• Title/Summary/Keyword: gut-brain axis

Search Result 26, Processing Time 0.026 seconds

Effect of Lactobacillus dominance modified by Korean Red Ginseng on the improvement of Alzheimer's disease in mice

  • Lee, Mijung;Lee, So-Hee;Kim, Min-Soo;Ahn, Kwang-Sung;Kim, Manho
    • Journal of Ginseng Research
    • /
    • v.46 no.3
    • /
    • pp.464-472
    • /
    • 2022
  • Background: Gut microbiota influence the central nervous system through gut-brain-axis. They also affect the neurological disorders. Gut microbiota differs in patients with Alzheimer's disease (AD), as a potential factor that leads to progression of AD. Oral intake of Korean Red Ginseng (KRG) improves the cognitive functions. Therefore, it can be proposed that KRG affect the microbiota on the gut-brain-axis to the brain. Methods: Tg2576 were used for the experimental model of AD. They were divided into four groups: wild type (n = 6), AD mice (n = 6), AD mice with 30 mg/kg/day (n = 6) or 100 mg/kg/day (n = 6) of KRG. Following two weeks, changes in gut microbiota were analyzed by Illumina HiSeq4000 platform 16S gene sequencing. Microglial activation were evaluated by quantitative Western blot analyses of Iba-1 protein. Claudin-5, occludin, laminin and CD13 assay were conducted for Blood-brain barrier (BBB) integrity. Amyloid beta (Aβ) accumulation demonstrated through Aβ 42/40 ratio was accessed by ELISA, and cognition were monitored by Novel object location test. Results: KRG improved the cognitive behavior of mice (30 mg/kg/day p < 0.05; 100 mg/kg/day p < 0.01), and decreased Aβ 42/40 ratio (p < 0.01) indicating reduced Aβ accumulation. Increased Iba-1 (p < 0.001) for reduced microglial activation, and upregulation of Claudin-5 (p < 0.05) for decreased BBB permeability were shown. In particular, diversity of gut microbiota was altered (30 mg/kg/day q-value<0.05), showing increased population of Lactobacillus species. (30 mg/kg/day 411%; 100 mg/kg/day 1040%). Conclusions: KRG administration showed the Lactobacillus dominance in the gut microbiota. Improvement of AD pathology by KRG can be medicated through gut-brain axis in mice model of AD.

Potential role of phytochemicals in brain plasticity: Focus on polyunsaturated fatty acids

  • Yook, Jang Soo;Lee, Minchul
    • Korean Journal of Exercise Nutrition
    • /
    • v.24 no.1
    • /
    • pp.14-18
    • /
    • 2020
  • [Purpose] Functional foods are thought to strongly influence the structure and function of the brain. Previous studies have reported that brain-boosting diets may enhance neuroprotective functions. Certain foods are particularly rich in nutrients like phytochemicals that are known to support brain plasticity; such foods are commonly referred to as brain foods. [Methods] In this review, we briefly explore the scientific evidence supporting the neuroprotective activity of a number of phytochemicals with a focus on phenols and polyunsaturated fatty acids such as flavonoid, olive oil, and omega-3 fatty acid. [Results] The aim of this study was to systematically examine the primary issues related to phytochemicals in the brain. These include (a) the brain-gut-microbiome axis; (b) the effects of phytochemicals on gut microbiome and their potential role in brain plasticity; (c) the role of polyunsaturated fatty acids in brain health; and (d) the effects of nutrition and exercise on brain function. [Conclusion] This review provides evidence supporting the view that phytochemicals from medicinal plants play a vital role in maintaining brain plasticity by influencing the brain-gut-microbiome axis. The consumption of brain foods may have neuroprotective effects, thus protecting against neurodegenerative disorders and promoting brain health.

