Microbiota Implications in Endocrine-Related Diseases: From Development to Novel Therapeutic Approaches
Abstract
:1. Introduction to the Human Microbiota and Its Composition
2. Overview of the Endocrine System and Its Role in Maintaining Homeostasis
3. The Emerging Concept of Microbiota-Endocrine System Interactions
3.1. Overview
3.2. Behavior and Emotional Regulation
3.3. Sex Hormones
3.4. Appetite
4. Microbiota Dysbiosis and Its Implications in Endocrine-Related Diseases
4.1. Overview
4.2. Diabetes and Obesity
4.3. Thyroid Disorders
4.4. Estrogens-Related Conditions
4.5. Bone Health
5. Mechanisms by Which Microbiota Can Influence Hormone Regulation
Gut, Hormonal Dysregulation, and Health
6. Microbiota’s Impact on Endocrine-Related Cancers
6.1. Tumor Development
6.2. Dysbiosis and Types of Cancer
6.3. Therapeutic Interventions
7. Clinical Manifestations of Microbiota-Related Endocrine Disorders
7.1. Overview
7.2. Metabolic Disorders Manifestations
7.3. Hormonal Dysregulation Manifestations
7.4. Osteoporosis Manifestations
8. Diagnostic Tools and Biomarkers for Assessing Microbiota-Endocrine Interactions
8.1. Overview
8.2. Metagenomic Sequencing
8.3. Metabolics and Proteomics
9. Current Therapeutic Approaches Targeting Microbiota-Endocrine Crosstalk
9.1. Overview
9.2. Pre and Probiotics
9.3. Diet
9.4. Fecal Transplant
10. Challenges and Limitations in Studying Microbiota Endocrine Interactions
- –
- Microbiota Complexity: The microbiota’s immense diversity poses a significant challenge.
- –
- Individual Variability: Microbiota variability among individuals is influenced by genetics, diet, environment, and lifestyle.
- –
- Correlational Studies Dominance: The majority of research relies on correlational studies, lacking causal establishment.
- –
- Dynamic Nature of Microbiota: The microbiota’s dynamic nature is changing rapidly in response to various factors.
- –
- Analytical Tools and Methodology: Need for more advanced analytical tools and methods
- –
- Promising yet Challenging Research Field: Despite the promising nature of microbiota-endocrine research
11. Future Directions for Research in this Field
11.1. Microbiota-Thyroid Research
11.2. Focus on the Interrelationship between Gut Bacteria and Hormone Control
11.3. Gut Microbes and Endocrine System Connections
11.4. Future Research Emphasis
- –
- Microbiota-Thyroid Axis: A New Frontier: The connections between gut health and thyroid disorders, emphasizing the potential of advanced analytical techniques like metabolomics and bioinformatics.
- –
- Interconnectedness of Gut Microbiota and Hormone Control: An exploration of the interplay between gut microbiota and sex hormones offers insights into reproductive health with potential health implications.
- –
- Comprehensive Investigation of the Microbiota-Endocrine Network: Research in this direction could unravel the complexity of how the gut microbiota influences multiple endocrine axes simultaneously.
12. Conclusions
- Understanding Microbiota’s Impact on Hormone Regulation:
- –
- Patient Education: Providing patients with detailed information about how the microbiota influences hormone regulation helps them understand the interconnected nature of their body systems.
- –
- Behavioral Changes: Encouraging patients to adopt lifestyle changes that promote healthy microbiota, such as a balanced diet, regular exercise, and stress management.
- Role of Microbiota in Endocrine Pathologies:
- –
- Early Detection and Monitoring: Emphasizing regular check-ups and tests to detect any alterations in the microbiota associated with endocrine pathologies, enabling early intervention.
- –
- Tailored Treatment Plans: Developing personalized treatment plans that consider the patient’s microbiota profile alongside traditional medical approaches for better outcomes.
- Microbiota Modulation Through Diet:
- –
- Nutritional Counseling: Providing personalized nutritional counseling based on the patient’s microbiota composition to optimize their diet for improved endocrine health.
- –
- Incorporating Probiotic-Rich Foods: Encouraging the inclusion of probiotic-rich foods, such as yogurt and fermented products, to naturally support a healthy microbiota.
- Probiotics and Prebiotics:
- –
- Supplementation Strategies: Recommending specific probiotic and prebiotic supplements tailored to the patient’s microbiota needs, potentially improving microbial diversity and function.
- –
- Educating Sources: Informing patients about the natural sources of probiotics and prebiotics in food enables them to make informed choices for their gut health.
- Fecal Microbiota Transplantation (FMT):
- –
- Consideration in Treatment Plans: Evaluating the potential benefits of FMT for specific endocrine conditions and incorporating it into treatment plans when deemed appropriate.
- –
- Ensuring Safety and Regulation: Ensuring that FMT procedures adhere to safety standards and regulatory guidelines to minimize risks and optimize therapeutic outcomes.
- Holistic Patient-Centric Approach:
- –
- Integration with Conventional Treatments: Integrating microbiota-focused interventions with conventional medical treatments to provide a comprehensive and holistic approach.
- –
- Patient Empowerment: Empowering patients to actively participate in their healthcare by making informed decisions about their diet, lifestyle, and treatment choices based on their microbiota profile.
Author Contributions
Funding
Acknowledgments
Conflicts of Interest
References
- Thursby, E.; Juge, N. Introduction to the Human Gut Microbiota. Biochem. J. 2017, 474, 1823–1836. [Google Scholar] [CrossRef] [PubMed]
- Clapp, M.; Aurora, N.; Herrera, L.; Bhatia, M.; Wilen, E.; Wakefield, S. Gut Microbiota’s Effect on Mental Health: The Gut-Brain Axis. Clin. Pract. 2017, 7, 987. [Google Scholar] [CrossRef] [PubMed]
- Gibbons, S.M.; Gilbert, J.A. Microbial Diversity-Exploration of Natural Ecosystems and Microbiomes. Curr. Opin. Genet. Dev. 2015, 35, 66–72. [Google Scholar] [CrossRef] [PubMed]
- Cahana, I.; Iraqi, F.A. Impact of Host Genetics on Gut Microbiome: Take-Home Lessons from Human and Mouse Studies. Anim. Models Exp. Med. 2020, 3, 229–236. [Google Scholar] [CrossRef] [PubMed]
- Dominguez-Bello, M.G.; Costello, E.K.; Contreras, M.; Magris, M.; Hidalgo, G.; Fierer, N.; Knight, R. Delivery Mode Shapes the Acquisition and Structure of the Initial Microbiota across Multiple Body Habitats in Newborns. Proc. Natl. Acad. Sci. USA 2010, 107, 11971–11975. [Google Scholar] [CrossRef] [PubMed]
- Ecklu-Mensah, G.; Gilbert, J.; Devkota, S. Dietary Selection Pressures and Their Impact on the Gut Microbiome. Cell. Mol. Gastroenterol. Hepatol. 2022, 13, 7–18. [Google Scholar] [CrossRef]
- Conlon, M.A.; Bird, A.R. The Impact of Diet and Lifestyle on Gut Microbiota and Human Health. Nutrients 2015, 7, 17–44. [Google Scholar] [CrossRef]
- Ayariga, J.A.; Ibrahim, I.; Gildea, L.; Abugri, J.; Villafane, R. Microbiota in a Long Survival Discourse with the Human Host. Arch. Microbiol. 2023, 205, 5. [Google Scholar] [CrossRef]
- Rowland, I.; Gibson, G.; Heinken, A.; Scott, K.; Swann, J.; Thiele, I.; Tuohy, K. Gut Microbiota Functions: Metabolism of Nutrients and Other Food Components. Eur. J. Nutr. 2018, 57, 1–24. [Google Scholar] [CrossRef] [PubMed]
- Degruttola, A.K.; Low, D.; Mizoguchi, A.; Mizoguchi, E. Current Understanding of Dysbiosis in Disease in Human and Animal Models. Inflamm. Bowel Dis. 2016, 22, 1137–1150. [Google Scholar] [CrossRef] [PubMed]
- Margolis, K.G.; Cryan, J.F.; Mayer, E.A. The Microbiota-Gut-Brain Axis: From Motility to Mood. Gastroenterology 2021, 160, 1486–1501. [Google Scholar] [CrossRef]
- Wu, J.; Wang, K.; Wang, X.; Pang, Y.; Jiang, C. The Role of the Gut Microbiome and Its Metabolites in Metabolic Diseases. Protein Cell 2021, 12, 360–373. [Google Scholar] [CrossRef] [PubMed]
- Johnson, J.S.; Spakowicz, D.J.; Hong, B.Y.; Petersen, L.M.; Demkowicz, P.; Chen, L.; Leopold, S.R.; Hanson, B.M.; Agresta, H.O.; Gerstein, M.; et al. Evaluation of 16S RRNA Gene Sequencing for Species and Strain-Level Microbiome Analysis. Nat. Commun. 2019, 10, 5029. [Google Scholar] [CrossRef]
- Maan, K.; Baghel, R.; Dhariwal, S.; Sharma, A.; Bakhshi, R.; Rana, P. Metabolomics and Transcriptomics Based Multi-Omics Integration Reveals Radiation-Induced Altered Pathway Networking and Underlying Mechanism. NPJ Syst. Biol. Appl. 2023, 9, 42. [Google Scholar] [CrossRef] [PubMed]
- Natarajan, A.; Bhatt, A.S. Microbes and Microbiomes in 2020 and Beyond. Nat. Commun. 2020, 11, 4988. [Google Scholar] [CrossRef] [PubMed]
- Ko, K.K.K.; Chng, K.R.; Nagarajan, N. Metagenomics-Enabled Microbial Surveillance. Nat. Microbiol. 2022, 7, 486–496. [Google Scholar] [CrossRef] [PubMed]
- Suzuki, T.A.; Ley, R.E. The Role of the Microbiota in Human Genetic Adaptation. Science 2020, 370, 1180. [Google Scholar] [CrossRef] [PubMed]
- Takiishi, T.; Fenero, C.I.M.; Câmara, N.O.S. Intestinal Barrier and Gut Microbiota: Shaping Our Immune Responses throughout Life. Tissue Barriers 2017, 5, e1373208. [Google Scholar] [CrossRef]
- Goenka, A.; Kollmann, T.R. Development of Immunity in Early Life. J. Infect. 2015, 71, S112–S120. [Google Scholar] [CrossRef]
- Uzbay, T. Germ-Free Animal Experiments in the Gut Microbiota Studies. Curr. Opin. Pharmacol. 2019, 49, 6–10. [Google Scholar] [CrossRef] [PubMed]
- Yoo, J.Y.; Groer, M.; Dutra, S.V.O.; Sarkar, A.; McSkimming, D.I. Gut Microbiota and Immune System Interactions. Microorganisms 2020, 8, 1587. [Google Scholar] [CrossRef] [PubMed]
- Carabotti, M.; Scirocco, A.; Maselli, M.A.; Severi, C. The Gut-Brain Axis: Interactions between Enteric Microbiota, Central and Enteric Nervous Systems. Ann. Gastroenterol. 2015, 28, 203–209. [Google Scholar] [PubMed]
- Appleton, J. The Gut-Brain Axis: Influence of Microbiota on Mood and Mental Health. Integr. Med. 2018, 17, 28–32. [Google Scholar]
- Gwak, M.G.; Chang, S.Y. Gut-Brain Connection: Microbiome, Gut Barrier, and Environmental Sensors. Immune Netw. 2021, 21, e20. [Google Scholar] [CrossRef] [PubMed]
- Hrncir, T. Gut Microbiota Dysbiosis: Triggers, Consequences, Diagnostic and Therapeutic Options. Microorganisms 2022, 10, 578. [Google Scholar] [CrossRef]
- Pryor, R.; Martinez-Martinez, D.; Quintaneiro, L.; Cabreiro, F. The Role of the Microbiome in Drug Response. Annu. Rev. Pharmacol. Toxicol. 2020, 60, 417–435. [Google Scholar] [CrossRef] [PubMed]
