1
|
Jiang Y, Huang Z, Sun W, Huang J, Xu Y, Liao Y, Jin T, Li Q, Ho IHT, Zou Y, Zhu W, Li Q, Qin F, Zhang X, Shi S, Zhang N, Yang S, Xie W, Wu S, Tan L, Zhang L, Chen H, Gin T, Chan MTV, Wu WKK, Xiao L, Liu X. Roseburia intestinalis-derived butyrate alleviates neuropathic pain. Cell Host Microbe 2025; 33:104-118.e7. [PMID: 39706182 DOI: 10.1016/j.chom.2024.11.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 10/30/2024] [Accepted: 11/25/2024] [Indexed: 12/23/2024]
Abstract
Approximately 20% of patients with shingles develop postherpetic neuralgia (PHN). We investigated the role of gut microbiota in shingle- and PHN-related pain. Patients with shingles or PHN exhibited significant alterations in their gut microbiota with microbial markers predicting PHN development among patients with shingles. Functionally, fecal microbiota transplantation from patients with PHN to mice heightened pain sensitivity. Administration of Roseburia intestinalis, a bacterium both depleted in patients with shingles and PHN, alleviated peripheral nerve injury-induced pain in mice. R. intestinalis enhanced vagal neurotransmission to the nucleus tractus solitarius (NTS) to suppress the central amygdala (CeA), a brain region involved in pain perception. R. intestinalis-generated butyrate activated vagal neurons through the receptor, G protein-coupled receptor 41 (GPR41). Vagal knockout of Gpr41 abolished the effects of R. intestinalis on the NTS-CeA circuit and reduced pain behaviors. Overall, we established a microbiota-based model for PHN risk assessment and identified R. intestinalis as a potential pain-alleviating probiotic.
Collapse
Affiliation(s)
- Yanjun Jiang
- Department of Anaesthesia and Intensive Care, The Chinese University of Hong Kong, Hong Kong SAR, China; Peter Hung Pain Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Ziheng Huang
- Department of Anaesthesia and Intensive Care, The Chinese University of Hong Kong, Hong Kong SAR, China; Peter Hung Pain Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Wuping Sun
- Department of Pain Medicine and Shenzhen Municipal Key Laboratory for Pain Medicine, Shenzhen Nanshan People's Hospital and the 6th Affiliated Hospital of Shenzhen University Medical School, National Key Clinical Pain Medicine of China, Shenzhen 518060, China
| | - Jiabin Huang
- Department of Pain Medicine and Shenzhen Municipal Key Laboratory for Pain Medicine, Shenzhen Nanshan People's Hospital and the 6th Affiliated Hospital of Shenzhen University Medical School, National Key Clinical Pain Medicine of China, Shenzhen 518060, China
| | - Yunlong Xu
- Shenzhen Key Laboratory of Drug Addiction, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; Department of Neonatology, Shenzhen Maternity & Child Healthcare Hospital, The First School of Clinical Medicine, Southern Medical University, Shenzhen 518055, China
| | - Yuliang Liao
- Department of Pain Medicine and Shenzhen Municipal Key Laboratory for Pain Medicine, Shenzhen Nanshan People's Hospital and the 6th Affiliated Hospital of Shenzhen University Medical School, National Key Clinical Pain Medicine of China, Shenzhen 518060, China
| | - Tingting Jin
- Department of Anaesthesia and Intensive Care, The Chinese University of Hong Kong, Hong Kong SAR, China; Peter Hung Pain Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Qing Li
- Department of Anaesthesia and Intensive Care, The Chinese University of Hong Kong, Hong Kong SAR, China; Peter Hung Pain Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China; Dermatology Hospital, Southern Medical University, Guangzhou, China
| | - Idy Hiu Ting Ho
- Department of Anaesthesia and Intensive Care, The Chinese University of Hong Kong, Hong Kong SAR, China; Peter Hung Pain Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Yidan Zou
- Department of Anaesthesia and Intensive Care, The Chinese University of Hong Kong, Hong Kong SAR, China; Peter Hung Pain Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Wenyi Zhu
- Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Qian Li
- Department of Anaesthesia and Intensive Care, The Chinese University of Hong Kong, Hong Kong SAR, China; Peter Hung Pain Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Fenfen Qin
- Department of Anaesthesia and Intensive Care, The Chinese University of Hong Kong, Hong Kong SAR, China; Peter Hung Pain Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Xinyi Zhang
- Department of Anaesthesia and Intensive Care, The Chinese University of Hong Kong, Hong Kong SAR, China; Peter Hung Pain Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Shuqi Shi
- Department of Pain Medicine and Shenzhen Municipal Key Laboratory for Pain Medicine, Shenzhen Nanshan People's Hospital and the 6th Affiliated Hospital of Shenzhen University Medical School, National Key Clinical Pain Medicine of China, Shenzhen 518060, China
| | - Na Zhang
- Department of Anaesthesia and Intensive Care, The Chinese University of Hong Kong, Hong Kong SAR, China; Peter Hung Pain Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Shaomin Yang
- Department of Pain Medicine and Shenzhen Municipal Key Laboratory for Pain Medicine, Shenzhen Nanshan People's Hospital and the 6th Affiliated Hospital of Shenzhen University Medical School, National Key Clinical Pain Medicine of China, Shenzhen 518060, China
| | - Wenhui Xie
- Department of Pain Medicine and Shenzhen Municipal Key Laboratory for Pain Medicine, Shenzhen Nanshan People's Hospital and the 6th Affiliated Hospital of Shenzhen University Medical School, National Key Clinical Pain Medicine of China, Shenzhen 518060, China
| | - Songbin Wu
- Department of Pain Medicine and Shenzhen Municipal Key Laboratory for Pain Medicine, Shenzhen Nanshan People's Hospital and the 6th Affiliated Hospital of Shenzhen University Medical School, National Key Clinical Pain Medicine of China, Shenzhen 518060, China
| | - Likai Tan
- Department of Anaesthesia and Intensive Care, The Chinese University of Hong Kong, Hong Kong SAR, China; Peter Hung Pain Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Lin Zhang
- Department of Anaesthesia and Intensive Care, The Chinese University of Hong Kong, Hong Kong SAR, China; Microbiota I-Center (MagIC), The Chinese University of Hong Kong, Hong Kong SAR, China; Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Huarong Chen
- Department of Anaesthesia and Intensive Care, The Chinese University of Hong Kong, Hong Kong SAR, China; Peter Hung Pain Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Tony Gin
- Department of Anaesthesia and Intensive Care, The Chinese University of Hong Kong, Hong Kong SAR, China; Peter Hung Pain Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Matthew Tak Vai Chan
- Department of Anaesthesia and Intensive Care, The Chinese University of Hong Kong, Hong Kong SAR, China; Peter Hung Pain Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China.
| | - William Ka Kei Wu
- Department of Anaesthesia and Intensive Care, The Chinese University of Hong Kong, Hong Kong SAR, China; Peter Hung Pain Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China; State Key Laboratory of Digestive Disease Institute of Digestive Disease and Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China.
| | - Lizu Xiao
- Department of Pain Medicine and Shenzhen Municipal Key Laboratory for Pain Medicine, Shenzhen Nanshan People's Hospital and the 6th Affiliated Hospital of Shenzhen University Medical School, National Key Clinical Pain Medicine of China, Shenzhen 518060, China.
| | - Xiaodong Liu
- Department of Anaesthesia and Intensive Care, The Chinese University of Hong Kong, Hong Kong SAR, China; Peter Hung Pain Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China.
| |
Collapse
|
2
|
Ohara TE, Hsiao EY. Microbiota-neuroepithelial signalling across the gut-brain axis. Nat Rev Microbiol 2025:10.1038/s41579-024-01136-9. [PMID: 39743581 DOI: 10.1038/s41579-024-01136-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/25/2024] [Indexed: 01/04/2025]
Abstract
Research over the past two decades has established a remarkable ability of the gut microbiota to modulate brain activity and behaviour. Conversely, signals from the brain can influence the composition and function of the gut microbiota. This bidirectional communication across the gut microbiota-brain axis, involving multiple biochemical and cellular mediators, is recognized as a major brain-body network that integrates cues from the environment and the body's internal state. Central to this network is the gut sensory system, formed by intimate connections between chemosensory epithelial cells and sensory nerve fibres, that conveys interoceptive signals to the central nervous system. In this Review, we provide a broad overview of the pathways that connect the gut and the brain, and explore the complex dialogue between microorganisms and neurons at this emerging intestinal neuroepithelial interface. We highlight relevant microbial factors, endocrine cells and neural mechanisms that govern gut microbiota-brain interactions and their implications for gastrointestinal and neuropsychiatric health.
Collapse
Affiliation(s)
- Takahiro E Ohara
- Department of Integrative Biology and Physiology, University of California, Los Angeles, CA, USA.
| | - Elaine Y Hsiao
- Department of Integrative Biology and Physiology, University of California, Los Angeles, CA, USA.
