetic drugs, as well as address corresponding microbial-based therapeutics, aiming to provide novel preventative strategies and personalized therapeutic targets in T2DM.
Collapse
Affiliation(s)
- Wenhui Liu
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
- Institute of Clinical Pharmacy, Central South University, Changsha, China
| | - Zhiying Luo
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
- Institute of Clinical Pharmacy, Central South University, Changsha, China
| | - Jiecan Zhou
- Institute of Clinical Medicine, The First Affiliated Hospital, University of South China, Hengyang, China
| | - Bao Sun
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
- Institute of Clinical Pharmacy, Central South University, Changsha, China
- *Correspondence: Bao Sun,
| |
Collapse
|
12
|
Peng X, Huang Y, Wang G, He Y, Hu L, Fang Z, Lin Y, Xu S, Feng B, Li J, Tang J, Hua L, Jiang X, Zhuo Y, Che L, Wu D. Maternal Long-Term Intake of Inulin Improves Fetal Development through Gut Microbiota and Related Metabolites in a Rat Model. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2022; 70:1840-1851. [PMID: 35129337 DOI: 10.1021/acs.jafc.1c07284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Adequate dietary fiber intake during gestation is critical for maternal-fetal health. This experiment aims to uncover the impacts of maternal long-term intake of inulin on fetal development and its underlying mechanism. Eighty female Sprague-Dawley rats were randomly assigned to two groups receiving either a fiber-free diet or an inulin diet (inulin) for three parities. On the 19th day of pregnancy in the third parity, blood, intestinal, placental, and colonic digesta samples were collected. Results showed that maternal intake of inulin significantly decreased the within-litter birth weight variation in parities 2 and 3. Inulin intake modified the gut microbiome profiles and elevated the colonic contents of short chain fatty acids (propionate and butyrate). Inulin decreased the serotonin (5-HT) concentration in the colon, whereas it increased the 5-HT concentrations in serum and placenta and the number of 5-HT+ enterochromaffin cells in the colon. The protein expression of melatonin-synthesizing enzyme (arylalkylamine N-acetyltransferase) and the melatonin concentration in the placenta were also increased by inulin. Inulin improved the placental redox status and nutrient transport. These findings indicated that maternal long-term intake of inulin improves fetal development by altering the intestinal microbiota and related metabolites in rats.
Collapse
Affiliation(s)
- Xie Peng
- Key Laboratory for Animal Disease Resistant Nutrition of the Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China
| | - Yingyan Huang
- Key Laboratory for Animal Disease Resistant Nutrition of the Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China
| | - Guixiang Wang
- Key Laboratory for Animal Disease Resistant Nutrition of the Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China
| | - Ying He
- Key Laboratory for Animal Disease Resistant Nutrition of the Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China
| | - Liang Hu
- College of Food Science, Sichuan Agricultural University, Ya'an 625014, China
| | - Zhengfeng Fang
- Key Laboratory for Animal Disease Resistant Nutrition of the Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China
| | - Yan Lin
- Key Laboratory for Animal Disease Resistant Nutrition of the Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China
| | - Shengyu Xu
- Key Laboratory for Animal Disease Resistant Nutrition of the Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China
| | - Bin Feng
- Key Laboratory for Animal Disease Resistant Nutrition of the Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China
| | - Jian Li
- Key Laboratory for Animal Disease Resistant Nutrition of the Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China
| | - Jiayong Tang
- Key Laboratory for Animal Disease Resistant Nutrition of the Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China
| | - Lun Hua
- Key Laboratory for Animal Disease Resistant Nutrition of the Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China
| | - Xuemei Jiang
- Key Laboratory for Animal Disease Resistant Nutrition of the Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China
| | - Yong Zhuo
- Key Laboratory for Animal Disease Resistant Nutrition of the Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China
| | - Lianqiang Che
- Key Laboratory for Animal Disease Resistant Nutrition of the Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China
| | - De Wu
- Key Laboratory for Animal Disease Resistant Nutrition of the Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China
| |
Collapse
|
13
|