- Pant, A.; Maiti, T.K.; Mahajan, D.; Das, B. Human Gut Microbiota and Drug Metabolism. Microb. Ecol. 2023, 86, 97–111. [Google Scholar] [CrossRef] [PubMed]
- Goetz, L.H.; Schork, N.J. Personalized medicine: Motivation, challenges, and progress. Fertil. Steril. 2018, 109, 952–963. [Google Scholar] [CrossRef]
- Hamjane, N.; Mechita, M.B.; Nourouti, N.G.; Barakat, A. Gut microbiota dysbiosis -associated obesity and its involvement in cardiovascular diseases and type 2 diabetes: A systematic review. Microvasc. Res. 2024, 151, 104601. [Google Scholar] [CrossRef] [PubMed]
- Matzaras, R.; Nikopoulou, A.; Protonotariou, E.; Christaki, E. Gut Microbiota Modulation and Prevention of Dysbiosis as an Alternative Approach to Antimicrobial Resistance: A Narrative Review. Yale J. Biol. Med. 2022, 95, 479–494. [Google Scholar] [PubMed]
- Liu, Y.; Wang, J.; Wu, C. Modulation of Gut Microbiota and Immune System by Probiotics, Pre-biotics, and Post-biotics. Front. Nutr. 2022, 8, 634897. [Google Scholar] [CrossRef]
- Clemente-Suárez, V.J.; Redondo-Flórez, L.; Beltrán-Velasco, A.I.; Ramos-Campo, D.J.; Belinchón-Demiguel, P.; Martinez-Guardado, I.; Dalamitros, A.A.; Yáñez-Sepúlveda, R.; Martín-Rodríguez, A.; Tornero-Aguilera, J.F. Mitochondria and Brain Disease: A Comprehensive Review of Pathological Mechanisms and Therapeutic Opportunities. Biomedicines 2023, 11, 2488. [Google Scholar] [CrossRef]
- Clemente-Suárez, V.J.; Martín-Rodríguez, A.; Redondo-Flórez, L.; López-Mora, C.; Yáñez-Sepúlveda, R.; Tornero-Aguilera, J.F. New Insights and Potential Therapeutic Interventions in Metabolic Diseases. Int. J. Mol. Sci. 2023, 24, 10672. [Google Scholar] [CrossRef] [PubMed]
- Clemente-Suárez, V.J.; Redondo-Flórez, L.; Beltrán-Velasco, A.I.; Martín-Rodríguez, A.; Martínez-Guardado, I.; Navarro-Jiménez, E.; Laborde-Cárdenas, C.C.; Tornero-Aguilera, J.F. The Role of Adipokines in Health and Disease. Biomedicines 2023, 11, 1290. [Google Scholar] [CrossRef]
- Hiller-Sturmhöfel, S.; Bartke, A. The Endocrine System—An Overview. Alcohol. Res. Health 1998, 22, 153–164. [Google Scholar]
- Mashinini, M. Pituitary gland and growth hormone. South. Afr. J. Anaesth. Analg. 2020, 26, S109–S112. [Google Scholar] [CrossRef]
- Campbell, M.; Jialal, I. Physiology, Endocrine Hormones; StatPearls: Treasure Island, FL, USA, 2020. [Google Scholar]
- Ramsay, D.S.; Woods, S.C. Physiological Regulation: How It Really Works. Cell Metab. 2016, 24, 361–364. [Google Scholar] [CrossRef]
- Ashwell, E. The endocrine system and associated disorders. Br. J. Nurs. 2022, 31, 316–320. [Google Scholar] [CrossRef] [PubMed]
- Ali, S.A.; Begum, T.; Reza, F. Hormonal Influences on Cognitive Function. Malays. J. Med. Sci. 2018, 25, 31–41. [Google Scholar] [CrossRef] [PubMed]
- Hegde, B.M. Textbook of Endocrine Physiology. Postgrad. Med. J. 1994, 70, 57. [Google Scholar] [CrossRef]
- Wassner, A.J.; Modi, B.P. Endocrine physiology in the newborn. Semin. Pediatr. Surg. 2013, 22, 205–210. [Google Scholar] [CrossRef] [PubMed]
- Lightman, S.L.; Birnie, M.T.; Conway-Campbell, B.L. Dynamics of ACTH and Cortisol Secretion and Implications for Disease. Endocr. Rev. 2021, 41, 470–490. [Google Scholar] [CrossRef] [PubMed]
- Wubben, R.; Efstathiou, C.; Stevenson, N.J. The Interplay between the Immune System and Viruses. Vitam. Hormon. 2021, 117, 1–15. [Google Scholar] [CrossRef]
- Vaillant, A.A.J.; Sabir, S.; Jan, A. Physiology, Immune Response; StatPearls: Treasure Island, FL, USA, 2022. [Google Scholar]
- Morey, J.N.; A Boggero, I.; Scott, A.B.; Segerstrom, S.C. Current directions in stress and human immune function. Curr. Opin. Psychol. 2015, 5, 13–17. [Google Scholar] [CrossRef]
- Jia, W.-Y.; Zhang, J.-J. Effects of glucocorticoids on leukocytes: Genomic and non-genomic mechanisms. World J. Clin. Cases 2022, 10, 7187–7194. [Google Scholar] [CrossRef] [PubMed]
- Khundmiri, S.J.; Murray, R.D.; Lederer, E. PTH and Vitamin D. Compr. Physiol. 2016, 6, 561–601. [Google Scholar] [CrossRef]
- Blau, J.E.; Collins, M.T. The PTH-Vitamin D-FGF23 axis. Rev. Endocr. Metab. Disord. 2015, 16, 165–174. [Google Scholar] [CrossRef]
- Dretakis, K.; Igoumenou, V.G. The role of parathyroid hormone (PTH) and vitamin D in falls and hip fracture type. Aging Clin. Exp. Res. 2019, 31, 1501–1507. [Google Scholar] [CrossRef]
- Friedman, J.M. Leptin and the endocrine control of energy balance. Nat. Metab. 2019, 1, 754–764. [Google Scholar] [CrossRef]
- Boucsein, A.; Kamstra, K.; Tups, A. Central signalling cross-talk between insulin and leptin in glucose and energy homeostasis. J. Neuroendocr. 2021, 33, e12944. [Google Scholar] [CrossRef]
- Timper, K.; Brüning, J.C. Hypothalamic circuits regulating appetite and energy homeostasis: Pathways to obesity. Dis. Model. Mech. 2017, 10, 679–689. [Google Scholar] [CrossRef] [PubMed]
- Mishra, K.; Sawant, N.; Garg, S. Management of psychiatric disorders in patients with endocrine disorders. Indian. J. Psychiatry 2022, 64, S402–S413. [Google Scholar] [CrossRef] [PubMed]
- Kundakovic, M.; Rocks, D. Sex hormone fluctuation and increased female risk for depression and anxiety disorders: From clinical evidence to molecular mechanisms. Front. Neuroendocr. 2022, 66, 101010. [Google Scholar] [CrossRef]
- Navarro-Pardo, E.; Holland, C.A.; Cano, A. Sex Hormones and Healthy Psychological Aging in Women. Front. Aging Neurosci. 2018, 9, 439. [Google Scholar] [CrossRef]
- Mikkelsen, M.B.; Tramm, G.; Zachariae, R.; Gravholt, C.H.; O’Toole, M.S. A systematic review and meta-analysis of the effect of emotion regulation on cortisol. Compr. Psychoneuroendocrinol. 2021, 5, 100020. [Google Scholar] [CrossRef]
- Tsoli, M.; Boutzios, G.; Kaltsas, G. Immune System Effects on the Endocrine System; Endotext: South Dartmouth, MA, USA, 2019. [Google Scholar]
- Feo, R.; Rasmussen, P.; Wiechula, R.; Conroy, T.; Kitson, A. Developing effective and caring nurse-patient relationships. Nurs. Stand. 2017, 31, 54–63. [Google Scholar] [CrossRef]
- Balagué, N.; Hristovski, R.; Almarcha, M.; Garcia-Retortillo, S.; Ivanov, P.C. Network Physiology of Exercise: Vision and Perspectives. Front. Physiol. 2020, 11, 1607. [Google Scholar] [CrossRef]
- Poon, M.M.L.; Farber, D.L. The Whole Body as the System in Systems Immunology. iScience 2020, 23, 101509. [Google Scholar] [CrossRef]
- Ledford, H. CRISPR cancer trial success paves the way for personalized treatments. Nature 2022, 611, 433–434. [Google Scholar] [CrossRef]
- Kalra, S.; Kapoor, N. Environmental Endocrinology: An Expanding Horizon; Endotext: South Dartmouth, MA, USA, 2000. [Google Scholar]
- Iatcu, C.O.; Steen, A.; Covasa, M. Gut Microbiota and Complications of Type-2 Diabetes. Nutrients 2021, 14, 166. [Google Scholar] [CrossRef]
- Abe, K.; Fujita, M.; Hayashi, M.; Okai, K.; Takahashi, A.; Ohira, H. Gut and oral microbiota in autoimmune liver disease. Fukushima J. Med. Sci. 2020, 65, 71–75. [Google Scholar] [CrossRef] [PubMed]
- Adak, A.; Khan, M.R. An insight into gut microbiota and its functionalities. Cell. Mol. Life Sci. 2018, 76, 473–493. [Google Scholar] [CrossRef]
- Sender, R.; Fuchs, S.; Milo, R. Revised Estimates for the Number of Human and Bacteria Cells in the Body. PLoS Biol. 2016, 14, e1002533. [Google Scholar] [CrossRef] [PubMed]
- Clemente, J.C.; Manasson, J.; Scher, J.U. The role of the gut microbiome in systemic inflammatory disease. BMJ 2018, 360, j5145. [Google Scholar] [CrossRef] [PubMed]
- Ang, Q.Y.; Alexander, M.; Newman, J.C.; Tian, Y.; Cai, J.; Upadhyay, V.; Turnbaugh, J.A.; Verdin, E.; Hall, K.D.; Leibel, R.L.; et al. Ketogenic Diets Alter the Gut Microbiome Resulting in Decreased Intestinal Th17 Cells. Cell 2020, 181, 1263–1275.e16. [Google Scholar] [CrossRef]
- Campaniello, D.; Corbo, M.R.; Sinigaglia, M.; Speranza, B.; Racioppo, A.; Altieri, C.; Bevilacqua, A. How Diet and Physical Activity Modulate Gut Microbiota: Evidence, and Perspectives. Nutrients 2022, 14, 2456. [Google Scholar] [CrossRef]
- Bartlett, A.; Kleiner, M. Dietary protein and the intestinal microbiota: An understudied relationship. iScience 2022, 25, 105313. [Google Scholar] [CrossRef]
- Shi, N.; Li, N.; Duan, X.; Niu, H. Interaction between the gut microbiome and mucosal immune system. Mil. Med. Res. 2017, 4, 1–7. [Google Scholar] [CrossRef]
- Lechuga, S.; Ivanov, A.I. Disruption of the epithelial barrier during intestinal inflammation: Quest for new molecules and mechanisms. Biochim. Biophys. Acta Mol. Cell Res. 2017, 1864, 1183–1194. [Google Scholar] [CrossRef]
- Portincasa, P.; Bonfrate, L.; Vacca, M.; De Angelis, M.; Farella, I.; Lanza, E.; Khalil, M.; Wang, D.Q.-H.; Sperandio, M.; Di Ciaula, A. Gut Microbiota and Short Chain Fatty Acids: Implications in Glucose Homeostasis. Int. J. Mol. Sci. 2022, 23, 1105. [Google Scholar] [CrossRef]
- Johnson, K.V.-A. Gut microbiome composition and diversity are related to human personality traits. Hum. Microbiome J. 2020, 15, 100069. [Google Scholar] [CrossRef] [PubMed]
- Bear, T.L.K.; Dalziel, J.E.; Coad, J.; Roy, N.C.; Butts, C.A.; Gopal, P.K. The Role of the Gut Microbiota in Dietary Interventions for Depression and Anxiety. Adv. Nutr. 2020, 11, 890–907. [Google Scholar] [CrossRef] [PubMed]
- Vicentini, F.A.; Keenan, C.M.; Wallace, L.E.; Woods, C.; Cavin, J.-B.; Flockton, A.R.; Macklin, W.B.; Belkind-Gerson, J.; Hirota, S.A.; Sharkey, K.A. Intestinal microbiota shapes gut physiology and regulates enteric neurons and glia. Microbiome 2021, 9, 210. [Google Scholar] [CrossRef]
- Obata, K. Synaptic inhibition and γ-aminobutyric acid in the mammalian central nervous system. Proc. Jpn. Acad. Ser. B Phys. Biol. Sci. 2013, 89, 139–156. [Google Scholar] [CrossRef] [PubMed]
- Osadchiy, V.; Martin, C.R.; Mayer, E.A. The gut-brain axis and the microbiome: Mechanisms and clinical implications. Clin. Gastroenterol. Hepatol. 2019, 17, 322–332. [Google Scholar] [CrossRef] [PubMed]
- Asadi, A.; Mehr, N.S.; Mohamadi, M.H.; Shokri, F.; Heidary, M.; Sadeghifard, N.; Khoshnood, S. Obesity and gut–microbiota–brain axis: A narrative review. J. Clin. Lab. Anal. 2022, 36, e24420. [Google Scholar] [CrossRef] [PubMed]
- Lyte, M. The role of microbial endocrinology in infectious disease. J. Endocrinol. 1993, 137, 343–345. [Google Scholar] [CrossRef]
- Lyte, M.; Bailey, M.T. Neuroendocrine–Bacterial Interactions in a Neurotoxin-Induced Model of Trauma. J. Surg. Res. 1997, 70, 195–201. [Google Scholar] [CrossRef]
- Iyer, L.M.; Aravind, L.; Coon, S.L.; Klein, D.C.; Koonin, E.V. Evolution of cell–cell signaling in animals: Did late horizontal gene transfer from bacteria have a role? Trends Genet. 2004, 20, 292–299. [Google Scholar] [CrossRef]