- UCLA Goodman-Luskin Microbiome Center, Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, UCLA David Geffen School of Medicine, Los Angeles, CA, USA.
| |
Collapse
|
3
|
Cheng C, Xue X, Jiao Y, Du M, Zhang M, Zeng X, Sun JB, Qin W, Deng H, Yang XJ. Can earlobe stimulation serve as a sham for transcutaneous auricular vagus stimulation? Evidence from an alertness study following sleep deprivation. Psychophysiology 2025; 62:e14744. [PMID: 39727264 DOI: 10.1111/psyp.14744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 11/17/2024] [Accepted: 11/29/2024] [Indexed: 12/28/2024]
Abstract
Transcutaneous auricular vagus nerve stimulation (taVNS) has garnered increasing attention as a safe and effective peripheral neuromodulation technique in various clinical and cognitive neuroscience fields. However, there is ongoing debate about whether the commonly used earlobe control interferes with the objective assessment of taVNS regulatory effects. This study aims to further explore the regulatory effects of taVNS and earlobe stimulation (ES) on alertness levels and physiological indicators following 24 h of sleep deprivation (SD), based on previous findings that both taVNS and ES showed significant positive effects. The goal is to evaluate whether ES can serve as a neutral sham condition. Using a within-subject randomized experimental design involving 56 participants, we assessed alertness, heart rate variability (HRV), and salivary alpha-amylase (sAA) levels in the morning of the first day. After 24 h of SD and 30 min of either taVNS or ES intervention, these indicators were re-evaluated, and the changes in both groups were analyzed. The results indicated that both taVNS and ES improved alertness levels following SD. However, taVNS significantly increased sAA levels, indicating activation of the LC-NE system, whereas ES significantly increased HR and reduced HRV, promoting sympathetic nervous activity. Additionally, the regulatory effect of taVNS on the alertness showed a higher correlation with SD impairment. Although taVNS and ES may involve different and separable neuromodulation mechanisms, both can enhance alertness following SD. Future studies should carefully consider the potential regulatory effects of ES when using it as a sham condition in taVNS research.
Collapse
Affiliation(s)
- Chen Cheng
- Engineering Research Center of Molecular and Neuro Imaging of the Ministry of Education, School of Life Science and Technology, Xidian University, Xi'an, China
- Intelligent Non-invasive Neuromodulation Technology and Transformation Joint Laboratory, Xidian University, Xi'an, China
| | - Xinxin Xue
- Engineering Research Center of Molecular and Neuro Imaging of the Ministry of Education, School of Life Science and Technology, Xidian University, Xi'an, China
- Intelligent Non-invasive Neuromodulation Technology and Transformation Joint Laboratory, Xidian University, Xi'an, China
| | - Yunyun Jiao
- Engineering Research Center of Molecular and Neuro Imaging of the Ministry of Education, School of Life Science and Technology, Xidian University, Xi'an, China
- Intelligent Non-invasive Neuromodulation Technology and Transformation Joint Laboratory, Xidian University, Xi'an, China
| | - Mengyu Du
- Engineering Research Center of Molecular and Neuro Imaging of the Ministry of Education, School of Life Science and Technology, Xidian University, Xi'an, China
- Intelligent Non-invasive Neuromodulation Technology and Transformation Joint Laboratory, Xidian University, Xi'an, China
| | - Mengkai Zhang
- Engineering Research Center of Molecular and Neuro Imaging of the Ministry of Education, School of Life Science and Technology, Xidian University, Xi'an, China
- Intelligent Non-invasive Neuromodulation Technology and Transformation Joint Laboratory, Xidian University, Xi'an, China
| | - Xiao Zeng
- Engineering Research Center of Molecular and Neuro Imaging of the Ministry of Education, School of Life Science and Technology, Xidian University, Xi'an, China
- Intelligent Non-invasive Neuromodulation Technology and Transformation Joint Laboratory, Xidian University, Xi'an, China
- Guangzhou Institute of Technology, Xidian University, Xi'an, China
| | - Jin-Bo Sun
- Engineering Research Center of Molecular and Neuro Imaging of the Ministry of Education, School of Life Science and Technology, Xidian University, Xi'an, China
- Intelligent Non-invasive Neuromodulation Technology and Transformation Joint Laboratory, Xidian University, Xi'an, China
- Guangzhou Institute of Technology, Xidian University, Xi'an, China
| | - Wei Qin
- Engineering Research Center of Molecular and Neuro Imaging of the Ministry of Education, School of Life Science and Technology, Xidian University, Xi'an, China
- Intelligent Non-invasive Neuromodulation Technology and Transformation Joint Laboratory, Xidian University, Xi'an, China
- Guangzhou Institute of Technology, Xidian University, Xi'an, China
| | - Hui Deng
- Engineering Research Center of Molecular and Neuro Imaging of the Ministry of Education, School of Life Science and Technology, Xidian University, Xi'an, China
- Center of Journal Publication, Xidian University, Xi'an, China
| | - Xue-Juan Yang
- Engineering Research Center of Molecular and Neuro Imaging of the Ministry of Education, School of Life Science and Technology, Xidian University, Xi'an, China
- Intelligent Non-invasive Neuromodulation Technology and Transformation Joint Laboratory, Xidian University, Xi'an, China
- Guangzhou Institute of Technology, Xidian University, Xi'an, China
| |
Collapse
|
4
|
Yao M, Qu Y, Zheng Y, Guo H. The effect of exercise on depression and gut microbiota: Possible mechanisms. Brain Res Bull 2025; 220:111130. [PMID: 39557221 DOI: 10.1016/j.brainresbull.2024.111130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Revised: 11/09/2024] [Accepted: 11/12/2024] [Indexed: 11/20/2024]
Abstract
Exercise can effectively prevent and treat depression and anxiety, with gut microbiota playing a crucial role in this process. Studies have shown that exercise can influence the diversity and composition of gut microbiota, which in turn affects depression through immune, endocrine, and neural pathways in the gut-brain axis. The effectiveness of exercise varies based on its type, intensity, and duration, largely due to the different changes in gut microbiota. This article summarizes the possible mechanisms by which exercise affects gut microbiota and how gut microbiota influences depression. Additionally, we reviewed literature on the effects of exercise on depression at different intensities, types, and durations to provide a reference for future exercise-based therapies for depression.
Collapse
Affiliation(s)
- Mingchen Yao
- School of Sports Medicine and Rehabilitation, Beijing Sport University, Beijing, China
| | - Yaqi Qu
- School of Sports Medicine and Rehabilitation, Beijing Sport University, Beijing, China
| | - Yalin Zheng
- School of Sports Medicine and Rehabilitation, Beijing Sport University, Beijing, China
| | - Hao Guo
- School of Sports Medicine and Rehabilitation, Beijing Sport University, Beijing, China.
| |
Collapse
|
5
|
Sun L, Bai Y, Kang F, Lei Y. Biosignals in the Gut-Brain Axis Transmission: Function and Detection. ACS APPLIED MATERIALS & INTERFACES 2024; 16:67045-67053. [PMID: 38572786 DOI: 10.1021/acsami.4c00194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/05/2024]
Abstract
The gut-brain axis (GBA) is an important information pathway connecting the brain, the central nervous system (CNS), and the gastrointestinal (GI) tract. On the one hand, gut microbiota can influence the function brain through GBA; on the other hand, the brain can also change the structural composition of gut microbiota via GBA. It contains a myriad of biosignals, such as monoamines, inflammatory cytokines, and macro-biomolecules, as the information carriers. Highly selective, sensitive, and reliable sensing techniques are essential to resolve the specific function of individual biosignals. This review summarizes the widely reported biosignals related to GBA and their functions, and organizes the latest sensing tools to provide feasible characterization ideas for GBA-related work. In addition, these low-cost, fast-responding sensors can also be used for early identification and diagnosis of GBA-related diseases (e.g., depression). Finally, the problems and deficiencies in this field are pointed out to provide a reference for the orientation of researchers in the sensing field.