Craciun CI, Neag MA, Catinean A, Mitre AO, Rusu A, Bala C, Roman G, Buzoianu AD, Muntean DM, Craciun AE. The Relationships between Gut Microbiota and Diabetes Mellitus, and Treatments for Diabetes Mellitus. Biomedicines 2022; 10:308. [PMID: 35203519 PMCID: PMC8869176 DOI: 10.3390/biomedicines10020308] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 01/15/2022] [Accepted: 01/26/2022] [Indexed: 02/04/2023] Open
Abstract
Diabetes mellitus is considered to be a global epidemic. The combination of genetic susceptibility and an unhealthy lifestyle is considered to be the main trigger of this metabolic disorder. Recently, there has been increased interest in the roles of gut microbiota as a new potential contributor to this epidemic. Research, in recent years, has contributed to an in-depth characterization of the human microbiome and its associations with various diseases, including metabolic diseases and diabetes mellitus. It is known that diet can change the composition of gut microbiota, but it is unclear how this, in turn, may influence metabolism. The main objective of this review is to evaluate the pathogenetic association between microbiota and diabetes and to explore any new therapeutic agents, including nutraceuticals that may modulate the microbiota. We also look at several mechanisms involved in this process. There is a clear, bidirectional relationship between microbiota and diabetes. Current treatments for diabetes influence microbiota in various ways, some beneficial, but others with still unclear effects. Microbiota-aimed treatments have seen no real-world significant effects on the progression of diabetes and its complications, with more studies needed in order to find a really beneficial agent.
Collapse
Affiliation(s)
- Cristian-Ioan Craciun
- Department of Pharmacology, Toxicology and Clinical Pharmacology, Iuliu Hatieganu University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania; (C.-I.C.); (A.-D.B.)
| | - Maria-Adriana Neag
- Department of Pharmacology, Toxicology and Clinical Pharmacology, Iuliu Hatieganu University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania; (C.-I.C.); (A.-D.B.)
| | - Adrian Catinean
- Department of Internal Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 400006 Cluj-Napoca, Romania;
| | - Andrei-Otto Mitre
- Faculty of Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania;
| | - Adriana Rusu
- Department of Diabetes, Nutrition, Metabolic Diseases, Iuliu Hatieganu University of Medicine and Pharmacy, 400006 Cluj-Napoca, Romania; (A.R.); (C.B.); (G.R.); (A.-E.C.)
| | - Cornelia Bala
- Department of Diabetes, Nutrition, Metabolic Diseases, Iuliu Hatieganu University of Medicine and Pharmacy, 400006 Cluj-Napoca, Romania; (A.R.); (C.B.); (G.R.); (A.-E.C.)
| | - Gabriela Roman
- Department of Diabetes, Nutrition, Metabolic Diseases, Iuliu Hatieganu University of Medicine and Pharmacy, 400006 Cluj-Napoca, Romania; (A.R.); (C.B.); (G.R.); (A.-E.C.)
| | - Anca-Dana Buzoianu
- Department of Pharmacology, Toxicology and Clinical Pharmacology, Iuliu Hatieganu University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania; (C.-I.C.); (A.-D.B.)
| | - Dana-Maria Muntean
- Department of Pharmaceutical Technology and Biopharmaceutics, Iuliu Hatieganu University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania;
| | - Anca-Elena Craciun
- Department of Diabetes, Nutrition, Metabolic Diseases, Iuliu Hatieganu University of Medicine and Pharmacy, 400006 Cluj-Napoca, Romania; (A.R.); (C.B.); (G.R.); (A.-E.C.)
| |
Collapse
|
14
|
Chen Y, Wang M. New Insights of Anti-Hyperglycemic Agents and Traditional Chinese Medicine on Gut Microbiota in Type 2 Diabetes. Drug Des Devel Ther 2021; 15:4849-4863. [PMID: 34876807 PMCID: PMC8643148 DOI: 10.2147/dddt.s334325] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Accepted: 11/19/2021] [Indexed: 12/11/2022] Open
Abstract
Type 2 diabetes mellitus (T2DM) is a widespread metabolic disease characterized by chronic hyperglycemia. Human microbiota, which is regarded as a “hidden organ”, plays an important role in the initiation and development of T2DM. In addition, anti-hyperglycemic agents and traditional Chinese medicine may affect the composition of gut microbiota and consequently improve glucose metabolism. However, the relationship between gut microbiota, T2DM and anti-hyperglycemic agents or traditional Chinese medicine is poorly understood. In this review, we summarized pre-clinical and clinical studies to elucidate the possible underlying mechanism. Some anti-hyperglycemic agents and traditional Chinese medicine may partly exert hypoglycemic effects by altering the gut microbiota composition in ways that reduce metabolic endotoxemia, maintain the integrity of intestinal mucosal barrier, promote the production of short-chain fatty acids (SCFAs), decrease trimethylamine-N-oxide (TMAO) and regulate bile acid metabolism. In conclusion, gut microbiota may provide some new therapeutic targets for treatment of patients with diabetes mellitus.