- Sperandio, V.; Torres, A.G.; Jarvis, B.; Nataro, J.P.; Kaper, J.B. Bacteria–host communication: The language of hormones. Proc. Natl. Acad. Sci. USA 2003, 100, 8951–8956. [Google Scholar] [CrossRef]
- Freestone, P.P.E.; Lyte, M. Chapter 2: Microbial Endocrinology: Experimental Design Issues in the Study of Interkingdom Sig-nalling in Infectious Disease. Adv. Appl. Microbiol. 2008, 64, 75–105. [Google Scholar] [CrossRef]
- Li, L.; Rao, S.; Cheng, Y.; Zhuo, X.; Deng, C.; Xu, N.; Zhang, H.; Yang, L. Microbial osteoporosis: The interplay between the gut microbiota and bones via host metabolism and immunity. Microbiologyopen 2019, 8, e00810. [Google Scholar] [CrossRef]
- Heijtz, R.D.; Wang, S.; Anuar, F.; Qian, Y.; Björkholm, B.; Samuelsson, A.; Hibberd, M.L.; Forssberg, H.; Pettersson, S. Normal gut microbiota modulates brain development and behavior. Proc. Natl. Acad. Sci. USA 2011, 108, 3047–3052. [Google Scholar] [CrossRef] [PubMed]
- Cryan, J.F.; O’Mahony, S.M. The microbiome-gut-brain axis: From bowel to behavior. Neurogastroenterol. Motil. 2011, 23, 187–192. [Google Scholar] [CrossRef] [PubMed]
- Karavolos, M.; Spencer, H.; Bulmer, D.; Thompson, A.; Winzer, K.; Williams, P.; Hinton, J.; Khan, C.A. Adrenaline modulates the global transcriptional profile of Salmonella revealing a role in the antimicrobial peptide and oxidative stress resistance responses. BMC Genom. 2008, 9, 458. [Google Scholar] [CrossRef]
- Huang, X.; Lao, W.; Zhou, Y.; Sun, Y.; Wang, Q. Glutamate dehydrogenase enables Salmonella to survive under oxidative stress and escape from clearance in macrophages. FEBS Lett. 2021, 596, 81–94. [Google Scholar] [CrossRef]
- Sudo, N.; Chida, Y.; Aiba, Y.; Sonoda, J.; Oyama, N.; Yu, X.N.; Kubo, C.; Koga, Y. Postnatal microbial colonization programs the hypothalamic–pituitary–adrenal system for stress response in mice. J. Physiol. 2004, 558, 263–275. [Google Scholar] [CrossRef]
- Huo, R.; Zeng, B.; Zeng, L.; Cheng, K.; Li, B.; Luo, Y.; Wang, H.; Zhou, C.; Fang, L.; Li, W.; et al. Microbiota Modulate Anxiety-Like Behavior and Endocrine Abnormalities in Hypothalamic-Pituitary-Adrenal Axis. Front. Cell. Infect. Microbiol. 2017, 7, 489. [Google Scholar] [CrossRef]
- Messaoudi, M.; Lalonde, R.; Violle, N.; Javelot, H.; Desor, D.; Nejdi, A.; Bisson, J.F.; Rougeot, C.; Pichelin, M.; Cazaubiel, M.; et al. Assessment of psychotropic-like properties of a probiotic formulation (Lactobacillus helveticus R0052 and Bifidobacterium longum R0175) in rats and human subjects. Br. J. Nutr. 2011, 105, 755–764. [Google Scholar] [CrossRef]
- Bravo, J.A.; Forsythe, P.; Chew, M.V.; Escaravage, E.; Savignac, H.M.; Dinan, T.G.; Bienenstock, J.; Cryan, J.F. Ingestion of Lactobacillus strain regulates emotional behavior and central GABA receptor expression in a mouse via the vagus nerve. Proc. Natl. Acad. Sci. USA 2011, 108, 16050–16055. [Google Scholar] [CrossRef]
- Kornman, K.S.; Loesche, W.J. Effects of estradiol and progesterone on Bacteroides melaninogenicus and Bacteroides gingivalis. Infect. Immun. 1982, 35, 256–263. [Google Scholar] [CrossRef] [PubMed]
- Menon, R.; Watson, S.E.; Thomas, L.N.; Allred, C.D.; Dabney, A.; Azcarate-Peril, M.A.; Sturino, J.M. Diet Complexity and Estrogen Receptor β Status Affect the Composition of the Murine Intestinal Microbiota. Appl. Environ. Microbiol. 2013, 79, 5763–5773. [Google Scholar] [CrossRef] [PubMed]
- Ridlon, J.M.; Ikegawa, S.; Alves, J.M.P.; Zhou, B.; Kobayashi, A.; Iida, T.; Mitamura, K.; Tanabe, G.; Serrano, M.; De Guzman, A.; et al. Clostridium scindens: A human gut microbe with a high potential to convert glucocorticoids into androgens. J. Lipid Res. 2013, 54, 2437–2449. [Google Scholar] [CrossRef] [PubMed]
- Adlercreutz, H.; Pulkkinen, M.; Hämäläinen, E.; Korpela, J. Studies on the role of intestinal bacteria in metabolism of synthetic and natural steroid hormones. J. Steroid Biochem. 1984, 20, 217–229. [Google Scholar] [CrossRef]
- Fuhrman, B.J.; Feigelson, H.S.; Flores, R.; Gail, M.H.; Xu, X.; Ravel, J.; Goedert, J.J. Associations of the Fecal Microbiome with Urinary Estrogens and Estrogen Metabolites in Postmenopausal Women. J. Clin. Endocrinol. Metab. 2014, 99, 4632–4640. [Google Scholar] [CrossRef]
- Cruz-Morales, P.; Orellana, C.A.; Moutafis, G.; Moonen, G.; Rincon, G.; Nielsen, L.K.; Marcellin, E.; Bapteste, E. Revisiting the Evolution and Taxonomy of Clostridia, a Phylogenomic Update. Genome Biol. Evol. 2019, 11, 2035–2044. [Google Scholar] [CrossRef]
- Cho, I.; Yamanishi, S.; Cox, L.; Methé, B.A.; Zavadil, J.; Li, K.; Gao, Z.; Mahana, D.; Raju, K.; Teitler, I.; et al. Antibiotics in early life alter the murine colonic microbiome and adiposity. Nature 2012, 488, 621–626. [Google Scholar] [CrossRef]
- Fukumoto, S.; Tatewaki, M.; Yamada, T.; Fujimiya, M.; Mantyh, C.; Voss, M.; Eubanks, S.; Harris, M.; Pappas, T.N.; Takahashi, T.; et al. Short-chain fatty acids stimulate colonic transit via intraluminal 5-HT release in rats. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2003, 284, R1269–R1276. [Google Scholar] [CrossRef]
- Ahmed, M.; Ahmed, S. Functional, Diagnostic and Therapeutic Aspects of Gastrointestinal Hormones. Gastroenterol. Res. 2019, 12, 233–244. [Google Scholar] [CrossRef]
- Alhabeeb, H.; AlFaiz, A.; Kutbi, E.; AlShahrani, D.; Alsuhail, A.; AlRajhi, S.; Alotaibi, N.; Alotaibi, K.; AlAmri, S.; Alghamdi, S.; et al. Gut Hormones in Health and Obesity: The Upcoming Role of Short Chain Fatty Acids. Nutrients 2021, 13, 481. [Google Scholar] [CrossRef]
- Ng, Q.X.; Lim, Y.L.; Yaow, C.Y.L.; Ng, W.K.; Thumboo, J.; Liew, T.M. Effect of Probiotic Supplementation on Gut Microbiota in Patients with Major Depressive Disorders: A Systematic Review. Nutrients 2023, 15, 1351. [Google Scholar] [CrossRef] [PubMed]
- Liang, D.; Leung, R.K.-K.; Guan, W.; Au, W.W. Involvement of gut microbiome in human health and disease: Brief overview, knowledge gaps and research opportunities. Gut Pathog. 2018, 10, 3. [Google Scholar] [CrossRef] [PubMed]
- Pham, V.T.; Dold, S.; Rehman, A.; Bird, J.K.; Steinert, R.E. Vitamins, the gut microbiome and gastrointestinal health in humans. Nutr. Res. 2021, 95, 35–53. [Google Scholar] [CrossRef]
- Petersen, C.; Round, J.L. Defining dysbiosis and its influence on host immunity and disease. Cell. Microbiol. 2014, 16, 1024–1033. [Google Scholar] [CrossRef] [PubMed]
- Kriss, M.; Hazleton, K.Z.; Nusbacher, N.M.; Martin, C.G.; A Lozupone, C. Low diversity gut microbiota dysbiosis: Drivers, functional implications and recovery. Curr. Opin. Microbiol. 2018, 44, 34–40. [Google Scholar] [CrossRef] [PubMed]
- Hufnagl, K.; Pali-Schöll, I.; Roth-Walter, F.; Jensen-Jarolim, E. Dysbiosis of the gut and lung microbiome has a role in asthma. Semin. Immunopathol. 2020, 42, 75–93. [Google Scholar] [CrossRef] [PubMed]
- Nishino, K.; Nishida, A.; Inoue, R.; Kawada, Y.; Ohno, M.; Sakai, S.; Inatomi, O.; Bamba, S.; Sugimoto, M.; Kawahara, M.; et al. Analysis of endoscopic brush samples identified mucosa-associated dysbiosis in inflammatory bowel disease. J. Gastroenterol. 2017, 53, 95–106. [Google Scholar] [CrossRef]
- Sanders, M.E.; Merenstein, D.J.; Reid, G.; Gibson, G.R.; Rastall, R.A. Probiotics and prebiotics in intestinal health and disease: From biology to the clinic. Nat. Rev. Gastroenterol. Hepatol. 2019, 16, 605–616. [Google Scholar] [CrossRef]
- Cunningham, A.L.; Stephens, J.W.; Harris, D.A. A review on gut microbiota: A central factor in the pathophysiology of obesity. Lipids Health Dis. 2021, 20, 65. [Google Scholar] [CrossRef]
- Yu, X.; Jiang, W.; Kosik, R.O.; Song, Y.; Luo, Q.; Qiao, T.; Tong, J.; Liu, S.; Deng, C.; Qin, S.; et al. Gut microbiota changes and its potential relations with thyroid carcinoma. J. Adv. Res. 2021, 35, 61–70. [Google Scholar] [CrossRef]
- Xu, H.; Wang, X.; Feng, W.; Liu, Q.; Zhou, S.; Liu, Q.; Cai, L. The gut microbiota and its interactions with cardiovascular disease. Microb. Biotechnol. 2020, 13, 637–656. [Google Scholar] [CrossRef] [PubMed]
- Clemente-Suárez, V.J.; Beltrán-Velasco, A.I.; Redondo-Flórez, L.; Martín-Rodríguez, A.; Yáñez-Sepúlveda, R.; Tornero-Aguilera, J.F. Neuro-Vulnerability in Energy Metabolism Regulation: A Comprehensive Narrative Review. Nutrients 2023, 15, 3106. [Google Scholar] [CrossRef]
- Mullaney, J.A.; Stephens, J.E.; Costello, M.-E.; Fong, C.; Geeling, B.E.; Gavin, P.G.; Wright, C.M.; Spector, T.D.; Brown, M.A.; Hamilton-Williams, E.E. Type 1 diabetes susceptibility alleles are associated with distinct alterations in the gut microbiota. Microbiome 2018, 6, 35, Erratum in Microbiome 2018, 6, 51. [Google Scholar] [CrossRef]
- Sato, K.; Takahashi, N.; Kato, T.; Matsuda, Y.; Yokoji, M.; Yamada, M.; Nakajima, T.; Kondo, N.; Endo, N.; Yamamoto, R.; et al. Aggravation of collagen-induced arthritis by orally administered Porphyromonas gingivalis through modulation of the gut microbiota and gut immune system. Sci. Rep. 2017, 7, 6955. [Google Scholar] [CrossRef] [PubMed]
- Corrêa, J.D.; Calderaro, D.C.; Ferreira, G.A.; Mendonça, S.M.S.; Fernandes, G.R.; Xiao, E.; Teixeira, A.L.; Leys, E.J.; Graves, D.T.; Silva, T.A. Subgingival microbiota dysbiosis in systemic lupus erythematosus: Association with periodontal status. Microbiome 2017, 5, 34. [Google Scholar] [CrossRef] [PubMed]
- Ni, J.; Wu, G.D.; Albenberg, L.; Tomov, V.T. Gut microbiota and IBD: Causation or correlation? Nat. Rev. Gastroenterol. Hepatol. 2017, 14, 573–584. [Google Scholar] [CrossRef]
- Suzuki, S.; Campos-Alberto, E.; Morita, Y.; Yamaguchi, M.; Toshimitsu, T.; Kimura, K.; Ikegami, S.; Katsuki, T.; Kohno, Y.; Shimojo, N. Low Interleukin 10 Production at Birth Is a Risk Factor for Atopic Dermatitis in Neonates with Bifidobacterium Colonization. Int. Arch. Allergy Immunol. 2018, 177, 342–349. [Google Scholar] [CrossRef]
- Chang, H.-W.; Yan, D.; Singh, R.; Liu, J.; Lu, X.; Ucmak, D.; Lee, K.; Afifi, L.; Fadrosh, D.; Leech, J.; et al. Alteration of the cutaneous microbiome in psoriasis and potential role in Th17 polarization. Microbiome 2018, 6, 154. [Google Scholar] [CrossRef]
- Cekanaviciute, E.; Yoo, B.B.; Runia, T.F.; Debelius, J.W.; Singh, S.; Nelson, C.A.; Kanner, R.; Bencosme, Y.; Lee, Y.K.; Hauser, S.L.; et al. Gut bacteria from multiple sclerosis patients modulate human T cells and exacerbate symptoms in mouse models. Proc. Natl. Acad. Sci. USA 2017, 114, 10713–10718. [Google Scholar] [CrossRef]