Collapse
Affiliation(s)
- Linxuan Sun
- Institute of Materials Research, Center of Double Helix, Guangdong Provincial Key Laboratory of Thermal Management Engineering and Materials, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, P. R. China
| | - Yichao Bai
- Institute of Materials Research, Center of Double Helix, Guangdong Provincial Key Laboratory of Thermal Management Engineering and Materials, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, P. R. China
| | - Feiyu Kang
- Institute of Materials Research, Center of Double Helix, Guangdong Provincial Key Laboratory of Thermal Management Engineering and Materials, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, P. R. China
| | - Yu Lei
- Institute of Materials Research, Center of Double Helix, Guangdong Provincial Key Laboratory of Thermal Management Engineering and Materials, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, P. R. China
| |
Collapse
|
6
|
Salaris A, Azevedo RT. Investigating the modulation of gastric sensations and disposition toward food with taVNS. Psychophysiology 2024:e14735. [PMID: 39614658 DOI: 10.1111/psyp.14735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 11/11/2024] [Accepted: 11/12/2024] [Indexed: 12/01/2024]
Abstract
Interoception, the perception of visceral sensations, is key for several survival functions, including those related to feeding behavior. Sensations of hunger and satiety are mediated by gastric signals transmitted via the vagus nerve to the Nucleus of Solitary Tract. Transcutaneous auricular vagus nerve stimulation (taVNS) has been shown to modulate brain-viscera communication and to impact interoceptive processing in the cardiac domain. Yet, its effect on gastric interoception remains unclear. The aim of this study was to investigate taVNS' modulatory effects on gastric interoception using the Water Load Test-II (WLT-II) and its impact on food-related dispositions through a disposition and willingness to eat task (DWET). Participants underwent active or sham taVNS while performing the WLT-II and DWET. Results showed no significant difference in gastric interoceptive accuracy and amount of water ingested between taVNS groups. However, we found a significant reduction in food liking after the fullness phase of the WLT-II in the active (vs sham) taVNS group, suggesting an influence of vagal activation in the inhibition of food enjoyment when satiated. These findings suggest that, while taVNS may not directly enhance gastric interoceptive accuracy at a conscious level, it influences food-related dispositions, likely by modulating the processing of gastric signals. Further research exploring the intricate relationship between vagal modulation, interoceptive abilities and eating behaviors is warranted to elucidate the underlying mechanisms and, possibly, develop targeted interventions for eating disorders.
Collapse
Affiliation(s)
- Andrea Salaris
- Department of Psychology, Sapienza University of Rome, Rome, Italy
- School of Psychology, University of Kent, Canterbury, UK
| | | |
Collapse
|
7
|
Dezfouli MA, Rashidi SK, Yazdanfar N, Khalili H, Goudarzi M, Saadi A, Kiani Deh Kiani A. The emerging roles of neuroactive components produced by gut microbiota. Mol Biol Rep 2024; 52:1. [PMID: 39570444 DOI: 10.1007/s11033-024-10097-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Accepted: 11/06/2024] [Indexed: 11/22/2024]
Abstract
BACKGROUND As a multifunctional ecosystem, the human digestive system contains a complex network of microorganisms, collectively known as gut microbiota. This consortium composed of more than 1013 microorganisms and Firmicutes and Bacteroidetes are the dominant microbes. Gut microbiota is increasingly recognized for its critical role in physiological processes beyond digestion. Gut microbiota participates in a symbiotic relationship with the host and takes advantage of intestinal nutrients and mutually participates in the digestion of complex carbohydrates and maintaining intestinal functions. METHOD AND RESULT We reviewed the neuroactive components produced by gut microbiota. Interestingly, microbiota plays a crucial role in regulating the activity of the intestinal lymphatic system, regulation of the intestinal epithelial barrier, and maintaining the tolerance to food immunostimulating molecules. The gut-brain axis is a two-way communication pathway that links the gut microbiota to the central nervous system (CNS) and importantly is involved in neurodevelopment, cognition, emotion and synaptic transmissions. The connections between gut microbiota and CNS are via endocrine system, immune system and vagus nerve. CONCLUSION The gut microbiota produces common neurotransmitters and neuromodulators of the nervous system. These compounds play a role in neuronal functions, immune system regulation, gastrointestinal homeostasis, permeability of the blood brain barrier and other physiological processes. This review investigates the essential aspects of the neurotransmitters and neuromodulators produced by gut microbiota and their implications in health and disease.
Collapse
Affiliation(s)
- Mitra Ansari Dezfouli
- Department of Neurology, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
- Student Research Committee, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| | - Seyed Khalil Rashidi
- Department of Medical Biotechnology, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Nada Yazdanfar
- Department of Neurology, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Hamidreza Khalili
- Department of Pharmacology, School of Pharmacy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mehdi Goudarzi
- Medicinal Plant Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Ali Saadi
- Department of Neurology, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
- Student Research Committee, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Ali Kiani Deh Kiani
- Department of Neurology, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
- Student Research Committee, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
8
|
Liu FJ, Wu J, Gong LJ, Yang HS, Chen H. Non-invasive vagus nerve stimulation in anti-inflammatory therapy: mechanistic insights and future perspectives. Front Neurosci 2024; 18:1490300. [PMID: 39605787 PMCID: PMC11599236 DOI: 10.3389/fnins.2024.1490300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Accepted: 10/24/2024] [Indexed: 11/29/2024] Open
Abstract
Non-invasive vagus nerve stimulation (VNS) represents a transformative approach for managing a broad spectrum of inflammatory and autoimmune conditions, including rheumatoid arthritis and inflammatory bowel disease. This comprehensive review delineates the mechanisms underlying VNS, emphasizing the cholinergic anti-inflammatory pathway, and explores interactions within the neuro-immune and vagus-gut axes based on both clinical outcomes and pre-clinical models. Clinical applications have confirmed the efficacy of VNS in managing specific autoimmune diseases, such as rheumatoid arthritis, and chronic inflammatory conditions like inflammatory bowel disease, showcasing the variability in stimulation parameters and patient responses. Concurrently, pre-clinical studies have provided insights into the potential of VNS in modulating cardiovascular and broader inflammatory responses, paving the way for its translational application in clinical settings. Innovations in non-invasive VNS technology and precision neuromodulation are enhancing its therapeutic potential, making it a viable option for patients who are unresponsive to conventional treatments. Nonetheless, the widespread adoption of this promising therapy is impeded by regulatory challenges, patient compliance issues, and the need for extensive studies on long-term efficacy and safety. Future research directions will focus on refining VNS technology, optimizing treatment parameters, and exploring synergistic effects with other therapeutic modalities, which could revolutionize the management of chronic inflammatory and autoimmune disorders.
Collapse
Affiliation(s)
- Fu-Jun Liu
- Neurology Medical Center II, Foresea Life Insurance Guangzhou General Hospital, Guangzhou, China
| | - Jing Wu
- Department of Medical Imaging, Foresea Life Insurance Guangzhou General Hospital, Guangzhou, China
| | - Li-Jun Gong
- Center of Surgical Anesthesia, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Hong-Shuai Yang
- Central Operating Room, Foresea Life Insurance Guangzhou General Hospital, Guangzhou, China
| | - Huan Chen
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| |
Collapse
|
9
|
Tavares-Silva E, de Aquino Lemos V, de França E, Silvestre J, dos Santos SA, Ravacci GR, Thomatieli-Santos RV. Protective Effects of Probiotics on Runners' Mood: Immunometabolic Mechanisms Post-Exercise. Nutrients 2024; 16:3761. [PMID: 39519594 PMCID: PMC11547471 DOI: 10.3390/nu16213761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Revised: 10/24/2024] [Accepted: 10/29/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND The gut-brain axis may mediate mood changes due to strenuous exercise. Therefore, probiotic supplementation may mitigate mood worsening. PURPOSE The present study aims to evaluate the effect of probiotic supplementation on mood and immunometabolic parameters after a marathon. MATERIALS AND METHODS Fourteen marathon runners were selected and divided into placebo and probiotic groups that were supplemented for 30 days. Before and after the marathon, mood (POMS) was assessed, and blood was collected for analysis of immunometabolic parameters. Statistical analysis was performed, and p < 0.05 was considered to determine statistically differences. RESULTS Tension decreased after the marathon in both groups. Vigor decreased only in the placebo group. Fatigue increased after the marathon in both groups. TMD increased after the marathon in placebo. The IL2/IL-4 ratio decreased in the probiotic group after the marathon compared to before and increased compared to the placebo group. The IL-10 increased after the marathon in placebo. TNF-α increased after the marathon in probiotics. The TNF-α/IL-10 ratio decreased after the marathon in both groups. LPS decreased in the probiotic group after the marathon compared to before and in the placebo group. CONCLUSIONS Thirty days of probiotic supplementation attenuated the impact of marathons on mood worsening. The decrease in LPS in the probiotic group mediated the change in the pro/anti-inflammatory balance, indicating an immunometabolic mechanism by which the gut-brain axis impacts mood after strenuous exercise.
Collapse
Affiliation(s)
- Edgar Tavares-Silva
- Post-Graduate Program in Psychobiology, Federal University of São Paulo, São Paulo 04040-003, SP, Brazil (V.d.A.L.)
| | - Valdir de Aquino Lemos
- Post-Graduate Program in Psychobiology, Federal University of São Paulo, São Paulo 04040-003, SP, Brazil (V.d.A.L.)
| | - Elias de França
- Department of Bioscience, Federal University of São Paulo, Rua Silva Jardim, 136, Vila Mathias, Santos 11015-021, SP, Brazil; (E.d.F.); (J.S.); (S.A.d.S.); (G.R.R.)
| | - Jean Silvestre
- Department of Bioscience, Federal University of São Paulo, Rua Silva Jardim, 136, Vila Mathias, Santos 11015-021, SP, Brazil; (E.d.F.); (J.S.); (S.A.d.S.); (G.R.R.)
| | - Samile Amorim dos Santos
- Department of Bioscience, Federal University of São Paulo, Rua Silva Jardim, 136, Vila Mathias, Santos 11015-021, SP, Brazil; (E.d.F.); (J.S.); (S.A.d.S.); (G.R.R.)
| | - Graziela Rosa Ravacci
- Department of Bioscience, Federal University of São Paulo, Rua Silva Jardim, 136, Vila Mathias, Santos 11015-021, SP, Brazil; (E.d.F.); (J.S.); (S.A.d.S.); (G.R.R.)
| | - Ronaldo Vagner Thomatieli-Santos
- Post-Graduate Program in Psychobiology, Federal University of São Paulo, São Paulo 04040-003, SP, Brazil (V.d.A.L.)