Alteration of Gut Microbiota in Autism Spectrum Disorder: An Overview

  • Oh, Donghun;Cheon, Keun-Ah
    • Journal of the Korean Academy of Child and Adolescent Psychiatry
    • /
    • v.31 no.3
    • /
    • pp.131-145
    • /
    • 2020
  • The microbiota-gut-brain axis, which refers to the bidirectional communication pathway between gut bacteria and the central nervous system, has a profound effect on important brain processes, from the synthesis of neurotransmitters to the modulation of complex behaviors such as sociability and anxiety. Previous studies have revealed that the gut microbiota is potentially related to not only gastrointestinal disturbances, but also social impairment and repetitive behavior-core symptoms of autism spectrum disorder (ASD). Although studies have been conducted to characterize the microbial composition in patients with ASD, the results are heterogeneous. Nevertheless, it is clear that there is a difference in the composition of the gut microbiota between ASD and typically developed individuals, and animal studies have repeatedly suggested that the gut microbiota plays an important role in ASD pathophysiology. This possibility is supported by abnormalities in metabolites produced by the gut microbiota and the association between altered immune responses and the gut microbiota observed in ASD patients. Based on these findings, various attempts have been made to use the microbiota in ASD treatment. The results reported to date suggest that microbiota-based therapies may be effective for ASD, but largescale, well-designed studies are needed to confirm this.

Effects of red ginseng on gut, microbiota, and brain in a mouse model of post-infectious irritable bowel syndrome

  • Yu, Seonhye;Chun, Eunho;Ji, Yeounjung;Lee, Young Joo;Jin, Mirim
    • Journal of Ginseng Research
    • /
    • v.45 no.6
    • /
    • pp.706-716
    • /
    • 2021
  • Background: Irritable bowel syndrome (IBS), the most common functional gastrointestinal disorder, is characterized by chronic abdominal pain and bowel habit changes. Although diverse complicated etiologies are involved in its pathogenesis, a dysregulated gut-brain axis may be an important factor. Red ginseng (RG), a traditional herbal medicine, is proven to have anti-inflammatory effects and improve brain function; however, these effects have not been investigated in IBS. Methods: Three-day intracolonic zymosan injections were used to induce post-infectious human IBS-like symptoms in mice. The animals were randomized to receive either phosphate-buffered saline (CG) or RG (30/100/300 mg/kg) for 10 days. Amitriptyline and sulfasalazine were used as positive controls. Macroscopic scoring was performed on day 4. Visceral pain and anxiety-like behaviors were assessed by colorectal distension and elevated plus maze and open field tests, respectively, on day 10. Next-generation sequencing of gut microbiota was performed, and biomarkers involved in gut-brain axis responses were analyzed. Results: Compared to CG, RG significantly decreased the macroscopic score, frequency of visceral pain, and anxiety-like behavior in the IBS mice. These effects were comparable to those after sulfasalazine and amitriptyline treatments. Moreover, RG significantly increased the proliferation of beneficial microbes, including Lactobacillus johnsonii, Lactobacillus reuteri, and Parabacteroides goldsteinii. RG significantly suppressed expression of IL-1β and c-fos in the gut and prefrontal cortex, respectively. Further, it restored the plasma levels of corticosterone to within the normal range, accompanied by an increase in adrenocorticotropic hormone. Conclusion: RG may be a potential therapeutic option for the management of human IBS.

Effect of Duloxetine in Functional Gastrointestinal Disorder : In the Perspective of 'Brain-Gut Axis' (기능성 위장관 장애에서 Duloxetine의 효과 : '뇌-장관 축' 모델을 중심으로)

  • Lee, Sang-Shin;Park, Si-Sung
    • Korean Journal of Psychosomatic Medicine
    • /
    • v.20 no.2
    • /
    • pp.135-138
    • /
    • 2012
  • The pathophysiology of functional gastrointestinal disorder(FGID) is not completely understood, but the importance of the 'Brain-Gut Axis(BGA)' model in FGID is being increasingly recognized. The BGA model is a bidirectional, hard-wired and homeostatic relationship between the central nervous system(CNS) and the enteric nervous system(ENS) via neural, neurohormonal and neuroimmunological pathways. In addition, the BGA model would provide a rationale for the use of psychotropics on FGID. The authors experienced two cases in which duloxetine, a serotonin-norepinephrine reuptake inhibitor, was effective in relieving FGID symptoms as well as psychiatric symptoms such as depression and hypochondriacal anxiety. Therefore we discuss the vignettes from the perspective of BGA theory. Duloxetine showed efficacy in these two patients by reducing visceral hypersensivity (bottom-up regulation) and by relieving depression and anxiety(top-down regulation).