- Hall, A.B.; Tolonen, A.C.; Xavier, R.J. Human genetic variation and the gut microbiome in disease. Nat. Rev. Genet. 2017, 18, 690–699. [Google Scholar] [CrossRef]
- Lin, X.; Li, H. Obesity: Epidemiology, Pathophysiology, and Therapeutics. Front. Endocrinol. 2021, 12, 706978. [Google Scholar] [CrossRef]
- Vijay, A.; Valdes, A.M. Role of the gut microbiome in chronic diseases: A narrative review. Eur. J. Clin. Nutr. 2021, 76, 489–501. [Google Scholar] [CrossRef] [PubMed]
- Lee, C.J.; Sears, C.L.; Maruthur, N. Gut microbiome and its role in obesity and insulin resistance. Ann. N. Y. Acad. Sci. 2020, 1461, 37–52. [Google Scholar] [CrossRef] [PubMed]
- Hu, Y.-H.; Meyer, K.; Lulla, A.; Lewis, C.E.; Carnethon, M.R.; Schreiner, P.J.; Sidney, S.; Shikany, J.M.; Meirelles, O.; Launer, L.J. Gut microbiome and stages of diabetes in middle-aged adults: CARDIA microbiome study. Nutr. Metab. 2023, 20, 3. [Google Scholar] [CrossRef] [PubMed]
- Sankararaman, S.; Noriega, K.; Velayuthan, S.; Sferra, T.; Martindale, R. Gut Microbiome and Its Impact on Obesity and Obesity-Related Disorders. Curr. Gastroenterol. Rep. 2023, 25, 31–44. [Google Scholar] [CrossRef]
- Barlow, G.M.; Yu, A.; Mathur, R. Role of the Gut Microbiome in Obesity and Diabetes Mellitus. Nutr. Clin. Pract. 2015, 30, 787–797. [Google Scholar] [CrossRef]
- Anderson, G. Type I Diabetes Pathoetiology and Pathophysiology: Roles of the Gut Microbiome, Pancreatic Cellular Interactions, and the ‘Bystander’ Activation of Memory CD8+ T Cells. Int. J. Mol. Sci. 2023, 24, 3300. [Google Scholar] [CrossRef]
- Sidhu, S.R.K.; Kok, C.W.; Kunasegaran, T.; Ramadas, A. Effect of Plant-Based Diets on Gut Microbiota: A Systematic Review of Interventional Studies. Nutrients 2023, 15, 1510. [Google Scholar] [CrossRef]
- Deandra, F.A.; Ketherin, K.; Rachmasari, R.; Sulijaya, B.; Takahashi, N. Probiotics and metabolites regulate the oral and gut microbiome composition as host modulation agents in periodontitis: A narrative review. Heliyon 2023, 9, e13475. [Google Scholar] [CrossRef]
- Sadagopan, A.; Mahmoud, A.; Begg, M.; Tarhuni, M.; Fotso, M.; Gonzalez, N.A.; Sanivarapu, R.R.; Osman, U.; Kumar, A.L.; Mohammed, L.; et al. Understanding the Role of the Gut Microbiome in Diabetes and Therapeutics Targeting Leaky Gut: A Systematic Review. Cureus 2023, 15, e41559. [Google Scholar] [CrossRef]
- Meslier, V.; Laiola, M.; Roager, H.M.; De Filippis, F.; Roume, H.; Quinquis, B.; Giacco, R.; Mennella, I.; Ferracane, R.; Pons, N.; et al. Mediterranean diet intervention in overweight and obese subjects lowers plasma cholesterol and causes changes in the gut microbiome and metabolome independently of energy intake. Gut 2020, 69, 1258–1268. [Google Scholar] [CrossRef]
- Jiang, W.; Lu, G.; Gao, D.; Lv, Z.; Li, D. The relationships between the gut microbiota and its metabolites with thyroid diseases. Front. Endocrinol. 2022, 13, 943408. [Google Scholar] [CrossRef]
- Knezevic, J.; Starchl, C.; Berisha, A.T.; Amrein, K. Thyroid-Gut-Axis: How Does the Microbiota Influence Thyroid Function? Nutrients 2020, 12, 1769. [Google Scholar] [CrossRef]
- Nicola, J.P.; Nazar, M.; Mascanfroni, I.D.; Pellizas, C.G.; Masini-Repiso, A.M. NF-κB p65 Subunit Mediates Lipopolysaccharide-Induced Na+/I− Symporter Gene Expression by Involving Functional Interaction with the Paired Domain Transcription Factor Pax8. Mol. Endocrinol. 2010, 24, 1846–1862. [Google Scholar] [CrossRef] [PubMed]
- Sponziello, M.; Scipioni, A.; Durante, C.; Verrienti, A.; Maranghi, M.; Giacomelli, L.; Ferretti, E.; Celano, M.; Filetti, S.; Russo, D. Regulation of Sodium/Iodide Symporter and Lactoperoxidase Expression in Four Human Breast Cancer Cell Lines. J. Endocrinol. Investig. 2010, 33, 529–534. [Google Scholar] [CrossRef] [PubMed]
- Yaylali, O.; Kirac, S.; Yilmaz, M.; Akin, F.; Yuksel, D.; Demirkan, N.; Akdag, B. Does Hypothyroidism Affect Gastrointestinal Motility? Gastroenterol. Res. Pract. 2009, 2009, 529802. [Google Scholar] [CrossRef] [PubMed]
- Hasham, A.; Tomer, Y. Genetic and epigenetic mechanisms in thyroid autoimmunity. Immunol. Res. 2012, 54, 204–213. [Google Scholar] [CrossRef]
- Ejtahed, H.-S.; Hasani-Ranjbar, S. Neuromodulatory effect of microbiome on gut-brain axis; new target for obesity drugs. J. Diabetes Metab. Disord. 2019, 18, 263–265. [Google Scholar] [CrossRef]
- Shen, C.; Zhang, Y.; Liu, Y.; Yin, S.; Zhang, X.; Wei, W.; Sun, Z.; Song, H.; Qiu, Z.; Wang, C.; et al. A distinct serum metabolic signature of distant metastatic papillary thyroid carcinoma. Clin. Endocrinol. 2017, 87, 844–852. [Google Scholar] [CrossRef]
- Vatanen, T.; Kostic, A.D.; D’hennezel, E.; Siljander, H.; Franzosa, E.A.; Yassour, M.; Kolde, R.; Vlamakis, H.; Arthur, T.D.; Hämäläinen, A.-M.; et al. Variation in Microbiome LPS Immunogenicity Contributes to Autoimmunity in Humans. Cell 2016, 165, 842–853. [Google Scholar] [CrossRef]
- Lindheim, L.; Bashir, M.; Münzker, J.; Trummer, C.; Zachhuber, V.; Leber, B.; Horvath, A.; Pieber, T.R.; Gorkiewicz, G.; Stadlbauer, V.; et al. Alterations in Gut Microbiome Composition and Barrier Function Are Associated with Reproductive and Metabolic Defects in Women with Polycystic Ovary Syndrome (PCOS): A Pilot Study. PLoS ONE 2017, 12, e0168390. [Google Scholar] [CrossRef]
- Schwarzer, M.; Makki, K.; Storelli, G.; Machuca-Gayet, I.; Srutkova, D.; Hermanova, P.; Martino, M.E.; Balmand, S.; Hudcovic, T.; Heddi, A.; et al. Lactobacillus plantarum strain maintains growth of infant mice during chronic undernutrition. Science 2016, 351, 854–857. [Google Scholar] [CrossRef]
- Clarke, G.; Stilling, R.M.; Kennedy, P.J.; Stanton, C.; Cryan, J.F.; Dinan, T.G. Minireview: Gut Microbiota: The Neglected Endocrine Organ. Mol. Endocrinol. 2014, 28, 1221–1238. [Google Scholar] [CrossRef]
- Canfora, E.E.; Jocken, J.W.; Blaak, E.E. Short-chain fatty acids in control of body weight and insulin sensitivity. Nat. Rev. Endocrinol. 2015, 11, 577–591. [Google Scholar] [CrossRef]
- Plottel, C.S.; Blaser, M.J. Microbiome and Malignancy. Cell Host Microbe 2011, 10, 324–335. [Google Scholar] [CrossRef]
- Foster, J.A.; McVey Neufeld, K.-A. Gut–brain axis: How the microbiome influences anxiety and depression. Trends Neurosci. 2013, 36, 305–312. [Google Scholar] [CrossRef]
- Vrieze, A.; Van Nood, E.; Holleman, F.; Salojärvi, J.; Kootte, R.S.; Bartelsman, J.F.; Dallinga-Thie, G.M.; Ackermans, M.T.; Serlie, M.J.; Oozeer, R.; et al. Transfer of Intestinal Microbiota from Lean Donors Increases Insulin Sensitivity in Individuals with Metabolic Syndrome. Gastroenterology 2012, 143, 913–916. [Google Scholar] [CrossRef] [PubMed]
- Fetissov, S.O.; Sinno, M.H.; Coëffier, M.; Bole-Feysot, C.; Ducrotté, P.; Hökfelt, T.; Déchelotte, P. Autoantibodies against appetite-regulating peptide hormones and neuropeptides: Putative modulation by gut microflora. Nutrition 2008, 24, 348–359. [Google Scholar] [CrossRef]
- Le Chatelier, E.; Nielsen, T.; Qin, J.; Prifti, E.; Hildebrand, F.; Falony, G.; Almeida, M.; Arumugam, M.; Batto, J.-M.; Kennedy, S.; et al. Richness of human gut microbiome correlates with metabolic markers. Nature 2013, 500, 541–546. [Google Scholar] [CrossRef] [PubMed]
- Strandwitz, P. Neurotransmitter modulation by the gut microbiota. Brain Res. 2018, 1693, 128–133. [Google Scholar] [CrossRef] [PubMed]
- Rook, G.A.W.; Raison, C.L.; Lowry, C.A. Microbiota, Immunoregulatory Old Friends and Psychiatric Disorders. Adv. Exp. Med. Biol. 2014, 817, 319–356. [Google Scholar] [CrossRef]
- Wahlström, A.; Sayin, S.I.; Marschall, H.-U.; Bäckhed, F. Intestinal Crosstalk between Bile Acids and Microbiota and Its Impact on Host Metabolism. Cell Metab. 2016, 24, 41–50. [Google Scholar] [CrossRef] [PubMed]
- Biagi, E.; Nylund, L.; Candela, M.; Ostan, R.; Bucci, L.; Pini, E.; Nikkïla, J.; Monti, D.; Satokari, R.; Franceschi, C.; et al. Through Ageing, and Beyond: Gut Microbiota and Inflammatory Status in Seniors and Centenarians. PLoS ONE 2010, 5, e10667. [Google Scholar] [CrossRef]
- Sanchez, M.; Darimont, C.; Drapeau, V.; Emady-Azar, S.; Lepage, M.; Rezzonico, E.; Ngom-Bru, C.; Berger, B.; Philippe, L.; Ammon-Zuffrey, C.; et al. Effect of Lactobacillus rhamnosusCGMCC1.3724 supplementation on weight loss and maintenance in obese men and women. Br. J. Nutr. 2014, 111, 1507–1519. [Google Scholar] [CrossRef] [PubMed]
- Hou, K.; Wu, Z.-X.; Chen, X.-Y.; Wang, J.-Q.; Zhang, D.; Xiao, C.; Zhu, D.; Koya, J.B.; Wei, L.; Li, J.; et al. Microbiota in health and diseases. Signal Transduct. Target. Ther. 2022, 7, 135. [Google Scholar] [CrossRef]
- Honda, K.; Littman, D.R. The microbiota in adaptive immune homeostasis and disease. Nature 2016, 535, 75–84. [Google Scholar] [CrossRef]
- Nova, E.; Wärnberg, J.; Gómez-Martínez, S.; Díaz, L.E.; Romeo, J.; Marcos, A. Immunomodulatory effects of probiotics in different stages of life. Br. J. Nutr. 2007, 98, S90–S95. [Google Scholar] [CrossRef]
- Schellenberg, J.; Smoragiewicz, W.; Karska-Wysocki, B. A rapid method combining immunofluorescence and flow cytometry for improved understanding of competitive interactions between lactic acid bacteria (LAB) and methicillin-resistant S. aureus (MRSA) in mixed culture. J. Microbiol. Methods 2006, 65, 1–9. [Google Scholar] [CrossRef]
- Ishikawa, H.; Akedo, I.; Otani, T.; Suzuki, T.; Nakamura, T.; Takeyama, I.; Ishiguro, S.; Miyaoka, E.; Sobue, T.; Kakizoe, T. Randomized trial of dietary fiber and Lactobacillus casei administration for prevention of colorectal tumors. Int. J. Cancer 2005, 116, 762–767. [Google Scholar] [CrossRef]
- Sanders, M.E.; Gibson, G.R.; Gill, H.S.; Guarner, F. Probiotics: Their Potential to Impact Human Health. Counc. Agric. Sci. Technol. 2007, 36, 1–20. [Google Scholar]
- Dai, Z.; Zhang, J.; Wu, Q.; Chen, J.; Liu, J.; Wang, L.; Chen, C.; Xu, J.; Zhang, H.; Shi, C.; et al. The role of microbiota in the development of colorectal cancer. Int. J. Cancer 2019, 145, 2032–2041. [Google Scholar] [CrossRef] [PubMed]
- Li, S.; Liu, J.; Zheng, X.; Ren, L.; Yang, Y.; Li, W.; Fu, W.; Wang, J.; Du, G. Tumorigenic bacteria in colorectal cancer: Mechanisms and treatments. Cancer Biol. Med. 2021, 18, 147–162. [Google Scholar] [CrossRef]
- Kostic, A.D.; Gevers, D.; Pedamallu, C.S.; Michaud, M.; Duke, F.; Earl, A.M.; Ojesina, A.I.; Jung, J.; Bass, A.J.; Tabernero, J.; et al. Genomic analysis identifies association of Fusobacterium with colorectal carcinoma. Genome Res. 2012, 22, 292–298. [Google Scholar] [CrossRef]