- Department of Bioscience, Federal University of São Paulo, Rua Silva Jardim, 136, Vila Mathias, Santos 11015-021, SP, Brazil; (E.d.F.); (J.S.); (S.A.d.S.); (G.R.R.)
| |
Collapse
|
10
|
Geloso MC, Zupo L, Corvino V. Crosstalk between peripheral inflammation and brain: Focus on the responses of microglia and astrocytes to peripheral challenge. Neurochem Int 2024; 180:105872. [PMID: 39362496 DOI: 10.1016/j.neuint.2024.105872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 09/18/2024] [Accepted: 09/30/2024] [Indexed: 10/05/2024]
Abstract
A growing body of evidence supports the link between peripheral inflammation and impairment of neurologic functions, including mood and cognitive abilities. The pathogenic event connecting peripheral inflammation and brain dysfunction is represented by neuroinflammation, a pathogenic phenomenon that provides an important contribution to neurodegeneration and cognitive decline also in Alzheimer's, Parkinson's, Huntington's diseases, as well as in Multiple Sclerosis. It is driven by resident brain immune cells, microglia and astrocytes, that acquire an activated phenotype in response to proinflammatory molecules moving from the periphery to the brain parenchyma. Although a huge progress has been made in clarifying cellular and molecular mechanisms bridging peripheral and central inflammation, a clear picture has not been achieved so far. Therefore, experimental models are of crucial relevance to clarify knowledge gaps in this regard. Many findings demonstrate that systemic inflammation induced by pathogen-associated molecular patterns, such as lipopolysaccharide (LPS), is able to trigger neuroinflammation. Therefore, LPS-administration is widely considered a useful tool to study this phenomenon. On this basis, the present review will focus on in vivo studies based on acute and subacute effects of systemic administration of LPS, with special attention on the state of art of microglia and astrocyte response to peripheral challenge.
Collapse
Affiliation(s)
- Maria Concetta Geloso
- Department of Neuroscience, Section of Human Anatomy, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168, Rome, Italy; Gemelli Science and Technology Park (GSTeP)-Organoids Research Core Facility, Fondazione Policlinico Agostino Gemelli IRCCS, Rome, Italy.
| | - Luca Zupo
- Department of Neuroscience, Section of Human Anatomy, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168, Rome, Italy
| | - Valentina Corvino
- Department of Neuroscience, Section of Human Anatomy, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168, Rome, Italy
| |
Collapse
|
11
|
Clarke GS, Page AJ, Eldeghaidy S. The gut-brain axis in appetite, satiety, food intake, and eating behavior: Insights from animal models and human studies. Pharmacol Res Perspect 2024; 12:e70027. [PMID: 39417406 PMCID: PMC11483575 DOI: 10.1002/prp2.70027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 09/02/2024] [Accepted: 09/24/2024] [Indexed: 10/19/2024] Open
Abstract
The gut-brain axis plays a pivotal role in the finely tuned orchestration of food intake, where both homeostatic and hedonic processes collaboratively control our dietary decisions. This interplay involves the transmission of mechanical and chemical signals from the gastrointestinal tract to the appetite centers in the brain, conveying information on meal arrival, quantity, and chemical composition. These signals are processed in the brain eventually leading to the sensation of satiety and the termination of a meal. However, the regulation of food intake and appetite extends beyond the realms of pure physiological need. Hedonic mechanisms, including sensory perception (i.e., through sight, smell, and taste), habitual behaviors, and psychological factors, exert profound influences on food intake. Drawing from studies in animal models and human research, this comprehensive review summarizes the physiological mechanisms that underlie the gut-brain axis and its interplay with the reward network in the regulation of appetite and satiety. The recent advancements in neuroimaging techniques, with a focus on human studies that enable investigation of the neural mechanisms underpinning appetite regulation are discussed. Furthermore, this review explores therapeutic/pharmacological strategies that hold the potential for controlling food intake.
Collapse
Affiliation(s)
- Georgia S. Clarke
- School of BiomedicineThe University of AdelaideAdelaideSouth AustraliaAustralia
- Robinson Research InstituteThe University of AdelaideAdelaideSouth AustraliaAustralia
- Nutrition, Diabetes and Gut Health, Lifelong Health ThemeSouth Australian Health and Medical Research Institute, SAHMRIAdelaideSouth AustraliaAustralia
| | - Amanda J. Page
- School of BiomedicineThe University of AdelaideAdelaideSouth AustraliaAustralia
- Nutrition, Diabetes and Gut Health, Lifelong Health ThemeSouth Australian Health and Medical Research Institute, SAHMRIAdelaideSouth AustraliaAustralia
| | - Sally Eldeghaidy
- Division of Food, Nutrition and DieteticsSchool of Biosciences, University of NottinghamNottinghamUK
- Sir Peter Mansfield Imaging CentreSchool of Physics and Astronomy, University of NottinghamNottinghamUK
| |
Collapse
|
12
|
Li S, Zhao L, Xiao J, Guo Y, Fu R, Zhang Y, Xu S. The gut microbiome: an important role in neurodegenerative diseases and their therapeutic advances. Mol Cell Biochem 2024; 479:2217-2243. [PMID: 37787835 DOI: 10.1007/s11010-023-04853-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 09/06/2023] [Indexed: 10/04/2023]
Abstract
There are complex interactions between the gut and the brain. With increasing research on the relationship between gut microbiota and brain function, accumulated clinical and preclinical evidence suggests that gut microbiota is intimately involved in the pathogenesis of neurodegenerative diseases (NDs). Increasingly studies are beginning to focus on the association between gut microbiota and central nervous system (CNS) degenerative pathologies to find potential therapies for these refractory diseases. In this review, we summarize the changes in the gut microbiota in Alzheimer's disease, Parkinson's disease, multiple sclerosis, and amyotrophic lateral sclerosis and contribute to our understanding of the function of the gut microbiota in NDs and its possible involvement in the pathogenesis. We subsequently discuss therapeutic approaches targeting gut microbial abnormalities in these diseases, including antibiotics, diet, probiotics, and fecal microbiota transplantation (FMT). Furthermore, we summarize some completed and ongoing clinical trials of interventions with gut microbes for NDs, which may provide new ideas for studying NDs.
Collapse
Affiliation(s)
- Songlin Li
- Medical Experiment Center, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Linna Zhao
- Medical Experiment Center, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Tianjin Key Laboratory of Translational Research of TCM Prescription and Syndrome, Tianjin, China
| | - Jie Xiao
- Medical Experiment Center, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yuying Guo
- Medical Experiment Center, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Tianjin Key Laboratory of Translational Research of TCM Prescription and Syndrome, Tianjin, China
| | - Rong Fu
- Medical Experiment Center, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yunsha Zhang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Shixin Xu
- Medical Experiment Center, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China.
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China.
- Tianjin Key Laboratory of Translational Research of TCM Prescription and Syndrome, Tianjin, China.
| |
Collapse
|
13
|
Verma A, Inslicht SS, Bhargava A. Gut-Brain Axis: Role of Microbiome, Metabolomics, Hormones, and Stress in Mental Health Disorders. Cells 2024; 13:1436. [PMID: 39273008 PMCID: PMC11394554 DOI: 10.3390/cells13171436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 08/19/2024] [Accepted: 08/23/2024] [Indexed: 09/15/2024] Open
Abstract
The influence of gut microbiome, metabolites, omics, hormones, and stress on general and mental health is increasingly being recognized. Ancient cultures recognized the importance of diet and gut health on the overall health of an individual. Western science and modern scientific methods are beginning to unravel the foundations and mechanisms behind some of the ancient beliefs and customs. The gut microbiome, an organ itself, is now thought to influence almost all other organs, ranging from the brain to the reproductive systems. Gut microbiome, metabolites, hormones, and biological sex also influence a myriad of health conditions that range from mental health disorders, obesity, gastrointestinal disorders, and cardiovascular diseases to reproductive health. Here, we review the history and current understanding of the gut-brain axis bidirectional talk in various mental health disorders with special emphasis on anxiety and depressive disorders, whose prevalence has increased by over 50% in the past three decades with COVID-19 pandemic being the biggest risk factor in the last few years. The vagal nerve is an important contributor to this bidirectional talk, but other pathways also contribute, and most remain understudied. Probiotics containing Lactobacillus and Bifidobacterium species seem to have the most impact on improvement in mental health symptoms, but the challenge appears to be maintaining sustained levels, especially since neither Lactobacillus nor Bifidobacterium can permanently colonize the gut. Ancient endogenous retroviral DNA in the human genome is also linked to several psychiatric disorders, including depression. These discoveries reveal the complex and intricately intertwined nature of gut health with mental health disorders.