Collapse
Affiliation(s)
- Yanxia Chen
- Department of Endocrinology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050051, People's Republic of China
| | - Mian Wang
- Department of Endocrinology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050051, People's Republic of China
| |
Collapse
|
15
|
Díaz-Perdigones CM, Muñoz-Garach A, Álvarez-Bermúdez MD, Moreno-Indias I, Tinahones FJ. Gut microbiota of patients with type 2 diabetes and gastrointestinal intolerance to metformin differs in composition and functionality from tolerant patients. Biomed Pharmacother 2021; 145:112448. [PMID: 34844104 DOI: 10.1016/j.biopha.2021.112448] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Revised: 10/30/2021] [Accepted: 11/16/2021] [Indexed: 12/12/2022] Open
Abstract
OBJECTIVE Metformin modifies the gut microbiome in type 2 diabetes and gastrointestinal tolerance to metformin could be mediated by the gut microbiome. METHODS We enrolled 35 patients with type 2 diabetes not receiving treatment with metformin due to suspected gastrointestinal intolerance. Metformin was reintroduced at 425 mg, increasing 425 mg every two weeks until reaching 1700 mg per day. According to the occurrence of metformin-related gastrointestinal symptoms, patients were classified into three groups: early intolerance, non-tolerant, and tolerant. Gut microbiota was profiled with 16 S rRNA. This sequencing aimed to determine the differences in the baseline gut microbiota in all groups and prospectively in the tolerant and non-tolerant groups. RESULTS The classification resulted in 15 early intolerant, 10 tolerant, and 10 non-tolerant subjects. Early tolerance was characterized by a higher abundance of Subdoligranulum; while Veillonella and Serratia were higher in the non-tolerant group. The tolerant group showed enrichment of Megamonas, Megamonas rupellensis, and Phascolarctobacterium spp; Ruminococcus gnavus was lower in the longitudinal analysis. At the end point Prevotellaceae, Prevotella stercorea, Megamonas funiformis, Bacteroides xylanisolvens, and Blautia producta had a higher relative abundance in the tolerant group compared to the non-tolerant group. Subdoligranulum, Ruminococcus torques_1, Phascolarctobacterium faecium, and Eubacterium were higher in the non-tolerant group. The PICRUSt analysis showed a lower activity of the amino acid biosynthesis pathways and a higher sugar degradation pathway in the intolerant groups. CONCLUSIONS Gut microbiota of subjects with gastrointestinal intolerance depicted taxonomic and functional differences compared to tolerant patients, and this changed differently after metformin administration.
Collapse
Affiliation(s)
- Cristina Ma Díaz-Perdigones
- Clinical Management Unit of Endocrinology and Nutrition, Laboratory of the Biomedical Research Institute of Málaga, Virgen de la Victoria University Hospital, Universidad de Málaga, Málaga, Spain; University of Málaga, Program in Biomedicine, Translational Research and New Technologies, Spain
| | - Araceli Muñoz-Garach
- Clinical Management Unit of Endocrinology and Nutrition, Laboratory of the Biomedical Research Institute of Málaga, Virgen de la Victoria University Hospital, Universidad de Málaga, Málaga, Spain
| | - María Dolores Álvarez-Bermúdez
- Clinical Management Unit of Endocrinology and Nutrition, Laboratory of the Biomedical Research Institute of Málaga, Virgen de la Victoria University Hospital, Universidad de Málaga, Málaga, Spain; University of Málaga, Program in Biomedicine, Translational Research and New Technologies, Spain; Centro de Investigación Biomédica en Red (CIBER) de Fisiopatología de la Obesidad y Nutrición, Instituto Salud Carlos III, Madrid, Spain
| | - Isabel Moreno-Indias
- Clinical Management Unit of Endocrinology and Nutrition, Laboratory of the Biomedical Research Institute of Málaga, Virgen de la Victoria University Hospital, Universidad de Málaga, Málaga, Spain; Centro de Investigación Biomédica en Red (CIBER) de Fisiopatología de la Obesidad y Nutrición, Instituto Salud Carlos III, Madrid, Spain.