- Docimo, G.; Cangiano, A.; Romano, R.M.; Pignatelli, M.F.; Offi, C.; Paglionico, V.A.; Galdiero, M.; Donnarumma, G.; Nigro, V.; Esposito, D.; et al. The Human Microbiota in Endocrinology: Implications for Pathophysiology, Treatment, and Prognosis in Thyroid Diseases. Front. Endocrinol. 2020, 11, 586529. [Google Scholar] [CrossRef]
- Chen, J.; Pitmon, E.; Wang, K. Microbiome, inflammation and colorectal cancer. Semin. Immunol. 2017, 32, 43–53. [Google Scholar] [CrossRef]
- Grivennikov, S.I.; Wang, K.; Mucida, D.; Stewart, C.A.; Schnabl, B.; Jauch, D.; Taniguchi, K.; Yu, G.Y.; Osterreicher, C.H.; Hung, K.E.; et al. Adenoma-linked barrier defects and microbial products drive IL-23/IL-17-mediated tumour growth. Nature 2013, 491, 254–258. [Google Scholar] [CrossRef]
- Pushalkar, S.; Hundeyin, M.; Daley, D.; Zambirinis, C.P.; Kurz, E.; Mishra, A.; Mohan, N.; Aykut, B.; Usyk, M.; Torres, L.E.; et al. The Pancreatic Cancer Microbiome Promotes Oncogenesis by Induction of Innate and Adaptive Immune Suppression. Cancer Discov. 2018, 8, 403–416. [Google Scholar] [CrossRef]
- Ochi, A.; Nguyen, A.H.; Bedrosian, A.S.; Mushlin, H.M.; Zarbakhsh, S.; Barilla, R.; Zambirinis, C.P.; Fallon, N.C.; Rehman, A.; Pylayeva-Gupta, Y.; et al. MyD88 inhibition amplifies dendritic cell capacity to promote pancreatic carcinogenesis via Th2 cells. J. Exp. Med. 2012, 209, 1671–1687. [Google Scholar] [CrossRef] [PubMed]
- Gui, Q.-F.; Lu, H.-F.; Zhang, C.-X.; Xu, Z.-R.; Yang, Y.-H. Well-balanced commensal microbiota contributes to anti-cancer response in a lung cancer mouse model. Evolution 2015, 14, 5642–5651. [Google Scholar] [CrossRef]
- Routy, B.; le Chatelier, E.; DeRosa, L.; Duong, C.P.M.; Alou, M.T.; Daillère, R.; Fluckiger, A.; Messaoudene, M.; Rauber, C.; Roberti, M.P.; et al. Gut microbiome influences efficacy of PD-1–based immunotherapy against epithelial tumors. Science 2018, 359, 91–97. [Google Scholar] [CrossRef] [PubMed]
- Zhang, W.-Q.; Zhao, S.-K.; Luo, J.-W.; Dong, X.-P.; Hao, Y.-T.; Li, H.; Shan, L.; Zhou, Y.; Shi, H.-B.; Zhang, Z.-Y.; et al. Alterations of fecal bacterial communities in patients with lung cancer. Am. J. Transl. Res. 2018, 10, 3171–3185. [Google Scholar] [PubMed]
- Gopalakrishnan, V.; Spencer, C.N.; Nezi, L.; Reuben, A.; Andrews, M.C.; Karpinets, T.V.; Prieto, P.A.; Vicente, D.; Hoffman, K.; Wei, S.C.; et al. Gut microbiome modulates response to anti–PD-1 immunotherapy in melanoma patients. Science 2018, 359, 97–103. [Google Scholar] [CrossRef] [PubMed]
- Iida, N.; Dzutsev, A.; Stewart, C.A.; Smith, L.; Bouladoux, N.; Weingarten, R.A.; Molina, D.A.; Salcedo, R.; Back, T.; Cramer, S.; et al. Commensal Bacteria Control Cancer Response to Therapy by Modulating the Tumor Microenvironment. Science 2019, 342, 967–970. [Google Scholar] [CrossRef]
- Bingula, R.; Filaire, M.; Radosevic-Robin, N.; Bey, M.; Berthon, J.-Y.; Bernalier-Donadille, A.; Vasson, M.-P.; Filaire, E. Desired Turbulence? Gut-Lung Axis, Immunity, and Lung Cancer. J. Oncol. 2017, 2017, 5035371. [Google Scholar] [CrossRef]
- Trompette, A.; Gollwitzer, E.S.; Yadava, K.; Sichelstiel, A.K.; Sprenger, N.; Ngom-Bru, C.; Blanchard, C.; Junt, T.; Nicod, L.P.; Harris, N.L.; et al. Gut microbiota metabolism of dietary fiber influences allergic airway disease and hematopoiesis. Nat. Med. 2014, 20, 159–166. [Google Scholar] [CrossRef]
- Boursier, J.; Mueller, O.; Barret, M.; Machado, M.; Fizanne, L.; Araujo-Perez, F.; Guy, C.D.; Seed, P.C.; Rawls, J.F.; David, L.A.; et al. The severity of nonalcoholic fatty liver disease is associated with gut dysbiosis and shift in the metabolic function of the gut microbiota. Hepatology 2016, 63, 764–775. [Google Scholar] [CrossRef] [PubMed]
- Zhu, L.; Baker, S.S.; Gill, C.; Liu, W.; Alkhouri, R.; Baker, R.D.; Gill, S.R. Characterization of gut microbiomes in nonalcoholic steatohepatitis (NASH) patients: A connection between endogenous alcohol and NASH. Hepatology 2013, 57, 601–609. [Google Scholar] [CrossRef]
- Bajaj, J.S.; Betrapally, N.S.; Gillevet, P.M. Decompensated cirrhosis and microbiome interpretation. Nature 2015, 525, E1–E2. [Google Scholar] [CrossRef]
- Dapito, D.H.; Mencin, A.; Gwak, G.-Y.; Pradere, J.-P.; Jang, M.-K.; Mederacke, I.; Caviglia, J.M.; Khiabanian, H.; Adeyemi, A.; Bataller, R.; et al. Promotion of Hepatocellular Carcinoma by the Intestinal Microbiota and TLR4. Cancer Cell 2012, 21, 504–516. [Google Scholar] [CrossRef]
- Yu, L.-X.; Schwabe, R.F. The gut microbiome and liver cancer: Mechanisms and clinical translation. Nat. Rev. Gastroenterol. Hepatol. 2017, 14, 527–539. [Google Scholar] [CrossRef]
- Loo, T.M.; Kamachi, F.; Watanabe, Y.; Yoshimoto, S.; Kanda, H.; Arai, Y.; Nakajima-Takagi, Y.; Iwama, A.; Koga, T.; Sugimoto, Y.; et al. Gut Microbiota Promotes Obesity-Associated Liver Cancer through PGE2-Mediated Suppression of Antitumor Immunity. Cancer Discov. 2017, 7, 522–538. [Google Scholar] [CrossRef] [PubMed]
- Gallage, S.; Kotsiliti, E.; Heikenwalder, M. When Soluble Fibers Meet Hepatocellular Carcinoma: The Dark Side of Fermentation. Cell Metab. 2018, 28, 673–675. [Google Scholar] [CrossRef]
- Singh, V.; San Yeoh, B.; Chassaing, B.; Xiao, X.; Saha, P.; Olvera, R.A.; Lapek, J.D.; Zhang, L.; Wang, W.B.; Hao, S.; et al. Dysregulated Microbial Fermentation of Soluble Fiber Induces Cholestatic Liver Cancer. Cell 2018, 175, 679–694.e22. [Google Scholar] [CrossRef]
- Furusawa, Y.; Obata, Y.; Fukuda, S.; Endo, T.A.; Nakato, G.; Takahashi, D.; Nakanishi, Y.; Uetake, C.; Kato, K.; Kato, T.; et al. Commensal microbe-derived butyrate induces the differentiation of colonic regulatory T cells. Nature 2013, 504, 446–450. [Google Scholar] [CrossRef] [PubMed]
- Chang, C.-W.; Lee, H.-C.; Li, L.-H.; Chiau, J.-S.C.; Wang, T.-E.; Chuang, W.-H.; Chen, M.-J.; Wang, H.-Y.; Shih, S.-C.; Liu, C.-Y.; et al. Fecal Microbiota Transplantation Prevents Intestinal Injury, Upregulation of Toll-Like Receptors, and 5-Fluorouracil/Oxaliplatin-Induced Toxicity in Colorectal Cancer. Int. J. Mol. Sci. 2020, 21, 386. [Google Scholar] [CrossRef] [PubMed]
- Fong, W.; Li, Q.; Yu, J. Gut microbiota modulation: A novel strategy for prevention and treatment of colorectal cancer. Oncogene 2020, 39, 4925–4943. [Google Scholar] [CrossRef] [PubMed]
- Oh, N.S.; Lee, J.Y.; Kim, Y.-T.; Kim, S.H.; Lee, J.-H. Cancer-protective effect of a synbiotic combination between Lactobacillus gasseri 505 and a Cudrania tricuspidata leaf extract on colitis-associated colorectal cancer. Gut Microbes 2020, 12, 1785803. [Google Scholar] [CrossRef]
- Yang, J.J.; Yu, D.; Xiang, Y.-B.; Blot, W.; White, E.; Robien, K.; Sinha, R.; Park, Y.; Takata, Y.; Lazovich, D.; et al. Association of Dietary Fiber and Yogurt Consumption with Lung Cancer Risk: A Pooled Analysis. JAMA Oncol. 2020, 6, e194107. [Google Scholar] [CrossRef]
- Alexander, J.L.; Wilson, I.D.; Teare, J.; Marchesi, J.R.; Nicholson, J.K.; Kinross, J.M. Gut microbiota modulation of chemotherapy efficacy and toxicity. Nat. Rev. Gastroenterol. Hepatol. 2017, 14, 356–365. [Google Scholar] [CrossRef]
- Grice, E.A.; Kong, H.H.; Conlan, S.; Deming, C.B.; Davis, J.; Young, A.C.; Bouffard, G.G.; Blakesley, R.W.; Murray, P.R.; Green, E.D.; et al. Topographical and Temporal Diversity of the Human Skin Microbiome. Science 2009, 324, 1190–1192. [Google Scholar] [CrossRef]
- Rad, S.S.; Nikkhah, A.; Orvatinia, M.; Ejtahed, H.-S.; Sarhangi, N.; Jamaldini, S.H.; Khodayari, N.; Meybodi, H.R.A.; Hasanzad, M. Gut microbiota: A perspective of precision medicine in endocrine disorders. J. Diabetes Metab. Disord. 2020, 19, 1827–1834. [Google Scholar] [CrossRef]
- Bäckhed, F.; Ley, R.E.; Sonnenburg, J.L.; Peterson, D.A.; Gordon, J.I. Host-Bacterial Mutualism in the Human Intestine. Science 2005, 307, 1915–1920. [Google Scholar] [CrossRef]
- Schroeder, B.O.; Bäckhed, F. Signals from the gut microbiota to distant organs in physiology and disease. Nat. Med. 2016, 22, 1079–1089. [Google Scholar] [CrossRef]
- Ley, R.E.; Peterson, D.A.; Gordon, J.I. Ecological and Evolutionary Forces Shaping Microbial Diversity in the Human Intestine. Cell 2006, 124, 837–848. [Google Scholar] [CrossRef] [PubMed]
- Shanahan, F. The Gut Microbiota in 2011: Translating the microbiota to medicine. Nat. Rev. Gastroenterol. Hepatol. 2012, 9, 72–74. [Google Scholar] [CrossRef]
- Montalto, M.; D’Onofrio, F.; Gallo, A.; Cazzato, A.; Gasbarrini, G. Intestinal microbiota and its functions. Dig. Liver Dis. Suppl. 2009, 3, 30–34. [Google Scholar] [CrossRef]
- Vehik, K.; Dabelea, D. Why Are C-Section Deliveries Linked to Childhood Type 1 Diabetes? Diabetes 2011, 61, 36–37. [Google Scholar] [CrossRef]
- Cardwell, C.R.; Stene, L.C.; Joner, G.; Cinek, O.; Svensson, J.; Goldacre, M.J.; Parslow, R.C.; Pozzilli, P.; Brigis, G.; Stoyanov, D.; et al. Caesarean section is associated with an increased risk of childhood-onset type 1 diabetes mellitus: A meta-analysis of observational studies. Diabetologia 2008, 51, 726–735. [Google Scholar] [CrossRef] [PubMed]
- Ejtahed, H.-S.; Soroush, A.-R.; Angoorani, P.; Larijani, B.; Hasani-Ranjbar, S. Gut Microbiota as a Target in the Pathogenesis of Metabolic Disorders: A New Approach to Novel Therapeutic Agents. Horm. Metab. Res. 2016, 48, 349–358. [Google Scholar] [CrossRef] [PubMed]
- Marchesi, J.R.; Adams, D.H.; Fava, F.; Hermes, G.D.A.; Hirschfield, G.M.; Hold, G.; Quraishi, M.N.; Kinross, J.; Smidt, H.; Tuohy, K.M.; et al. The gut microbiota and host health: A new clinical frontier. Gut 2016, 65, 330–339. [Google Scholar] [CrossRef]
- Zhou, L.; Li, X.; Ahmed, A.; Wu, D.; Liu, L.; Qiu, J.; Yan, Y.; Jin, M.; Xin, Y. Gut Microbe Analysis between Hyperthyroid and Healthy Individuals. Curr. Microbiol. 2014, 69, 675–680. [Google Scholar] [CrossRef] [PubMed]
- Sanz, Y.; Olivares, M.; Moya-Pérez, Á.; Agostoni, C. Understanding the role of gut microbiome in metabolic disease risk. Pediatr. Res. 2014, 77, 236–244. [Google Scholar] [CrossRef]
- Zhang, X.; Shen, D.; Fang, Z.; Jie, Z.; Qiu, X.; Zhang, C.; Chen, Y.; Ji, L. Human Gut Microbiota Changes Reveal the Progression of Glucose Intolerance. PLoS ONE 2013, 8, e71108. [Google Scholar] [CrossRef]
- Karlsson, F.H.; Tremaroli, V.; Nookaew, I.; Bergström, G.; Behre, C.J.; Fagerberg, B.; Nielsen, J.; Bäckhed, F. Gut metagenome in European women with normal, impaired and diabetic glucose control. Nature 2013, 498, 99–103. [Google Scholar] [CrossRef] [PubMed]