Collapse
Affiliation(s)
- Ankita Verma
- Department of Obstetrics, Gynecology & Reproductive Sciences, University of California at San Francisco, 513 Parnassus Avenue, San Francisco, CA 94143, USA
| | - Sabra S Inslicht
- San Francisco VA Health Care System, San Francisco, CA 94121, USA
- Department of Psychiatry and Behavioral Sciences, University of California at San Francisco, San Francisco, CA 94143, USA
| | - Aditi Bhargava
- Department of Obstetrics, Gynecology & Reproductive Sciences, University of California at San Francisco, 513 Parnassus Avenue, San Francisco, CA 94143, USA
| |
Collapse
|
14
|
Kerschner A, Hassan H, Kern M, Edeani F, Mei L, Sanvanson P, Shaker R, Yu E. Parkinson's disease is associated with low striated esophagus contractility potentially contributing to the development of dysphagia. Neurogastroenterol Motil 2024; 36:e14822. [PMID: 38798058 PMCID: PMC11246227 DOI: 10.1111/nmo.14822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 05/03/2024] [Accepted: 05/09/2024] [Indexed: 05/29/2024]
Abstract
BACKGROUND Parkinson's disease (PD) is the second most common neurodegenerative disorder, and more than 80% of PD patients will develop oropharyngeal dysphagia. Despite its striated histology, proximity to airway, and potential negative impact of its dysfunction on bolus transport and airway safety, the contractile function of the striated esophagus in PD patients has not been systematically studied. METHODS Using our repository of clinical manometry and the Milwaukee ManoBank, we analyzed high-resolution manometry (HRM) studies of 20 PD patients, mean age 69.1 (range 38-87 years); 30 non-PD patients with dysphagia, mean age 64.0 (44-86 years); and 32 healthy volunteers, mean age 65.3 (39-86 years). Patients with abnormal findings based on Chicago Classification 4.0 were identified. Repeat analysis was performed in 20% of the manometric tracings by a different investigator with inter-rater concordance between 0.91 and 0.99. KEY RESULTS The striated esophageal contractile integral in PD patients was significantly lower than that in non-PD dysphagic patients and healthy controls (p = 0.03 and <0.01, respectively). This significant difference persisted after excluding patients with concurrent Chicago Classification motility disorders (p = 0.02 and 0.01, respectively). In both analyses, the distal esophageal contractile integral did not show any significant difference between groups (p = 0.58 and 0.93, respectively). CONCLUSIONS & INFERENCES PD is associated with a significant decrease in striated esophagus contractility compared to non-PD and healthy controls. This finding may play a pathophysiologic role in development of dysphagia in this patient population.
Collapse
Affiliation(s)
- Alexander Kerschner
- Department of Internal Medicine, The Hub for Collaborative Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Hamza Hassan
- Division of Gastroenterology and Hepatology, The Hub for Collaborative Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Mark Kern
- Division of Gastroenterology and Hepatology, The Hub for Collaborative Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Francis Edeani
- Division of Gastroenterology and Hepatology, The Hub for Collaborative Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Ling Mei
- Division of Gastroenterology and Hepatology, The Hub for Collaborative Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Patrick Sanvanson
- Division of Gastroenterology and Hepatology, The Hub for Collaborative Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Reza Shaker
- Division of Gastroenterology and Hepatology, The Hub for Collaborative Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Elliot Yu
- Division of Gastroenterology and Hepatology, The Hub for Collaborative Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| |
Collapse
|
15
|
Wang L, Taché Y. The parasympathetic and sensory innervation of the proximal and distal colon in male mice. Front Neuroanat 2024; 18:1422403. [PMID: 39045348 PMCID: PMC11263295 DOI: 10.3389/fnana.2024.1422403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Accepted: 06/24/2024] [Indexed: 07/25/2024] Open
Abstract
Introduction The distributions of extrinsic neurons innervating the colon show differences in experimental animals from humans, including the vagal and spinal parasympathetic innervation to the distal colon. The neuroanatomical tracing to the mouse proximal colon has not been studied in details. This study aimed to trace the locations of extrinsic neurons projecting to the mouse proximal colon compared to the distal colon using dual retrograde tracing. Methods The parasympathetic and sensory neurons projecting to colon were assessed using Cholera Toxin subunit B conjugated to Alexa-Fluor 488 or 555 injected in the proximal and distal colon of the same mice. Results Retrograde tracing from the proximal and distal colon labeled neurons in the dorsal motor nucleus of the vagus (DMV) and the nodose ganglia, while the tracing from the distal colon did not label the parasympathetic neurons in the lumbosacral spinal cord at L6-S1. Neurons in the pelvic ganglia which were cholinergic projected to the distal colon. There were more neurons in the DMV and nodose ganglia projecting to the proximal than distal colon. The right nodose ganglion had a higher number of neurons than the left ganglion innervating the proximal colon. In the dorsal root ganglia (DRG), the highest number of neurons traced from the distal colon were at L6, and those from the proximal colon at T12. DRG neurons projected closely to the cholinergic neurons in the intermediolateral column of L6 spinal cord. Small percentages of neurons with dual projections to both the proximal and distal colon existed in the DMV, nodose ganglia and DRG. We also observed long projecting neurons traced from the caudal distal colon to the transverse and proximal colon, some of which were calbindin immunoreactive, while there were no retrogradely labeled neurons traced from the proximal to distal colon. Discussion These data demonstrated that the vagal motor and motor and sensory neurons innervate both the proximal and distal colon in mice, and the autonomic neurons in the intermediate zone of the lumbosacral spinal cord do not project directly to the mouse colon, which differs from that in humans.
Collapse
Affiliation(s)
- Lixin Wang
- CURE/Digestive Diseases Research Center, Department of Medicine, Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine, UCLA, Los Angeles, CA, United States
- Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, CA, United States
| | - Yvette Taché
- CURE/Digestive Diseases Research Center, Department of Medicine, Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine, UCLA, Los Angeles, CA, United States
- Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, CA, United States
| |
Collapse
|
16
|
Xu H, Luo Y, Li Q, Zhu H. Acupuncture influences multiple diseases by regulating gut microbiota. Front Cell Infect Microbiol 2024; 14:1371543. [PMID: 39040602 PMCID: PMC11260648 DOI: 10.3389/fcimb.2024.1371543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 06/13/2024] [Indexed: 07/24/2024] Open
Abstract
Acupuncture, an important green and side effect-free therapy in traditional Chinese medicine, is widely use both domestically and internationally. Acupuncture can interact with the gut microbiota and influence various diseases, including metabolic diseases, gastrointestinal diseases, mental disorders, nervous system diseases, and other diseases. This review presents a thorough analysis of these interactions and their impacts and examines the alterations in the gut microbiota and the potential clinical outcomes following acupuncture intervention to establish a basis for the future utilization of acupuncture in clinical treatments.
Collapse
Affiliation(s)
- Huimin Xu
- Department of Abdominal Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Yingzhe Luo
- Department of Oncology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Qiaoqi Li
- Department of Abdominal Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Hong Zhu
- Department of Abdominal Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
17
|
Al Shamsi HSS, Rainey-Smith SR, Gardener SL, Sohrabi HR, Canovas R, Martins RN, Fernando WMADB. The Relationship between Diet, Depression, and Alzheimer's Disease: A Narrative Review. Mol Nutr Food Res 2024; 68:e2300419. [PMID: 38973221 DOI: 10.1002/mnfr.202300419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Revised: 02/02/2024] [Indexed: 07/09/2024]
Abstract
PURPOSE OF REVIEW This narrative review evaluates the role of diet in the relationship between depression and Alzheimer's disease (AD). RECENT FINDINGS AD and depression are often comorbid, and depression appears to independently increase the future risk of AD. Evidence suggests diet influences the risk of both conditions directly and indirectly. Diet impacts neurochemical and biological processes that may affect the development and progression of depression and cognitive dysfunction. The dietary components offering the greatest protection against depression and AD are yet to be determined. Current evidence highlights the importance of polyphenolic compounds, folate, B vitamins, and polyunsaturated fatty acids, along with adherence to dietary patterns like the Mediterranean diet, which includes multiple beneficial dietary factors. SUMMARY The investigation of dietary factors in the prevention of depression and AD is a comparatively young field of research. Comprehensive highly characterised longitudinal datasets and advanced analytical approaches are required to further examine the complex relationship between diet, depression, and AD. There is a critical need for more research in this area to develop effective preventive strategies aimed at maintaining mental and physical health with advancing age.