| | - Francisco J Tinahones
- Clinical Management Unit of Endocrinology and Nutrition, Laboratory of the Biomedical Research Institute of Málaga, Virgen de la Victoria University Hospital, Universidad de Málaga, Málaga, Spain; University of Málaga, Program in Biomedicine, Translational Research and New Technologies, Spain; Centro de Investigación Biomédica en Red (CIBER) de Fisiopatología de la Obesidad y Nutrición, Instituto Salud Carlos III, Madrid, Spain.
| |
Collapse
|
16
|
Fan P, Kim M, Liu G, Zhai Y, Liu T, Driver JD, Jeong KC. The Gut Microbiota of Newborn Calves and Influence of Potential Probiotics on Reducing Diarrheic Disease by Inhibition of Pathogen Colonization. Front Microbiol 2021; 12:772863. [PMID: 34745079 PMCID: PMC8567051 DOI: 10.3389/fmicb.2021.772863] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 09/30/2021] [Indexed: 01/17/2023] Open
Abstract
Calf diarrhea is one of the most concerning challenges facing both the dairy and beef cattle industry. Maintaining healthy gut microbiota is essential for preventing gastrointestinal disorders. Here, we observed significantly less bacterial richness in the abnormal feces with watery or hemorrhagic morphology compared to the normal solid feces. The normal solid feces showed high relative abundances of Osllospiraceae, Christensenellaceae, Barnesiella, and Lactobacillus, while the abnormal feces contained more bacterial taxa of Negativicutes, Tyzzerella, Parasutterella, Veillonella, Fusobacterium, and Campylobacter. Healthy calves had extensive bacterial-bacterial correlations, with negative correlation between Lactobacillus and potential diarrheagenic Escherichia coli-Shigella, but not in the abnormal feces. We isolated Lactobacillus species (L. reuteri, L. johnsonii, L. amylovorus, and L. animalis), with L. reuteri being the most abundant, from the healthy gut microbiota. Isolated Lactobacillus strains inhibited pathogenic strains including E. coli K88 and Salmonella Typhimurium. These findings indicate the importance of a diverse gut microbiota in newborn calf’s health and provide multiple potential probiotics that suppress pathogen colonization in the gastrointestinal tract to prevent calf diarrhea.
Collapse
Affiliation(s)
- Peixin Fan
- Emerging Pathogens Institute, University of Florida, Gainesville, FL, United States.,Department of Animal Sciences, University of Florida, Gainesville, FL, United States
| | - Miju Kim
- Emerging Pathogens Institute, University of Florida, Gainesville, FL, United States.,Department of Animal Sciences, University of Florida, Gainesville, FL, United States
| | - Grace Liu
- Emerging Pathogens Institute, University of Florida, Gainesville, FL, United States.,Department of Animal Sciences, University of Florida, Gainesville, FL, United States
| | - Yuting Zhai
- Emerging Pathogens Institute, University of Florida, Gainesville, FL, United States.,Department of Animal Sciences, University of Florida, Gainesville, FL, United States
| | - Ting Liu
- Emerging Pathogens Institute, University of Florida, Gainesville, FL, United States.,Department of Animal Sciences, University of Florida, Gainesville, FL, United States
| | - Joseph Danny Driver
- Department of Animal Sciences, University of Florida, Gainesville, FL, United States
| | - Kwangcheol C Jeong
- Emerging Pathogens Institute, University of Florida, Gainesville, FL, United States.,Department of Animal Sciences, University of Florida, Gainesville, FL, United States
| |
Collapse
|
17
|
Multi-Omics Analysis to Generate Hypotheses for Mild Health Problems in Monkeys. Metabolites 2021; 11:metabo11100701. [PMID: 34677416 PMCID: PMC8538200 DOI: 10.3390/metabo11100701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 09/28/2021] [Accepted: 10/09/2021] [Indexed: 11/17/2022] Open
Abstract
Certain symptoms associated with mild sickness and lethargy have not been categorized as definitive diseases. Confirming such symptoms in captive monkeys (Macaca fascicularis, known as cynomolgus monkeys) can be difficult; however, it is possible to observe and analyze their feces. In this study, we investigated the relationship between stool state and various omics data by considering objective and quantitative values of stool water content as a phenotype for analysis. By examining the food intake of the monkeys and assessing their stool, urine, and plasma, we attempted to obtain a comprehensive understanding of the health status of individual monkeys and correlate it with the stool condition. Our metabolomics data strongly suggested that many lipid-related metabolites were correlated with the stool water content. The lipidomic analysis revealed the involvement of saturated and oxidized fatty acids, metallomics revealed the contribution of selenium (a bio-essential trace element), and intestinal microbiota analysis revealed the association of several bacterial species with the stool water content. Based on our results, we hypothesize that the redox imbalance causes minor health problems. However, it is not possible to make a definite conclusion using multi-omics alone, and other hypotheses could be proposed.