- Wang, J.; Qin, J.; Li, Y.; Cai, Z.; Li, S.; Zhu, J.; Zhang, F.; Liang, S.; Zhang, W.; Guan, Y.; et al. A metagenome-wide association study of gut microbiota in type 2 diabetes. Nature 2012, 490, 55–60. [Google Scholar] [CrossRef]
- Mykhalchyshyn, G.; Kobyliak, N.; Bodnar, P. Diagnostic accuracy of acyl-ghrelin and it association with non-alcoholic fatty liver disease in type 2 diabetic patients. J. Diabetes Metab. Disord. 2015, 14, 44. [Google Scholar] [CrossRef] [PubMed]
- Cummings, J.H. Short chain fatty acids in the human colon. Gut 1981, 22, 763–779. [Google Scholar] [CrossRef]
- Roy, C.C.; Kien, C.L.; Bouthillier, L.; Levy, E. Short-Chain Fatty Acids: Ready for Prime Time? Nutr. Clin. Pract. 2006, 21, 351–366. [Google Scholar] [CrossRef]
- Vernia, P.; Gnaedinger, A.; Hauck, W.; Breuer, R.I. Organic anions and the diarrhea of inflammatory bowel disease. Dig. Dis. Sci. 1988, 33, 1353–1358. [Google Scholar] [CrossRef]
- Scheppach, W. Effects of short chain fatty acids on gut morphology and function. Gut 1994, 35, S35–S38. [Google Scholar] [CrossRef]
- Schwiertz, A.; Taras, D.; Schäfer, K.; Beijer, S.; Bos, N.A.; Donus, C.; Hardt, P.D. Microbiota and SCFA in Lean and Overweight Healthy Subjects. Obesity 2010, 18, 190–195. [Google Scholar] [CrossRef]
- Hartstra, A.V.; Bouter, K.E.; Bäckhed, F.; Nieuwdorp, M. Insights Into the Role of the Microbiome in Obesity and Type 2 Diabetes. Diabetes Care 2014, 38, 159–165. [Google Scholar] [CrossRef] [PubMed]
- Cotillard, A.; Kennedy, S.P.; Kong, L.C.; Prifti, E.; Pons, N.; Le Chatelier, E.; Almeida, M.; Quinquis, B.; Levenez, F.; Galleron, N.; et al. Dietary intervention impact on gut microbial gene richness. Nature 2013, 500, 585–588. [Google Scholar] [CrossRef] [PubMed]
- Creely, S.J.; McTernan, P.G.; Kusminski, C.M.; Fisher, F.M.; Da Silva, N.F.; Khanolkar, M.; Evans, M.; Harte, A.L.; Kumar, S. Lipopolysaccharide activates an innate immune system response in human adipose tissue in obesity and type 2 diabetes. Am. J. Physiol. Endocrinol. Metab. 2007, 292, E740–E747. [Google Scholar] [CrossRef] [PubMed]
- Chen, X.; Devaraj, S. Gut Microbiome in Obesity, Metabolic Syndrome, and Diabetes. Curr. Diabetes Rep. 2018, 18, 129. [Google Scholar] [CrossRef]
- Zoetendal, E.G.; Akkermans, A.D.L.; Akkermans-van Vliet, W.M.; De Visser, J.A.G.M.; De Vos, W.M. The Host Genotype Affects the Bacterial Community in the Human Gastronintestinal Tract. Microb. Ecol. Heal. Dis. 2001, 13, 129–134. [Google Scholar] [CrossRef]
- Bäckhed, F.; Ding, H.; Wang, T.; Hooper, L.V.; Koh, G.Y.; Nagy, A.; Semenkovich, C.F.; Gordon, J.I. The Gut Microbiota as an Environmental Factor That Regulates Fat Storage. Proc. Natl. Acad. Sci. USA 2004, 101, 15718–15723. [Google Scholar] [CrossRef]
- Baothman, O.A.; Zamzami, M.A.; Taher, I.; Abubaker, J.; Abu-Farha, M. The role of Gut Microbiota in the development of obesity and Diabetes. Lipids Health Dis. 2016, 15, 108. [Google Scholar] [CrossRef]
- Insenser, M.; Murri, M.; Del Campo, R.; Martínez-García, M.; Fernández-Durán, E.; Escobar-Morreale, H.F. Gut Microbiota and the Polycystic Ovary Syndrome: Influence of Sex, Sex Hormones, and Obesity. J. Clin. Endocrinol. Metab. 2018, 103, 2552–2562. [Google Scholar] [CrossRef]
- Pi-Sunyer, F.X. The Obesity Epidemic: Pathophysiology and Consequences of Obesity. Obes. Res. 2002, 10, 97S–104S. [Google Scholar] [CrossRef]
- Afzaal, M.; Saeed, F.; Shah, Y.A.; Hussain, M.; Rabail, R.; Socol, C.T.; Hassoun, A.; Pateiro, M.; Lorenzo, J.M.; Rusu, A.V.; et al. Human gut microbiota in health and disease: Unveiling the relationship. Front. Microbiol. 2022, 13, 999001. [Google Scholar] [CrossRef]
- Blaser, M.J.; Falkow, S. What are the consequences of the disappearing human microbiota? Nat. Rev. Microbiol. 2009, 7, 887–894. [Google Scholar] [CrossRef] [PubMed]
- Hooper, L.V.; Gordon, J.I. Commensal Host-Bacterial Relationships in the Gut. Science 2001, 292, 1115–1118. [Google Scholar] [CrossRef] [PubMed]
- Neish, A.S. Microbes in Gastrointestinal Health and Disease. Gastroenterology 2009, 136, 65–80. [Google Scholar] [CrossRef] [PubMed]
- Crovesy, L.; Masterson, D.; Rosado, E.L. Profile of the gut microbiota of adults with obesity: A systematic review. Eur. J. Clin. Nutr. 2020, 74, 1251–1262. [Google Scholar] [CrossRef]
- Ley, R.E.; Turnbaugh, P.J.; Klein, S.; Gordon, J.I. Human Gut Microbes Associated with Obesity. Nature 2006, 444, 1022–1023. [Google Scholar] [CrossRef] [PubMed]
- Ley, R.E.; Bäckhed, F.; Turnbaugh, P.; Lozupone, C.A.; Knight, R.D.; Gordon, J.I. Obesity alters gut microbial ecology. Proc. Natl. Acad. Sci. USA 2005, 102, 11070–11075. [Google Scholar] [CrossRef] [PubMed]
- Turnbaugh, P.J.; Ley, R.E.; Mahowald, M.A.; Magrini, V.; Mardis, E.R.; Gordon, J.I. An Obesity-Associated Gut Microbiome with Increased Capacity for Energy Harvest. Nature 2006, 444, 1027–1031. [Google Scholar] [CrossRef]
- Guo, Y.; Qi, Y.; Yang, X.; Zhao, L.; Wen, S.; Liu, Y.; Tang, L. Association between Polycystic Ovary Syndrome and Gut Microbiota. PLoS ONE 2016, 11, e0153196. [Google Scholar] [CrossRef]
- Yurtdaş, G.; Akdevelioğlu, Y. A New Approach to Polycystic Ovary Syndrome: The Gut Microbiota. J. Am. Coll. Nutr. 2020, 39, 371–382. [Google Scholar] [CrossRef]
- Markle, J.G.M.; Frank, D.N.; Mortin-Toth, S.; Robertson, C.E.; Feazel, L.M.; Rolle-Kampczyk, U.; von Bergen, M.; McCoy, K.D.; Macpherson, A.J.; Danska, J.S. Sex Differences in the Gut Microbiome Drive Hormone-Dependent Regulation of Autoimmunity. Science 2013, 339, 1084–1088. [Google Scholar] [CrossRef] [PubMed]
- Tremellen, K.; Pearce, K. Dysbiosis of Gut Microbiota (DOGMA)—A novel theory for the development of Polycystic Ovarian Syndrome. Med. Hypotheses 2012, 79, 104–112. [Google Scholar] [CrossRef] [PubMed]
- Escobar-Morreale, H.F.; Luque-Ramírez, M.; Millán, J.L.S. The Molecular-Genetic Basis of Functional Hyperandrogenism and the Polycystic Ovary Syndrome. Endocr. Rev. 2005, 26, 251–282. [Google Scholar] [CrossRef] [PubMed]
- Kageyama, A.; Benno, Y. Catenibacterium mitsuokai gen. nov., sp. nov., a gram-positive anaerobic bacterium isolated from human faeces. Int. J. Syst. Evol. Microbiol. 2000, 50, 1595–1599. [Google Scholar] [CrossRef] [PubMed]
- Liu, R.; Zhang, C.; Shi, Y.; Zhang, F.; Li, L.; Wang, X.; Ling, Y.; Fu, H.; Dong, W.; Shen, J.; et al. Dysbiosis of Gut Microbiota Associated with Clinical Parameters in Polycystic Ovary Syndrome. Front. Microbiol. 2017, 8, 324. [Google Scholar] [CrossRef] [PubMed]
- Fröhlich, E.; Wahl, R. Microbiota and Thyroid Interaction in Health and Disease. Trends Endocrinol. Metab. 2019, 30, 479–490. [Google Scholar] [CrossRef] [PubMed]
- Navarro, A.M.; Suen, V.M.M.; Souza, I.M.; De Oliveira, J.E.D.; Marchini, J.S. Patients with severe bowel malabsorption do not have changes in iodine status. Nutrition 2005, 21, 895–900. [Google Scholar] [CrossRef]
- Hrdina, J.; Banning, A.; Kipp, A.; Loh, G.; Blaut, M.; Brigelius-Flohé, R. The gastrointestinal microbiota affects the selenium status and selenoprotein expression in mice. J. Nutr. Biochem. 2009, 20, 638–648. [Google Scholar] [CrossRef]
- Jonklaas, J.; Bianco, A.C.; Bauer, A.J.; Burman, K.D.; Cappola, A.R.; Celi, F.S.; Cooper, D.S.; Kim, B.W.; Peeters, R.P.; Rosenthal, M.S.; et al. Guidelines for the Treatment of Hypothyroidism: Prepared by the American Thyroid Association Task Force on Thyroid Hormone Replacement. Thyroid 2014, 24, 1670–1751. [Google Scholar] [CrossRef]
- Ejtahed, H.-S.; Angoorani, P.; Soroush, A.-R.; Siadat, S.-D.; Shirzad, N.; Hasani-Ranjbar, S.; Larijani, B. Our Little Friends with Big Roles: Alterations of the Gut Microbiota in Thyroid Disorders. Endocrine, Metab. Immune Disord. Drug Targets 2020, 20, 344–350. [Google Scholar] [CrossRef]
- Bahn, R.S.; Burch, H.B.; Cooper, D.S.; Garber, J.R.; Greenlee, M.C.; Klein, I.; Laurberg, P.; McDougall, I.R.; Montori, V.M.; Rivkees, S.A.; et al. Hyperthyroidism and Other Causes of Thyrotoxicosis: Management Guidelines of the American Thyroid Association and American Association of Clinical Endocrinologists. Thyroid 2011, 21, 593–646. [Google Scholar] [CrossRef] [PubMed]
- Upton, N. Developing our understanding of nutrition in depression. Br. J. Nutr. 2021, 127, 1010–1017. [Google Scholar] [CrossRef] [PubMed]
- Zhao, F.; Feng, J.; Li, J.; Zhao, L.; Liu, Y.; Chen, H.; Jin, Y.; Zhu, B.; Wei, Y. Alterations of the Gut Microbiota in Hashimoto’s Thyroiditis Patients. Thyroid 2018, 28, 175–186. [Google Scholar] [CrossRef]
- Köhling, H.L.; Plummer, S.F.; Marchesi, J.R.; Davidge, K.S.; Ludgate, M. The microbiota and autoimmunity: Their role in thyroid autoimmune diseases. Clin. Immunol. 2017, 183, 63–74. [Google Scholar] [CrossRef] [PubMed]
- Jeon, Y.K.; Kim, B.H.; Kim, I.J. The diagnosis of osteoporosis. J. Korean Med. Assoc. 2016, 59, 842–846. [Google Scholar] [CrossRef]
- Kanis, J.A.; Cooper, C.; Rizzoli, R.; Reginster, J.-Y. European Guidance for the Diagnosis and Management of Osteoporosis in Postmenopausal Women. Osteoporos. Int. 2019, 30, 3–44. [Google Scholar] [CrossRef] [PubMed]
- Blain, H.; Rolland, Y.; Beauchet, O.; Annweiler, C.; Benhamou, C.-L.; Benetos, A.; Berrut, G.; Audran, M.; Bendavid, S.; Bousson, V.; et al. Usefulness of bone density measurement in fallers. Jt. Bone Spine 2014, 81, 403–408. [Google Scholar] [CrossRef]
- Riggs, B.L.; Melton, L.J.; A Robb, R.; Camp, J.J.; Atkinson, E.J.; Peterson, J.M.; A Rouleau, P.; McCollough, C.H.; Bouxsein, M.L.; Khosla, S. Population-Based Study of Age and Sex Differences in Bone Volumetric Density, Size, Geometry, and Structure at Different Skeletal Sites. J. Bone Miner. Res. 2004, 19, 1945–1954. [Google Scholar] [CrossRef]
- Neugebauer, J.; Heilig, J.; Hosseinibarkooie, S.; Ross, B.C.; Mendoza-Ferreira, N.; Nolte, F.; Peters, M.; Hölker, I.; Hupperich, K.; Tschanz, T.; et al. Plastin 3 influences bone homeostasis through regulation of osteoclast activity. Hum. Mol. Genet. 2018, 27, 4249–4262. [Google Scholar] [CrossRef]
- Fabbiano, S.; Suárez-Zamorano, N.; Chevalier, C.; Lazarević, V.; Kieser, S.; Rigo, D.; Leo, S.; Veyrat-Durebex, C.; Gaïa, N.; Maresca, M.; et al. Functional Gut Microbiota Remodeling Contributes to the Caloric Restriction-Induced Metabolic Improvements. Cell Metab. 2018, 28, 907–921.e7. [Google Scholar] [CrossRef]
- Huang, K.-P.; Raybould, H.E. Estrogen and gut satiety hormones in vagus-hindbrain axis. Peptides 2020, 133, 170389. [Google Scholar] [CrossRef]
- Whisner, C.M.; Castillo, L.F. Prebiotics, Bone and Mineral Metabolism. Calcif. Tissue Int. 2018, 102, 443–479. [Google Scholar] [CrossRef] [PubMed]