Collapse
Affiliation(s)
- Hilal Salim Said Al Shamsi
- Centre of Excellence for Alzheimer's Disease Research and Care, School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, 6027, Australia
| | - Stephanie R Rainey-Smith
- Centre of Excellence for Alzheimer's Disease Research and Care, School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, 6027, Australia
- Centre for Healthy Ageing, Health Futures Institute, Murdoch University, Murdoch, Western Australia, 6150, Australia
- Alzheimer's Research Australia, Ralph and Patricia Sarich Neuroscience Research Institute, Nedlands, Western Australia, 6009, Australia
- Lifestyle Approaches Towards Cognitive Health Research Group, Murdoch University, Murdoch, Western Australia, 6150, Australia
- School of Psychological Science, University of Western Australia, Crawley, Western Australia, 6009, Australia
| | - Samantha L Gardener
- Centre of Excellence for Alzheimer's Disease Research and Care, School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, 6027, Australia
- Alzheimer's Research Australia, Ralph and Patricia Sarich Neuroscience Research Institute, Nedlands, Western Australia, 6009, Australia
- Lifestyle Approaches Towards Cognitive Health Research Group, Murdoch University, Murdoch, Western Australia, 6150, Australia
| | - Hamid R Sohrabi
- Centre of Excellence for Alzheimer's Disease Research and Care, School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, 6027, Australia
- Centre for Healthy Ageing, Health Futures Institute, Murdoch University, Murdoch, Western Australia, 6150, Australia
- Alzheimer's Research Australia, Ralph and Patricia Sarich Neuroscience Research Institute, Nedlands, Western Australia, 6009, Australia
- Department of Biomedical Sciences, Macquarie University, Macquarie Park, New South Wales, 2109, Australia
| | - Rodrigo Canovas
- Health & Biosecurity, The Commonwealth Scientific and Industrial Research Organisation, Herston, Queensland, 4029, Australia
| | - Ralph N Martins
- Centre of Excellence for Alzheimer's Disease Research and Care, School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, 6027, Australia
- Centre for Healthy Ageing, Health Futures Institute, Murdoch University, Murdoch, Western Australia, 6150, Australia
- Alzheimer's Research Australia, Ralph and Patricia Sarich Neuroscience Research Institute, Nedlands, Western Australia, 6009, Australia
- Department of Biomedical Sciences, Macquarie University, Macquarie Park, New South Wales, 2109, Australia
| | - Warnakulasuriya Mary Ann Dipika Binosha Fernando
- Centre of Excellence for Alzheimer's Disease Research and Care, School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, 6027, Australia
- Alzheimer's Research Australia, Ralph and Patricia Sarich Neuroscience Research Institute, Nedlands, Western Australia, 6009, Australia
| |
Collapse
|
18
|
Lee B, Lee SM, Song JW, Choi JW. Gut Microbiota Metabolite Messengers in Brain Function and Pathology at a View of Cell Type-Based Receptor and Enzyme Reaction. Biomol Ther (Seoul) 2024; 32:403-423. [PMID: 38898687 PMCID: PMC11214962 DOI: 10.4062/biomolther.2024.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 05/02/2024] [Accepted: 05/25/2024] [Indexed: 06/21/2024] Open
Abstract
The human gastrointestinal (GI) tract houses a diverse microbial community, known as the gut microbiome comprising bacteria, viruses, fungi, and protozoa. The gut microbiome plays a crucial role in maintaining the body's equilibrium and has recently been discovered to influence the functioning of the central nervous system (CNS). The communication between the nervous system and the GI tract occurs through a two-way network called the gut-brain axis. The nervous system and the GI tract can modulate each other through activated neuronal cells, the immune system, and metabolites produced by the gut microbiome. Extensive research both in preclinical and clinical realms, has highlighted the complex relationship between the gut and diseases associated with the CNS, such as Alzheimer's disease, Parkinson's disease, and amyotrophic lateral sclerosis. This review aims to delineate receptor and target enzymes linked with gut microbiota metabolites and explore their specific roles within the brain, particularly their impact on CNS-related diseases.
Collapse
Affiliation(s)
- Bada Lee
- Department of Biomedicinal and Pharmaceutical Sciences, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Soo Min Lee
- Department of Biomedicinal and Pharmaceutical Sciences, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Jae Won Song
- Department of Regulatory Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Jin Woo Choi
- Department of Biomedicinal and Pharmaceutical Sciences, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
- Department of Regulatory Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| |
Collapse
|
19
|
Varley AN, Browning KN. Gastrointestinal dysfunction in the valproic acid induced model of social deficit in rats. Auton Neurosci 2024; 253:103161. [PMID: 38461695 PMCID: PMC11128350 DOI: 10.1016/j.autneu.2024.103161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 02/28/2024] [Accepted: 02/28/2024] [Indexed: 03/12/2024]
Abstract
Autism spectrum disorder (ASD) has increased in incidence over the past several decades, and is associated with a range of co-morbidities including gastrointestinal (GI) dysfunctions including gastroesophageal reflux, abdominal pain, bloating, constipation and/or diarrhea. Several animal models have been used that replicate several aspects of ASD but no single model has been able to replicate the entire disease pathophysiology. In humans, prenatal exposure to valproic acid (VPA) has been identified as a significant risk factor and rodent models have shown that in utero VPA exposure leads to behavioral deficits in offspring. The present study aimed to investigate whether in utero exposure to VPA induces GI dysfunction in rats. Timed pregnant Sprague-Dawley rats were injected with a single dose of VPA at embryonic day 12.5. Both male and female offspring subsequently underwent behavioral studies and assessment of GI function in adulthood. In utero VPA treatment induced social deficits in both male and female offspring, decreasing sociability and social novelty. Histological examination showed that VPA treated offspring had decreased thickness of GI muscle and mucosa, while immunohistochemical studies showed a decrease in myenteric neuron number in the fundus. Functional studies showed that both male and female VPA offspring had a delay in gastric emptying compared to vehicle treated offspring. Results of the current study suggest that the rat VPA model of behavioral deficits may be a convenient model by which both mechanistic and functional insights into GI dysfunction may be studied.
Collapse
Affiliation(s)
- Ashley N Varley
- Department of Comparative Medicine, Penn State College of Medicine, Hershey, PA, United States of America
| | - Kirsteen N Browning
- Department of Neural and Behavioral Sciences, Penn State College of Medicine, Hershey, PA, United States of America.
| |
Collapse
|
20
|
McKay DM, Defaye M, Rajeev S, MacNaughton WK, Nasser Y, Sharkey KA. Neuroimmunophysiology of the gastrointestinal tract. Am J Physiol Gastrointest Liver Physiol 2024; 326:G712-G725. [PMID: 38626403 PMCID: PMC11376980 DOI: 10.1152/ajpgi.00075.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 04/11/2024] [Accepted: 04/15/2024] [Indexed: 04/18/2024]
Abstract
Gut physiology is the epicenter of a web of internal communication systems (i.e., neural, immune, hormonal) mediated by cell-cell contacts, soluble factors, and external influences, such as the microbiome, diet, and the physical environment. Together these provide the signals that shape enteric homeostasis and, when they go awry, lead to disease. Faced with the seemingly paradoxical tasks of nutrient uptake (digestion) and retarding pathogen invasion (host defense), the gut integrates interactions between a variety of cells and signaling molecules to keep the host nourished and protected from pathogens. When the system fails, the outcome can be acute or chronic disease, often labeled as "idiopathic" in nature (e.g., irritable bowel syndrome, inflammatory bowel disease). Here we underscore the importance of a holistic approach to gut physiology, placing an emphasis on intercellular connectedness, using enteric neuroimmunophysiology as the paradigm. The goal of this opinion piece is to acknowledge the pace of change brought to our field via single-cell and -omic methodologies and other techniques such as cell lineage tracing, transgenic animal models, methods for culturing patient tissue, and advanced imaging. We identify gaps in the field and hope to inspire and challenge colleagues to take up the mantle and advance awareness of the subtleties, intricacies, and nuances of intestinal physiology in health and disease by defining communication pathways between gut resident cells, those recruited from the circulation, and "external" influences such as the central nervous system and the gut microbiota.