Collapse
|
18
|
Shoshan-Barmatz V, Anand U, Nahon-Crystal E, Di Carlo M, Shteinfer-Kuzmine A. Adverse Effects of Metformin From Diabetes to COVID-19, Cancer, Neurodegenerative Diseases, and Aging: Is VDAC1 a Common Target? Front Physiol 2021; 12:730048. [PMID: 34671273 PMCID: PMC8521008 DOI: 10.3389/fphys.2021.730048] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Accepted: 08/24/2021] [Indexed: 12/13/2022] Open
Abstract
Metformin has been used for treating diabetes mellitus since the late 1950s. In addition to its antihyperglycemic activity, it was shown to be a potential drug candidate for treating a range of other diseases that include various cancers, cardiovascular diseases, diabetic kidney disease, neurodegenerative diseases, renal diseases, obesity, inflammation, COVID-19 in diabetic patients, and aging. In this review, we focus on the important aspects of mitochondrial dysfunction in energy metabolism and cell death with their gatekeeper VDAC1 (voltage-dependent anion channel 1) as a possible metformin target, and summarize metformin's effects in several diseases and gut microbiota. We question how the same drug can act on diseases with opposite characteristics, such as increasing apoptotic cell death in cancer, while inhibiting it in neurodegenerative diseases. Interestingly, metformin's adverse effects in many diseases all show VDAC1 involvement, suggesting that it is a common factor in metformin-affecting diseases. The findings that metformin has an opposite effect on various diseases are consistent with the fact that VDAC1 controls cell life and death, supporting the idea that it is a target for metformin.
Collapse
Affiliation(s)
- Varda Shoshan-Barmatz
- Department of Life Sciences, Ben-Gurion University of the Negev, Beersheba, Israel
- National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beersheba, Israel
| | - Uttpal Anand
- Department of Life Sciences, Ben-Gurion University of the Negev, Beersheba, Israel
| | | | - Marta Di Carlo
- Institute for Biomedical Research and Innovation, National Research Council, Palermo, Italy
| | - Anna Shteinfer-Kuzmine
- National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beersheba, Israel
| |
Collapse
|
19
|
Balvers M, van den Born BJH, Levin E, Nieuwdorp M. Impact drugs targeting cardiometabolic risk on the gut microbiota. Curr Opin Lipidol 2021; 32:38-54. [PMID: 33332920 DOI: 10.1097/mol.0000000000000727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
PURPOSE OF REVIEW Alterations in the gut microbiome composition or function are associated with risk factors for cardiometabolic diseases, including hypertension, hyperlipidemia and hyperglycemia. Based on recent evidence that also oral medications used to treat these conditions could alter the gut microbiome composition and function and, vice versa, that the gut microbiome could affect the efficacy of these treatments, we reviewed the literature on these observed interactions. RECENT FINDINGS While the interaction of metformin with the gut microbiome has been studied most, other drugs that target cardiometabolic risk are gaining attention and often showed associations with alterations in microbiome-related features, including alterations in specific microbial taxa or pathways, microbiome composition or microbiome-derived metabolites, while the gut microbiome was also involved in drug metabolism and drug efficacy. As for metformin, for some of them even a potential therapeutic effect via the gut microbiome is postulated. However, exact mechanisms remain to be elucidated. SUMMARY There is growing interest in clarifying the interactions between the gut microbiome and drugs to treat hypertension, hyperlipidemia and hyperglycemia as well as the first pass effect of microbiome on drug efficacy. While mostly analysed in animal models, also human studies are gaining more and more traction. Improving the understanding of the gut microbiome drug interaction can provide clinical directions for therapy by optimizing drug efficacy or providing new targets for drug development.