- Veziant, J.; Villéger, R.; Barnich, N.; Bonnet, M. Gut Microbiota as Potential Biomarker and/or Therapeutic Target to Improve the Management of Cancer: Focus on Colibactin-Producing Escherichia coli in Colorectal Cancer. Cancers 2021, 13, 2215. [Google Scholar] [CrossRef]
- Kau, A.L.; Ahern, P.P.; Griffin, N.W.; Goodman, A.L.; Gordon, J.I. Human nutrition, the gut microbiome and the immune system. Nature 2011, 474, 327–336. [Google Scholar] [CrossRef] [PubMed]
- Garrett, W.S.; Gordon, J.I.; Glimcher, L.H. Homeostasis and Inflammation in the Intestine. Cell 2010, 140, 859–870. [Google Scholar] [CrossRef] [PubMed]
- Cullin, N.; Antunes, C.A.; Straussman, R.; Stein-Thoeringer, C.K.; Elinav, E. Microbiome and cancer. Cancer Cell 2021, 39, 1317–1341. [Google Scholar] [CrossRef] [PubMed]
- Zhang, Y.-J.; Li, S.; Gan, R.-Y.; Zhou, T.; Xu, D.-P.; Li, H.-B. Impacts of Gut Bacteria on Human Health and Diseases. Int. J. Mol. Sci. 2015, 16, 7493–7519. [Google Scholar] [CrossRef]
- Hooper, L.V.; Littman, D.R.; MacPherson, A.J. Interactions between the Microbiota and the Immune System. Science 2012, 336, 1268–1273. [Google Scholar] [CrossRef]
- Costello, E.K.; Stagaman, K.; Dethlefsen, L.; Bohannan, B.J.M.; Relman, D.A. The Application of Ecological Theory Toward an Understanding of the Human Microbiome. Science 2012, 336, 1255–1262. [Google Scholar] [CrossRef]
- Mallick, H.; Ma, S.; Franzosa, E.A.; Vatanen, T.; Morgan, X.C.; Huttenhower, C. Experimental design and quantitative analysis of microbial community multiomics. Genome Biol. 2017, 18, 228. [Google Scholar] [CrossRef]
- Pérez-Cobas, A.E.; Gomez-Valero, L.; Buchrieser, C. Metagenomic approaches in microbial ecology: An update on whole-genome and marker gene sequencing analyses. Microb. Genom. 2020, 6, e000409. [Google Scholar] [CrossRef]
- Wilson, S.; Steele, S.; Adeli, K. Innovative technological advancements in laboratory medicine: Predicting the lab of the future. Biotechnol. Biotechnol. Equip. 2022, 36, S9–S21. [Google Scholar] [CrossRef]
- Virolainen, S.J.; VonHandorf, A.; Viel, K.C.M.F.; Weirauch, M.T.; Kottyan, L.C. Gene–environment interactions and their impact on human health. Genes. Immun. 2023, 24, 1–11. [Google Scholar] [CrossRef]
- Blanco-Míguez, A.; Beghini, F.; Cumbo, F.; McIver, L.J.; Thompson, K.N.; Zolfo, M.; Manghi, P.; Dubois, L.; Huang, K.D.; Thomas, A.M.; et al. Extending and improving metagenomic taxonomic profiling with uncharacterized species using MetaPhlAn 4. Nat. Biotechnol. 2023, 41, 1633–1644. [Google Scholar] [CrossRef]
- Awasthi, M.K.; Ravindran, B.; Sarsaiya, S.; Chen, H.; Wainaina, S.; Singh, E.; Liu, T.; Kumar, S.; Pandey, A.; Singh, L.; et al. Metagenomics for taxonomy profiling: Tools and approaches. Bioengineered 2020, 11, 356–374. [Google Scholar] [CrossRef] [PubMed]
- Schluter, J.; Peled, J.U.; Taylor, B.P.; Markey, K.A.; Smith, J.A.; Taur, Y.; Niehus, R.; Staffas, A.; Dai, A.; Fontana, E.; et al. The gut microbiota is associated with immune cell dynamics in humans. Nature 2020, 588, 303–307. [Google Scholar] [CrossRef] [PubMed]
- Schlechte, J.; Zucoloto, A.Z.; Yu, I.-L.; Doig, C.J.; Dunbar, M.J.; McCoy, K.D.; McDonald, B. Dysbiosis of a microbiota–immune metasystem in critical illness is associated with nosocomial infections. Nat. Med. 2023, 29, 1017–1027. [Google Scholar] [CrossRef] [PubMed]
- Sánchez-Sánchez, P.; Santonja, F.J.; Benítez-Páez, A. Assessment of human microbiota stability across longitudinal samples using iteratively growing-partitioned clustering. Brief. Bioinform. 2022, 23, bbac055. [Google Scholar] [CrossRef]
- Kodikara, S.; Ellul, S.; Cao, K.-A.L. Statistical challenges in longitudinal microbiome data analysis. Brief. Bioinform. 2022, 23, bbac273. [Google Scholar] [CrossRef]
- Gerber, G.K. Longitudinal microbiome data analysis. In Metagenomics for Microbiology; Frontiers: Lausanne, Switzerland, 2015. [Google Scholar] [CrossRef]
- Lema, N.K.; Gemeda, M.T.; Woldesemayat, A.A. Recent Advances in Metagenomic Approaches, Applications, and Challenges. Curr. Microbiol. 2023, 80, 347. [Google Scholar] [CrossRef]
- Knights, D.; Parfrey, L.W.; Zaneveld, J.; Lozupone, C.; Knight, R. Human-Associated Microbial Signatures: Examining Their Predictive Value. Cell Host Microbe 2011, 10, 292–296. [Google Scholar] [CrossRef] [PubMed]
- Rintala, A.; Pietilä, S.; Munukka, E.; Eerola, E.; Pursiheimo, J.-P.; Laiho, A.; Pekkala, S.; Huovinen, P. Gut Microbiota Analysis Results Are Highly Dependent on the 16S rRNA Gene Target Region, Whereas the Impact of DNA Extraction Is Minor. J. Biomol. Tech. 2017, 28, 19–30. [Google Scholar] [CrossRef] [PubMed]
- Metwaly, A.; Reitmeier, S.; Haller, D. Microbiome risk profiles as biomarkers for inflammatory and metabolic disorders. Nat. Rev. Gastroenterol. Hepatol. 2022, 19, 383–397. [Google Scholar] [CrossRef]
- Balser, T.C.; McMahon, K.D.; Bart, D.; Bronson, D.; Coyle, D.R.; Craig, N.; Flores-Mangual, M.L.; Forshay, K.; Jones, S.E.; Kent, A.E.; et al. Bridging the gap between micro- and macro-scale perspectives on the role of microbial communities in global change ecology. Plant Soil. 2006, 289, 59–70. [Google Scholar] [CrossRef]
- Satam, H.; Joshi, K.; Mangrolia, U.; Waghoo, S.; Zaidi, G.; Rawool, S.; Thakare, R.P.; Banday, S.; Mishra, A.K.; Das, G.; et al. Next-Generation Sequencing Technology: Current Trends and Advancements. Biology 2023, 12, 997. [Google Scholar] [CrossRef]
- Peters, D.L.; Wang, W.; Zhang, X.; Ning, Z.; Mayne, J.; Figeys, D. Metaproteomic and Metabolomic Approaches for Characterizing the Gut Microbiome. Proteomics 2019, 19, e1800363. [Google Scholar] [CrossRef]
- Qiu, S.; Cai, Y.; Yao, H.; Lin, C.; Xie, Y.; Tang, S.; Zhang, A. Small molecule metabolites: Discovery of biomarkers and therapeutic targets. Signal Transduct. Target. Ther. 2023, 8, 132. [Google Scholar] [CrossRef] [PubMed]
- Muthubharathi, B.C.; Gowripriya, T.; Balamurugan, K. Metabolomics: Small molecules that matter more. Mol. Omics 2021, 17, 210–229. [Google Scholar] [CrossRef]
- De Vos, W.M.; Tilg, H.; Van Hul, M.; Cani, P.D. Gut microbiome and health: Mechanistic insights. Gut 2022, 71, 1020–1032. [Google Scholar] [CrossRef]
- Krajmalnik-Brown, R.; Ilhan, Z.E.; Kang, D.W.; DiBaise, J.K. Effects of Gut Microbes on Nutrient Absorption and Energy Regulation. Nutr. Clin. Pract. 2012, 27, 201–214. [Google Scholar] [CrossRef]
- Danzi, F.; Pacchiana, R.; Mafficini, A.; Scupoli, M.T.; Scarpa, A.; Donadelli, M.; Fiore, A. To metabolomics and beyond: A technological portfolio to investigate cancer metabolism. Signal Transduct. Target. Ther. 2023, 8, 137. [Google Scholar] [CrossRef]
- Monteiro, M.S.; Carvalho, M.; Bastos, M.L.; Guedes de Pinho, P. Metabolomics Analysis for Biomarker Discovery: Advances and Challenges. Curr. Med. Chem. 2013, 20, 257–271. [Google Scholar] [CrossRef] [PubMed]
- Al-Amrani, S.; Al-Jabri, Z.; Al-Zaabi, A.; Alshekaili, J.; Al-Khabori, M. Proteomics: Concepts and applications in human medicine. World J. Biol. Chem. 2021, 12, 57–69. [Google Scholar] [CrossRef] [PubMed]
- Santiago-Rodriguez, T.M.; Hollister, E.B. Multi ‘omic data integration: A review of concepts, considerations, and approaches. Semin. Perinatol. 2021, 45, 151456. [Google Scholar] [CrossRef] [PubMed]
- Liu, H.; Liao, C.; Wu, L.; Tang, J.; Chen, J.; Lei, C.; Zheng, L.; Zhang, C.; Liu, Y.-Y.; Xavier, J.; et al. Ecological dynamics of the gut microbiome in response to dietary fiber. ISME J. 2022, 16, 2040–2055. [Google Scholar] [CrossRef]
- Neuman, H.; Debelius, J.W.; Knight, R.; Koren, O. Microbial endocrinology: The interplay between the microbiota and the endocrine system. FEMS Microbiol. Rev. 2015, 39, 509–521. [Google Scholar] [CrossRef]
- Blaak, E.E.; Canfora, E.E.; Theis, S.; Frost, G.; Groen, A.K.; Mithieux, G.; Nauta, A.; Scott, K.; Stahl, B.; Van Harsselaar, J.; et al. Short chain fatty acids in human gut and metabolic health. Benef. Microbes 2020, 11, 411–455. [Google Scholar] [CrossRef]
- Bakadia, B.M.; Manan, S.; Ul-Islam, M.; Mukole, B.M.; Shahzad, A.; Abdalla, A.M.E.; Ullah, M.W.; Yang, G. Microbiome as cancer biomarkers. In Cancer Biomarkers in Diagnosis and Therapeutics; Springer: Berlin/Heidelberg, Germany, 2022. [Google Scholar] [CrossRef]
- Herren, C.M.; McMahon, K.D. Keystone taxa predict compositional change in microbial communities. Environ. Microbiol. 2018, 20, 2207–2217. [Google Scholar] [CrossRef]
- Leeuwendaal, N.K.; Cryan, J.F.; Schellekens, H. Gut peptides and the microbiome: Focus on ghrelin. Curr. Opin. Endocrinol. Diabetes 2021, 28, 243–252. [Google Scholar] [CrossRef]
- Rosario, S.R.; Dong, B.; Zhang, Y.; Hsiao, H.-H.; Isenhart, E.; Wang, J.; Siegel, E.M.; Monjazeb, A.M.; Owen, D.H.; Dey, P.; et al. Metabolic Dysregulation Explains the Diverse Impacts of Obesity in Males and Females with Gastrointestinal Cancers. Int. J. Mol. Sci. 2023, 24, 10847. [Google Scholar] [CrossRef]
- Shen, Y.; Nie, J.; Kuang, L.; Zhang, J.; Li, H. DNA sequencing, genomes and genetic markers of microbes on fruits and vegetables. Microb. Biotechnol. 2021, 14, 323–362. [Google Scholar] [CrossRef] [PubMed]
- Slikker, W. Biomarkers and their impact on precision medicine. Exp. Biol. Med. 2018, 243, 211–212. [Google Scholar] [CrossRef]
- Wilson, J.L.; Altman, R.B. Biomarkers: Delivering on the expectation of molecularly driven, quantitative health. Exp. Biol. Med. 2018, 243, 313–322. [Google Scholar] [CrossRef]
- Smith, K.; Climer, S. Heterogeneity Impacts Biomarker Discovery for Precision Medicine. medRxiv 2022. [Google Scholar] [CrossRef]
- Pataky, Z.; Bobbioni-Harsch, E.; Hadengue, A.; Carpentier, A.; Golay, A. Gut microbiota, responsible for our body weight? Rev. Med. Suisse 2009, 5, 662–664+666. [Google Scholar]
- Kowalewska, B.; Zorena, K.; Szmigiero-Kawko, M.; Wąż, P.; Myśliwiec, M. Higher diversity in fungal species discriminates children with type 1 diabetes mellitus from healthy control. Patient Prefer. Adherence 2016, 10, 591–599. [Google Scholar] [CrossRef] [PubMed]
- Mazloom, K.; Siddiqi, I.; Covasa, M. Probiotics: How Effective Are They in the Fight. against Obesity? Nutrients 2019, 11, 258. [Google Scholar] [CrossRef]
- Ozbey, G.; Hanafiah, A.; Sproston, E. Helicobacter pylori Infection and Gastric Microbiota. Euroasian J. Hepato-Gastroenterol. 2020, 10, 36–41. [Google Scholar] [CrossRef]