Collapse
Affiliation(s)
- Derek M McKay
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
- Gastrointestinal Research Group, University of Calgary, Calgary, Alberta, Canada
- Inflammation Research Network, University of Calgary, Calgary, Alberta, Canada
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Manon Defaye
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
- Gastrointestinal Research Group, University of Calgary, Calgary, Alberta, Canada
- Inflammation Research Network, University of Calgary, Calgary, Alberta, Canada
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Sruthi Rajeev
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
- Gastrointestinal Research Group, University of Calgary, Calgary, Alberta, Canada
- Inflammation Research Network, University of Calgary, Calgary, Alberta, Canada
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Wallace K MacNaughton
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
- Gastrointestinal Research Group, University of Calgary, Calgary, Alberta, Canada
- Inflammation Research Network, University of Calgary, Calgary, Alberta, Canada
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
| | - Yasmin Nasser
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
- Gastrointestinal Research Group, University of Calgary, Calgary, Alberta, Canada
- Inflammation Research Network, University of Calgary, Calgary, Alberta, Canada
- Department of Medicine, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Keith A Sharkey
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
- Gastrointestinal Research Group, University of Calgary, Calgary, Alberta, Canada
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
21
|
Wang X, Wen X, Yuan S, Zhang J. Gut-brain axis in the pathogenesis of sepsis-associated encephalopathy. Neurobiol Dis 2024; 195:106499. [PMID: 38588753 DOI: 10.1016/j.nbd.2024.106499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 03/31/2024] [Accepted: 04/04/2024] [Indexed: 04/10/2024] Open
Abstract
The gut-brain axis is a bidirectional communication network linking the gut and the brain, overseeing digestive functions, emotional responses, body immunity, brain development, and overall health. Substantial research highlights a connection between disruptions of the gut-brain axis and various psychiatric and neurological conditions, including depression and Alzheimer's disease. Given the impact of the gut-brain axis on behavior, cognition, and brain diseases, some studies have started to pay attention to the role of the axis in sepsis-associated encephalopathy (SAE), where cognitive impairment is the primary manifestation. SAE emerges as the primary and earliest form of organ dysfunction following sepsis, potentially leading to acute cognitive impairment and long-term cognitive decline in patients. Notably, the neuronal damage in SAE does not stem directly from the central nervous system (CNS) infection but rather from an infection occurring outside the brain. The gut-brain axis is posited as a pivotal factor in this process. This review will delve into the gut-brain axis, exploring four crucial pathways through which inflammatory signals are transmitted and elevate the incidence of SAE. These pathways encompass the vagus nerve pathway, the neuroendocrine pathway involving the hypothalamic-pituitary-adrenal (HPA) axis and serotonin (5-HT) regulation, the neuroimmune pathway, and the microbial regulation. These pathways can operate independently or collaboratively on the CNS to modulate brain activity. Understanding how the gut affects and regulates the CNS could offer the potential to identify novel targets for preventing and treating this condition, ultimately enhancing the prognosis for individuals with SAE.
Collapse
Affiliation(s)
- Xin Wang
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, PR China; Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, PR China
| | - Xiaoyue Wen
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, PR China; Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, PR China
| | - Shiying Yuan
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, PR China; Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, PR China.
| | - Jiancheng Zhang
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, PR China; Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, PR China.
| |
Collapse
|
22
|
Boyajian JL, Islam P, Abosalha A, Schaly S, Thareja R, Kassab A, Arora K, Santos M, Shum-Tim C, Prakash S. Probiotics, prebiotics, synbiotics and other microbiome-based innovative therapeutics to mitigate obesity and enhance longevity via the gut-brain axis. MICROBIOME RESEARCH REPORTS 2024; 3:29. [PMID: 39421246 PMCID: PMC11480732 DOI: 10.20517/mrr.2024.05] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 04/18/2024] [Accepted: 05/11/2024] [Indexed: 10/19/2024]
Abstract
The global prevalence of obesity currently exceeds 1 billion people and is accompanied by an increase in the aging population. Obesity and aging share many hallmarks and are leading risk factors for cardiometabolic disease and premature death. Current anti-obesity and pro-longevity pharmacotherapies are limited by side effects, warranting the development of novel therapies. The gut microbiota plays a major role in human health and disease, with a dysbiotic composition evident in obese and aged individuals. The bidirectional communication system between the gut and the central nervous system, known as the gut-brain axis, may link obesity to unhealthy aging. Modulating the gut with microbiome-targeted therapies, such as biotics, is a novel strategy to treat and/or manage obesity and promote longevity. Biotics represent material derived from living or once-living organisms, many of which have therapeutic effects. Pre-, pro-, syn- and post-biotics may beneficially modulate gut microbial composition and function to improve obesity and the aging process. However, the investigation of biotics as next-generation therapeutics has only just begun. Further research is needed to identify therapeutic biotics and understand their mechanisms of action. Investigating the function of the gut-brain axis in obesity and aging may lead to novel therapeutic strategies for obese, aged and comorbid (e.g., sarcopenic obese) patient populations. This review discusses the interrelationship between obesity and aging, with a particular emphasis on the gut microbiome, and presents biotics as novel therapeutic agents for obesity, aging and related disease states.
Collapse
Affiliation(s)
- Jacqueline L. Boyajian
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering, Faculty of Medicine and Health Sciences, McGill University, Montreal H3A 2B4, Quebec, Canada
| | - Paromita Islam
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering, Faculty of Medicine and Health Sciences, McGill University, Montreal H3A 2B4, Quebec, Canada
| | - Ahmed Abosalha
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering, Faculty of Medicine and Health Sciences, McGill University, Montreal H3A 2B4, Quebec, Canada
- Pharmaceutical Technology Department, Faculty of Pharmacy, Tanta University, Tanta 31111, Egypt
| | - Sabrina Schaly
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering, Faculty of Medicine and Health Sciences, McGill University, Montreal H3A 2B4, Quebec, Canada
| | - Rahul Thareja
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering, Faculty of Medicine and Health Sciences, McGill University, Montreal H3A 2B4, Quebec, Canada
| | - Amal Kassab
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering, Faculty of Medicine and Health Sciences, McGill University, Montreal H3A 2B4, Quebec, Canada
| | - Karan Arora
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering, Faculty of Medicine and Health Sciences, McGill University, Montreal H3A 2B4, Quebec, Canada
| | - Madison Santos
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering, Faculty of Medicine and Health Sciences, McGill University, Montreal H3A 2B4, Quebec, Canada
| | - Cedrique Shum-Tim
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering, Faculty of Medicine and Health Sciences, McGill University, Montreal H3A 2B4, Quebec, Canada
| | - Satya Prakash
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering, Faculty of Medicine and Health Sciences, McGill University, Montreal H3A 2B4, Quebec, Canada
| |
Collapse
|
23
|
Sahu PK, Ranjan P, Agrawal D, Singh G. Effect of Transcutaneous Electrical Nerve Stimulation of T6 Dermatome on Appetite and Weight Reduction in Obese Individuals: A Pilot Study. Neurol India 2024; 72:534-539. [PMID: 39041969 DOI: 10.4103/neuroindia.ni_1063_20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Accepted: 04/27/2022] [Indexed: 07/24/2024]
Abstract
BACKGROUND Previous work demonstrated the beneficial effect of T6 dermatomal stimulation by the percutaneous method in managing obesity. However, a simple, cost-effective, and feasible intervention stimulating the dermatome can be a potential solution for obesity care. OBJECTIVE The objective of this study was to find the effect of transcutaneous electrical nerve stimulation (TENS) of T6 dermatome on appetite and weight loss in obese individuals. MATERIAL AND METHODS In this prospective single-arm experimental study, 20 obese patients received TENS treatment of T6 dermatome for 30 min, once a week and for 12 weeks. Outcome measures such as appetite level in the visual analog scale (VAS), weight in kg, and basal metabolic index (BMI) in kg/m2 were assessed at baseline, 12 weeks -post-intervention, and at 8 weeks of follow-up. RESULTS A statistically significant difference in all the variables was found from baseline to 12 weeks of intervention (P < 0.001) and maintained till 8 weeks of follow-up. From pre- to post-treatment, the effect size for appetite reduction was large (>1) while for weight and BMI loss was (0.14, 0.16), respectively small. Additionally, a weak correlation was found between pre- and follow-up appetite and weight loss (r = 0.25, P = 0.294). CONCLUSIONS Twelve weeks of TENS treatment of T6 dermatome showed a positive effect in reducing appetite with sustained reduction up to 8 weeks of follow-up even after completion of the intervention. However, in the absence of dietary modification, TENS treatment was associated with low effect sizes weight and BMI loss.