Collapse
Affiliation(s)
- Manon Balvers
- Department of Internal and Vascular Medicine, Amsterdam UMC, Location AMC, University of Amsterdam, Amsterdam
- Horaizon BV, Delft
| | - Bert-Jan H van den Born
- Department of Internal and Vascular Medicine, Amsterdam UMC, Location AMC, University of Amsterdam, Amsterdam
- Department of Public Health, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Evgeni Levin
- Department of Internal and Vascular Medicine, Amsterdam UMC, Location AMC, University of Amsterdam, Amsterdam
- Horaizon BV, Delft
| | - Max Nieuwdorp
- Department of Internal and Vascular Medicine, Amsterdam UMC, Location AMC, University of Amsterdam, Amsterdam
| |
Collapse
|
20
|
Huang QY, Yao F, Zhou CR, Huang XY, Wang Q, Long H, Wu QM. Role of gut microbiome in regulating the effectiveness of metformin in reducing colorectal cancer in type 2 diabetes. World J Clin Cases 2020; 8:6213-6228. [PMID: 33392303 PMCID: PMC7760447 DOI: 10.12998/wjcc.v8.i24.6213] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 10/13/2020] [Accepted: 11/02/2020] [Indexed: 02/05/2023] Open
Abstract
The prevalence of colorectal cancer (CRC) and type 2 diabetes mellitus (T2DM) is increasing globally. It is rarely noticed that the incidence of CRC is higher in patients with T2DM. What needs to be mentioned is that metformin, a commonly used clinical drug for T2DM, attracts scholars’ attention because of its benefits in lowering the risk of developing CRC. Hence, we try to find the common grounds of initiation of T2DM and CRC and the reason why metformin reduces the risk of CRC in patients with T2DM. We noticed consistent changes of gut microbiota, such as elevated Bacteroides, Prevotella and Bifidobacterium and depressed Firmicutes and Lactobacillus. Furthermore, many studies in recent years have proved that the efficacy of metformin, such as improving blood glucose, depends on the gut microbiota. Coincidentally, the progression of CRC is inseparable from the contributions of gut microbiota. Therefore, we first proposed the concept of the metformin-gut microbiota–CRC (in T2DM) axis to explain the effect of metformin in reducing CRC in patients with T2DM. In this review, we elaborated the new concept and its potential clinical application value.
Collapse
Affiliation(s)
- Qi-You Huang
- Institute of Infection, Immunology and Tumor Microenvironment, Medical College, Wuhan University of Science and Technology, Wuhan 430065, Hubei Province, China
| | - Fei Yao
- Institute of Infection, Immunology and Tumor Microenvironment, Medical College, Wuhan University of Science and Technology, Wuhan 430065, Hubei Province, China
| | - Chuan-Ren Zhou
- Institute of Infection, Immunology and Tumor Microenvironment, Medical College, Wuhan University of Science and Technology, Wuhan 430065, Hubei Province, China
| | - Xiao-Ying Huang
- Institute of Infection, Immunology and Tumor Microenvironment, Medical College, Wuhan University of Science and Technology, Wuhan 430065, Hubei Province, China
| | - Qiang Wang
- Institute of Infection, Immunology and Tumor Microenvironment, Medical College, Wuhan University of Science and Technology, Wuhan 430065, Hubei Province, China
| | - Hui Long
- Department of Gastroenterology, Tianyou Affiliated Hospital, Wuhan University of Science and Technology, Wuhan 430064, Hubei Province, China
| | - Qing-Ming Wu
- Institute of Infection, Immunology and Tumor Microenvironment, Medical College, Wuhan University of Science and Technology, Wuhan 430065, Hubei Province, China
| |
Collapse
|