- Curimbaba, T.F.S.; Almeida-Junior, L.D.; Chagas, A.S.; Quaglio, A.E.V.; Herculano, A.M.; Di Stasi, L.C. Prebiotic, antioxidant and anti-inflammatory properties of edible Amazon fruits. Food Biosci. 2020, 36, 100599. [Google Scholar] [CrossRef]
- Rajkumar, H.; Kumar, M.; Das, N.; Kumar, S.N.; Challa, H.R.; Nagpal, R. Effect of Probiotic Lactobacillus salivarius UBL S22 and Prebiotic Fructo-oligosaccharide on Serum Lipids, Inflammatory Markers, Insulin Sensitivity, and Gut Bacteria in Healthy Young Volunteers: A Randomized Controlled Single-Blind Pilot Study. J. Cardiovasc. Pharmacol. Ther. 2015, 20, 289–298. [Google Scholar] [CrossRef]
- Firouzi, S.; Majid, H.A.; Ismail, A.; Kamaruddin, N.A.; Barakatun-Nisak, M.-Y. Effect of multi-strain probiotics (multi-strain microbial cell preparation) on glycemic control and other diabetes-related outcomes in people with type 2 diabetes: A randomized controlled trial. Eur. J. Nutr. 2017, 56, 1535–1550. [Google Scholar] [CrossRef]
- Rioux, K.P.; Madsen, K.L.; Fedorak, R.N. The Role of Enteric Microflora in Inflammatory Bowel Disease: Human and Animal Studies with Probiotics and Prebiotics. Gastroenterol. Clin. N. Am. 2005, 34, 465–482. [Google Scholar] [CrossRef] [PubMed]
- Fontané, L.; Benaiges, D.; Goday, A.; Llauradó, G.; Pedro-Botet, J. Influence of the Microbiota and Probiotics in Obesity. Clin. Investig. Arterioscler. 2018, 30, 271–279. [Google Scholar] [CrossRef]
- Kootte, R.S.; Levin, E.; Salojärvi, J.; Smits, L.P.; Hartstra, A.V.; Udayappan, S.D.; Hermes, G.; Bouter, K.E.; Koopen, A.M.; Holst, J.J.; et al. Improvement of Insulin Sensitivity after Lean Donor Feces in Metabolic Syndrome Is Driven by Baseline Intestinal Microbiota Composition. Cell Metab. 2017, 26, 611–619.e6. [Google Scholar] [CrossRef] [PubMed]
- Qi, X.; Yun, C.; Sun, L.; Xia, J.; Wu, Q.; Wang, Y.; Wang, L.; Zhang, Y.; Liang, X.; Wang, L.; et al. Gut microbiota–bile acid–interleukin-22 axis orchestrates polycystic ovary syndrome. Nat. Med. 2019, 25, 1225–1233. [Google Scholar] [CrossRef] [PubMed]
- Beam, A.; Clinger, E.; Hao, L. Effect of Diet and Dietary Components on the Composition of the Gut Microbiota. Nutrients 2021, 13, 2795. [Google Scholar] [CrossRef] [PubMed]
- Wang, C.-H.; Yen, H.-R.; Lu, W.-L.; Ho, H.-H.; Lin, W.-Y.; Kuo, Y.-W.; Huang, Y.-Y.; Tsai, S.-Y.; Lin, H.-C. Adjuvant Probiotics of Lactobacillus salivarius subsp. salicinius AP-32, L. johnsonii MH-68, and Bifidobacterium animalis subsp. lactis CP-9 Attenuate Glycemic Levels and Inflammatory Cytokines in Patients with Type 1 Diabetes Mellitus. Front. Endocrinol. 2022, 13, 754401. [Google Scholar] [CrossRef]
- Hsieh, P.-S.; Ho, H.-H.; Tsao, S.P.; Hsieh, S.-H.; Lin, W.-Y.; Chen, J.-F.; Kuo, Y.-W.; Tsai, S.-Y.; Huang, H.-Y. Multi-strain probiotic supplement attenuates streptozotocin-induced type-2 diabetes by reducing inflammation and β-cell death in rats. PLoS ONE 2021, 16, e0251646. [Google Scholar] [CrossRef]
- Shyangdan, D.S.; Royle, P.; Clar, C.; Sharma, P.; Waugh, N.; Snaith, A. Glucagon-like peptide analogues for type 2 diabetes mellitus. Cochrane Database Syst. Rev. 2011, 2011, CD006423. [Google Scholar] [CrossRef]
- Hu, Y.; Ding, M.; Sampson, L.; Willett, W.C.; Manson, J.E.; Wang, M.; Rosner, B.; Hu, F.B.; Sun, Q. Intake of whole grain foods and risk of type 2 diabetes: Results from three prospective cohort studies. BMJ 2020, 370, m2206. [Google Scholar] [CrossRef]
- Candela, M.; Biagi, E.; Soverini, M.; Consolandi, C.; Quercia, S.; Severgnini, M.; Peano, C.; Turroni, S.; Rampelli, S.; Pozzilli, P.; et al. Modulation of gut microbiota dysbioses in type 2 diabetic patients by macrobiotic Ma-Pi 2 diet. Br. J. Nutr. 2016, 116, 80–93. [Google Scholar] [CrossRef]
- Fallucca, F.; Porrata, C.; Fallucca, S.; Pianesi, M. Influence of diet on gut microbiota, inflammation and type 2 diabetes mellitus. First experience with macrobiotic Ma-Pi 2 diet. Diabetes/Metabolism. Res. Rev. 2014, 30, 48–54. [Google Scholar] [CrossRef]
- Psichas, A.; Sleeth, M.L.; Murphy, K.G.; Brooks, L.; Bewick, G.A.; Hanyaloglu, A.C.; Ghatei, M.A.; Bloom, S.R.; Frost, G. The short chain fatty acid propionate stimulates GLP-1 and PYY secretion via free fatty acid receptor 2 in rodents. Int. J. Obes. 2015, 39, 424. [Google Scholar] [CrossRef]
- Sourabh, A.; Kanwar, S.S.; Sud, R.G.; Ghabru, A.; Sharma, O.P. Influence of phenolic compounds of Kangra tea [Camellia sinensis (L) O Kuntze] on bacterial pathogens and indigenous bacterial probiotics of Western Himalayas. Braz. J. Microbiol. 2013, 44, 709–715. [Google Scholar] [CrossRef]
- Shen, X.; Sun, X.; Xie, Q.; Liu, H.; Zhao, Y.; Pan, Y.; Hwang, C.-A.; Wu, V.C. Antimicrobial effect of blueberry (Vaccinium corymbosum L.) extracts against the growth of Listeria monocytogenes and Salmonella enteritidis. Food Control 2013, 35, 159–165. [Google Scholar] [CrossRef]
- Nohynek, L.J.; Alakomi, H.-L.; Kähkönen, M.P.; Heinonen, M.; Helander, I.M.; Oksman-Caldentey, K.-M.; Puupponen-Pimiä, R.H. Berry Phenolics: Antimicrobial Properties and Mechanisms of Action Against Severe Human Pathogens. Nutr. Cancer 2006, 54, 18–32. [Google Scholar] [CrossRef]
- Singh, R.; Chandrashekharappa, S.; Bodduluri, S.R.; Baby, B.V.; Hegde, B.; Kotla, N.G.; Hiwale, A.A.; Saiyed, T.; Patel, P.; Vijay-Kumar, M.; et al. Enhancement of the gut barrier integrity by a microbial metabolite through the Nrf2 pathway. Nat. Commun. 2019, 10, 89. [Google Scholar] [CrossRef] [PubMed]
- Banerjee, A.; Dhar, P. Amalgamation of polyphenols and probiotics induce health promotion. Crit. Rev. Food Sci. Nutr. 2019, 59, 2903–2926. [Google Scholar] [CrossRef] [PubMed]
- Rupasinghe, H.P.V.; Parmar, I.; Neir, S.V. Biotransformation of Cranberry Proanthocyanidins to Probiotic Metabolites by Lactobacillus rhamnosus Enhances Their Anticancer Activity in HepG2 Cells In Vitro. Oxidative Med. Cell. Longev. 2019, 2019, 4750795. [Google Scholar] [CrossRef]
- Dos Santos, A.S.; de Albuquerque, T.M.R.; Alves, J.L.d.B.; de Souza, E.L. Effects of Quercetin and Resveratrol on in vitro Properties Related to the Functionality of Potentially Probiotic Lactobacillus Strains. Front. Microbiol. 2019, 10, 2229. [Google Scholar] [CrossRef]
- Westfall, S.; Lomis, N.; Prakash, S. A novel polyphenolic prebiotic and probiotic formulation have synergistic effects on the gut microbiota influencing Drosophila melanogaster physiology. Artif. Cells Nanomed. Biotechnol. 2018, 46, 441–455. [Google Scholar] [CrossRef]
- Watson, H.; Mitra, S.; Croden, F.C.; Taylor, M.; Wood, H.M.; Perry, S.L.; Spencer, J.A.; Quirke, P.; Toogood, G.J.; Lawton, C.L.; et al. A randomised trial of the effect of omega-3 polyunsaturated fatty acid supplements on the human intestinal microbiota. Gut 2018, 67, 1974–1983. [Google Scholar] [CrossRef]
- Shama, S.; Liu, W. Omega-3 Fatty Acids and Gut Microbiota: A Reciprocal Interaction in Nonalcoholic Fatty Liver Disease. Dig. Dis. Sci. 2020, 65, 906–910. [Google Scholar] [CrossRef] [PubMed]
- De Groot, P.; Nikolic, T.; Pellegrini, S.; Sordi, V.; Imangaliyev, S.; Rampanelli, E.; Hanssen, N.; Attaye, I.; Bakker, G.; Duinkerken, G.; et al. Faecal microbiota transplantation halts progression of human new-onset type 1 diabetes in a randomised controlled trial. Gut 2020, 70, 92–105. [Google Scholar] [CrossRef]
- McLeod, K.; Mason, L.; Mariño, E. Transplantation of Fecal Microbiota Shaped by Diet. Bio-Protocol 2018, 8, e2683. [Google Scholar] [CrossRef]
- Turnbaugh, P.J.; Ridaura, V.K.; Faith, J.J.; Rey, F.E.; Knight, R.; Gordon, J.I. The Effect of Diet on the Human Gut Microbiome: A Metagenomic Analysis in Humanized Gnotobiotic Mice. Sci. Transl. Med. 2009, 1, 6ra14. [Google Scholar] [CrossRef]
- Virili, C.; Stramazzo, I.; Centanni, M. Gut microbiome and thyroid autoimmunity. Best. Pract. Res. Clin. Endocrinol. Metab. 2021, 35, 101506. [Google Scholar] [CrossRef]
- Virili, C.; Fallahi, P.; Antonelli, A.; Benvenga, S.; Centanni, M. Gut microbiota and Hashimoto’s thyroiditis. Rev. Endocr. Metab. Disord. 2018, 19, 293–300. [Google Scholar] [CrossRef] [PubMed]
- Ishaq, H.M.; Mohammad, I.S.; Shahzad, M.; Ma, C.; Raza, M.A.; Wu, X.; Guo, H.; Shi, P.; Xu, J. Molecular Alteration Analysis of Human Gut Microbial Composition in Graves’ disease Patients. Int. J. Biol. Sci. 2018, 14, 1558–1570. [Google Scholar] [CrossRef] [PubMed]
- Li, T.; Chiang, J.Y.L. Bile acid-based therapies for non-alcoholic steatohepatitis and alcoholic liver disease. HepatoBiliary Surg. Nutr. 2020, 9, 152–169. [Google Scholar] [CrossRef]
- Torres, P.J.; Siakowska, M.; Banaszewska, B.; Pawelczyk, L.; Duleba, A.J.; Kelley, S.T.; Thackray, V.G. Gut Microbial Diversity in Women with Polycystic Ovary Syndrome Correlates with Hyperandrogenism. J. Clin. Endocrinol. Metab. 2018, 103, 1502–1511. [Google Scholar] [CrossRef] [PubMed]
- Bansal, A.; Henao-Mejia, J.; Simmons, R.A. Immune System: An Emerging Player in Mediating Effects of Endocrine Disruptors on Metabolic Health. Endocrinology 2017, 159, 32–45. [Google Scholar] [CrossRef]
- Kwaifa, I.K.; Bahari, H.; Yong, Y.K.; Noor, S.M. Endothelial Dysfunction in Obesity-Induced Inflammation: Molecular Mechanisms and Clinical Implications. Biomolecules 2020, 10, 291. [Google Scholar] [CrossRef] [PubMed]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2024 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Clemente-Suárez, V.J.; Redondo-Flórez, L.; Rubio-Zarapuz, A.; Martín-Rodríguez, A.; Tornero-Aguilera, J.F. Microbiota Implications in Endocrine-Related Diseases: From Development to Novel Therapeutic Approaches. Biomedicines 2024, 12, 221. https://doi.org/10.3390/biomedicines12010221
Clemente-Suárez VJ, Redondo-Flórez L, Rubio-Zarapuz A, Martín-Rodríguez A, Tornero-Aguilera JF. Microbiota Implications in Endocrine-Related Diseases: From Development to Novel Therapeutic Approaches. Biomedicines. 2024; 12(1):221. https://doi.org/10.3390/biomedicines12010221
Chicago/Turabian StyleClemente-Suárez, Vicente Javier, Laura Redondo-Flórez, Alejandro Rubio-Zarapuz, Alexandra Martín-Rodríguez, and José Francisco Tornero-Aguilera. 2024. "Microbiota Implications in Endocrine-Related Diseases: From Development to Novel Therapeutic Approaches" Biomedicines 12, no. 1: 221. https://doi.org/10.3390/biomedicines12010221
APA StyleClemente-Suárez, V. J., Redondo-Flórez, L., Rubio-Zarapuz, A., Martín-Rodríguez, A., & Tornero-Aguilera, J. F. (2024). Microbiota Implications in Endocrine-Related Diseases: From Development to Novel Therapeutic Approaches. Biomedicines, 12(1), 221. https://doi.org/10.3390/biomedicines12010221