  • PDF

Fishing for synucleinopathy models

  • Noor, Suzita Mohd;Norazit, Anwar
    • Fisheries and Aquatic Sciences
    • /
    • v.25 no.3
    • /
    • pp.117-139
    • /
    • 2022
  • Synucleinopathies such as Parkinson's disease (PD) are incurable neurodegenerative conditions characterised by the abnormal aggregation of α-synuclein protein in neuronal cells. In PD, fibrillary synuclein aggregation forms Lewy bodies and Lewy neurites in the substantia nigra and cortex on the brain. Dementia with Lewy bodies and multiple system atrophy are also associated with α-synuclein protein abnormalities. α-synuclein is one of three synuclein proteins, and while its precise function is still unknown, one hypothesis posits that α-synuclein propagates from the enteric nervous system through the vagus nerve and into the brain, resulting in synucleinopathy. Studies on synucleinopathies should thus encompass not only the central nervous system but must necessarily include the gut and microbiome. The zebrafish (Danio rerio) is a well-established model for human neuronal pathologies and have been used in studies ranging from genetic models of hereditary disorders to neurotoxin-induced neurodegeneration as well as gut-brain-axis studies. There is significant genetic homology between zebrafish and mammalian vertebrates which is what makes the zebrafish so amenable to modelling human conditions but in the case of synucleinopathies, the zebrafish notably does not possess an α-synuclein homolog. Synuclein orthologs are present in the zebrafish however, and transgenic zebrafish that carry human α-synuclein have been generated. In addition, the zebrafish is a highly advantageous model and ideal replacement for reducing the use of mammalian models. This review discusses the application of the zebrafish as a model for synucleinopathies in efforts to further understand synuclein function and explore therapeutic strategies.

6-Shogaol, an Active Ingredient of Ginger, Improves Intestinal and Brain Abnormalities in Proteus Mirabilis-Induced Parkinson's Disease Mouse Model

  • Eugene Huh;Jin Gyu Choi;Yujin Choi;In Gyoung Ju;Dongjin Noh;Dong-yun Shin;Dong Hyun Kim;Hi-Joon Park;Myung Sook Oh
    • Biomolecules & Therapeutics
    • /
    • v.31 no.4
    • /
    • pp.417-424
    • /
    • 2023
  • Parkinson's disease (PD) which has various pathological mechanisms, recently, it is attracting attention to the mechanism via microbiome-gut-brain axis. 6-Shogaol, a representative compound of ginger, have been known for improving PD phenotypes by reducing neuroinflammatory responses. In the present study, we investigated whether 6-shogaol and ginger attenuate degeneration induced by Proteus mirabilis (P. mirabilis) on the intestine and brain, simultaneously. C57BL/6J mice received P. mirabilis for 5 days. Ginger (300 mg/kg) and 6-shogaol (10 mg/kg) were treated by gavage feeding for 22 days including the period of P. mirabilis treatment. Results showed that 6-shogaol and ginger improved motor dysfunction and dopaminergic neuronal death induced by P. mirabilis treatment. In addition, they suppressed P. mirabilis-induced intestinal barrier disruption, pro-inflammatory signals such as toll-like receptor and TNF-α, and intestinal α-synuclein aggregation. Moreover, ginger and 6-shogaol significantly inhibited neuroinflammation and α-synuclein in the brain. Taken together, 6-shogaol and ginger have the potential to ameliorate PD-like motor behavior and degeneration of dopaminergic neurons induced by P. mirabilis in mice. Here, these findings are meaningful in that they provide the first experimental evidence that 6-shogaol might attenuate PD via regulating gut-brain axis.

Breastfeeding and Melatonin (모유 수유와 멜라토닌)

  • Song, Minyu;Park, Won Seo;Yoo, Jayeon;Ham, Jun-Sang
    • Journal of Dairy Science and Biotechnology
    • /
    • v.36 no.3
    • /
    • pp.133-145
    • /
    • 2018
  • Breastfeeding is highly recommended due to its benefits for both the infant and mother; however, most mothers predominantly use formula feed. Breastfeeding affords protection against a wide variety of medical conditions that may emerge at different time points over the lifespan, including hospital admissions for respiratory infections and neonatal fever, offspring childhood obesity, and cancer as well as cardiovascular disease, hyperlipidemia, hypertension, and diabetes. Moreover, breastfeeding is expected to decrease the risk of adolescent depression and other psychopathologies. It is also important for the development of the gut, gut-brain axis, and immune system, and night-time breast milk is likely to have higher antioxidant, anti-inflammatory, and immune regulatory effects due to the impact of breast milk melatonin on the infant's developing microbiome and gut permeability. Melatonin can be added to a night-time-specific formula feed; however, it is not included in the Korean Food Additive Codex.

Water Extract of Ecklonia cava Protects against Fine Dust (PM2.5)-Induced Health Damage by Regulating Gut Health

  • Park, Seon Kyeong;Kang, Jin Yong;Kim, Jong Min;Kim, Min Ji;Lee, Hyo Lim;Moon, Jong Hyun;Jeong, Hye Rin;Kim, Hyun-Jin;Heo, Ho Jin
    • Journal of Microbiology and Biotechnology
    • /
    • v.32 no.7
    • /
    • pp.927-937
    • /
    • 2022
  • To confirm the therapeutic effect of the water extract from Ecklonia cava (WEE) against PM2.5 induced systemic health damage, we evaluated gut health with a focus on the microbiota and metabolites. Systemic damage in mice was induced through PM2.5 exposure for 12 weeks in a whole-body chamber. After exposure for 12 weeks, body weight and food intake decreased, and WEE at 200 mg/kg body weight (mpk) alleviated these metabolic efficiency changes. In addition, PM2.5 induced changes in the length of the colon and fecal water content. The administration of the WEE at 200 mpk oral dose effectively reduced changes in the colon caused by PM2.5 exposure. We also attempted to confirm whether the effect of the WEE is mediated via regulation of the microbiota-gut-brain axis in mice with PM2.5 induced systemic damage. We examined changes in the fecal microbiota and gut metabolites such as short-chain fatty acids (SCFAs) and kynurenine metabolites. In the PM2.5 exposed group, a decrease in the abundance of Lactobacillus (Family: Lactobacillaceae) and an increase in the abundance of Alistipes (Family: Rikenellaceae) were observed, and the administration of the WEE showed a beneficial effect on the gut microbiota. In addition, the WEE effectively increased the levels of SCFAs (acetate, propionate, and butyrate). Furthermore, kynurenic acid (KYNA), which is a critical neuroprotective metabolite in the gut-brain axis, was increased by the administration of the WEE. Our findings suggest that the WEE could be used as a potential therapeutic against PM2.5 induced health damage by regulating gut function.

Understanding Neurogastroenterology From Neuroimaging Perspective: A Comprehensive Review of Functional and Structural Brain Imaging in Functional Gastrointestinal Disorders

  • Kano, Michiko;Dupont, Patrick;Aziz, Qasim;Fukudo, Shin
    • Journal of Neurogastroenterology and Motility
    • /
    • v.24 no.4
    • /
    • pp.512-527
    • /
    • 2018
  • This review provides a comprehensive overview of brain imaging studies of the brain-gut interaction in functional gastrointestinal disorders (FGIDs). Functional neuroimaging studies during gut stimulation have shown enhanced brain responses in regions related to sensory processing of the homeostatic condition of the gut (homeostatic afferent) and responses to salience stimuli (salience network), as well as increased and decreased brain activity in the emotional response areas and reduced activation in areas associated with the top-down modulation of visceral afferent signals. Altered central regulation of the endocrine and autonomic nervous responses, the key mediators of the brain-gut axis, has been demonstrated. Studies using resting-state functional magnetic resonance imaging reported abnormal local and global connectivity in the areas related to pain processing and the default mode network (a physiological baseline of brain activity at rest associated with self-awareness and memory) in FGIDs. Structural imaging with brain morphometry and diffusion imaging demonstrated altered gray- and white-matter structures in areas that also showed changes in functional imaging studies, although this requires replication. Molecular imaging by magnetic resonance spectroscopy and positron emission tomography in FGIDs remains relatively sparse. Progress using analytical methods such as machine learning algorithms may shift neuroimaging studies from brain mapping to predicting clinical outcomes. Because several factors contribute to the pathophysiology of FGIDs and because its population is quite heterogeneous, a new model is needed in future studies to assess the importance of the factors and brain functions that are responsible for an optimal homeostatic state.