Collapse
Affiliation(s)
- Pradeep Kumar Sahu
- Department of Exercise Science, University of South Carolina, Columbia, SC, United States
| | - Piyush Ranjan
- Department of Medicine, All India Institute of Medical Sciences, New Delhi, India
| | - Deepak Agrawal
- Department of Neurosurgery, Neurosciences Centre, All India Institute of Medical Sciences, New Delhi, India
| | - Garima Singh
- Neurosciences Centre, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
24
|
Colucci Cante R, Nigro F, Passannanti F, Lentini G, Gallo M, Nigro R, Budelli AL. Gut health benefits and associated systemic effects provided by functional components from the fermentation of natural matrices. Compr Rev Food Sci Food Saf 2024; 23:e13356. [PMID: 38767859 DOI: 10.1111/1541-4337.13356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 02/26/2024] [Accepted: 04/06/2024] [Indexed: 05/22/2024]
Abstract
Recently, the role of the gut microbiota in metabolic health, immunity, behavioral balance, longevity, and intestine comfort has been the object of several studies from scientific communities. They were encouraged by a growing interest from food industries and consumers toward novel fermented ingredients and formulations with powerful biological effects, such as pre, pro, and postbiotic products. Depending on the selected strains, the operating conditions, the addition of suitable reagents or enzymes, the equipment, and the reactor configurations, functional compounds with high bioactivity, such as short-chain fatty acids, gamma-aminobutyric acid, bioactive peptides, and serotonin, can be enhanced and/or produced through fermentation of several vegetable matrices. Otherwise, their formation can also be promoted directly in the gut after the dietary intake of fermented foods: In this case, fermentation will aim to increase the content of precursor substances, such as indigestible fibers, polyphenols, some amino acids, and resistant starch, which can be potentially metabolized by endogenous gut microorganisms and converted in healthy molecules. This review provides an overview of the main functional components currently investigated in literature and the associated gut health benefits. The current state of the art about fermentation technology as a promising functionalization tool to promote the direct or indirect formation of gut-health-enhancing components was deepened, highlighting the importance of optimizing microorganism selection, system setups, and process conditions according to the target compound of interest. The collected data suggested the possibility of gaining novel functional food ingredients or products rich in functional molecules through fermentation without performing additional extraction and purification stages, which are needed when conventional culture broths are used.
Collapse
Affiliation(s)
- Rosa Colucci Cante
- Department of Chemical Engineering, Materials, and Industrial Production, University of Naples Federico II, Naples, Italy
- Department of Industrial Engineering, University of Niccolò Cusano, Rome, Italy
| | - Federica Nigro
- I. T. P. Innovation and Technology Provider S.r.l., Naples, Italy
| | - Francesca Passannanti
- Department of Chemical Engineering, Materials, and Industrial Production, University of Naples Federico II, Naples, Italy
- I. T. P. Innovation and Technology Provider S.r.l., Naples, Italy
| | - Giulia Lentini
- Department of Chemical Engineering, Materials, and Industrial Production, University of Naples Federico II, Naples, Italy
| | - Marianna Gallo
- Department of Chemical Engineering, Materials, and Industrial Production, University of Naples Federico II, Naples, Italy
- Department of Industrial Engineering, University of Niccolò Cusano, Rome, Italy
- I. T. P. Innovation and Technology Provider S.r.l., Naples, Italy
| | - Roberto Nigro
- Department of Chemical Engineering, Materials, and Industrial Production, University of Naples Federico II, Naples, Italy
| | | |
Collapse
|
25
|
Hedley KE, Cuskelly A, Callister RJ, Horvat JC, Hodgson DM, Tadros MA. The medulla oblongata shows a sex-specific inflammatory response to systemic neonatal lipopolysaccharide. J Neuroimmunol 2024; 389:578316. [PMID: 38394966 DOI: 10.1016/j.jneuroim.2024.578316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 02/12/2024] [Accepted: 02/14/2024] [Indexed: 02/25/2024]
Abstract
Early life inflammation has been linked to long-term modulation of behavioural outcomes due to the central nervous system, but it is now becoming apparent it is also linked to dysfunction of visceral physiology. The medulla oblongata contains a number of nuclei critical for homeostasis, therefore we utilised the well-established model of neonatal lipopolysaccharide (LPS) exposure to examine the immediate and long-term impacts of systemic inflammation on the medulla oblongata. Wistar rats were injected with LPS or saline on postnatal days 3 and 5, with tissues collected on postnatal days 7 or 90 in order to assess expression of inflammatory mediators and microglial morphology in autonomic regions of the medulla oblongata. We observed a distinct sex-specific response of all measured inflammatory mediators at both ages, as well as significant neonatal sex differences in inflammatory mediators within saline groups. At both ages, microglial morphology had significant changes in branch length and soma size in a sex-specific manner in response to LPS exposure. This data not only highlights the strong sex-specific response of neonates to LPS administration, but also the significant life-long impact on the medulla oblongata and the potential altered control of visceral organs.
Collapse
Affiliation(s)
- Kateleen E Hedley
- School of Biomedical Sciences & Pharmacy, University of Newcastle, NSW, Australia; Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Annalisa Cuskelly
- Hunter Medical Research Institute, New Lambton Heights, NSW, Australia; School of Psychological Sciences, University of Newcastle, NSW, Australia; School of Education, University of Newcastle, NSW, Australia
| | - Robert J Callister
- School of Biomedical Sciences & Pharmacy, University of Newcastle, NSW, Australia; Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Jay C Horvat
- School of Biomedical Sciences & Pharmacy, University of Newcastle, NSW, Australia; Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Deborah M Hodgson
- Hunter Medical Research Institute, New Lambton Heights, NSW, Australia; School of Psychological Sciences, University of Newcastle, NSW, Australia
| | - Melissa A Tadros
- School of Biomedical Sciences & Pharmacy, University of Newcastle, NSW, Australia; Hunter Medical Research Institute, New Lambton Heights, NSW, Australia.
| |
Collapse
|
26
|
Tao W, Zhang Y, Wang B, Nie S, Fang L, Xiao J, Wu Y. Advances in molecular mechanisms and therapeutic strategies for central nervous system diseases based on gut microbiota imbalance. J Adv Res 2024:S2090-1232(24)00124-3. [PMID: 38579985 DOI: 10.1016/j.jare.2024.03.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Revised: 03/12/2024] [Accepted: 03/29/2024] [Indexed: 04/07/2024] Open
Abstract
BACKGROUD Central nervous system (CNS) diseases pose a serious threat to human health, but the regulatory mechanisms and therapeutic strategies of CNS diseases need to be further explored. It has been demonstrated that the gut microbiota (GM) is closely related to CNS disease. GM structure disorders, abnormal microbial metabolites, intestinal barrier destruction and elevated inflammation exist in patients with CNS diseases and promote the development of CNS diseases. More importantly, GM remodeling alleviates CNS pathology to some extent. AIM OF REVIEW Here, we have summarized the regulatory mechanism of the GM in CNS diseases and the potential treatment strategies for CNS repair based on GM regulation, aiming to provide safer and more effective strategies for CNS repair from the perspective of GM regulation. KEY SCIENTIFIC CONCEPTS OF REVIEW The abundance and composition of GM is closely associated with the CNS diseases. On the basis of in-depth analysis of GM changes in mice with CNS disease, as well as the changes in its metabolites, therapeutic strategies, such as probiotics, prebiotics, and FMT, may be used to regulate GM balance and affect its microbial metabolites, thereby promoting the recovery of CNS diseases.
Collapse
Affiliation(s)
- Wei Tao
- The Institute of Life Sciences, Wenzhou University, Wenzhou 325035, China
| | - Yanren Zhang
- The Institute of Life Sciences, Wenzhou University, Wenzhou 325035, China
| | - Bingbin Wang
- The Institute of Life Sciences, Wenzhou University, Wenzhou 325035, China
| | - Saiqun Nie
- The Institute of Life Sciences, Wenzhou University, Wenzhou 325035, China
| | - Li Fang
- The Institute of Life Sciences, Wenzhou University, Wenzhou 325035, China
| | - Jian Xiao
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China.
| | - Yanqing Wu
- The Institute of Life Sciences, Wenzhou University, Wenzhou 325035, China.
| |
Collapse
|
27
|
Ma SN, Liu XH, Cai WS. Preventive noninvasive vagal nerve stimulation reduces insufficient sleep-induced depression by improving the autonomic nervous system. Biomed Pharmacother 2024; 173:116344. [PMID: 38412716 DOI: 10.1016/j.biopha.2024.116344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 02/16/2024] [Accepted: 02/23/2024] [Indexed: 02/29/2024] Open
Abstract
BACKGROUND Depression is closely linked to an imbalance in the autonomic nervous system (ANS). However, the role of this imbalance in mediating the effects of sleep deprivation (SD) and vagus nerve stimulation (VNS) on emotional well-being is not fully understood. METHODS A population-based analysis was conducted to explore the relationship between sleep duration, depression scores, and heart rate variability (HRV). Additionally, the chronic SD mouse model was established to assess the impact of preventive transcutaneous auricular VNS (taVNS) on pathological and behavioral changes. RESULTS Our study found a significant link between sleep duration, depression severity, and HRV. Shorter sleep duration was associated with higher depression scores and lower RMSSD (a measure of HRV). In our rat model, insufficient sleep consistently impaired HRV. This effect was mitigated by taVNS, accompanied by corresponding changes in levels of IL-1β and IL-6, astrocyte and microglia activation, and tail suspension times. CONCLUSIONS Using VNS as a preventive treatment for depression-risk individuals with insufficient sleep shows promise. It not only broadens the potential applications of VNS but also sheds light on its mechanism-particularly its role in enhancing vagal nerve function and balancing the ANS, as evidenced by HRV measurements.
Collapse
Affiliation(s)
- Sai-Nan Ma
- Department of Clinical Epidemiology, Shengjing Hospital of China Medical University, Shenyang, China; Clinical Research Center, Shengjing Hospital of China Medical University, Shenyang, China; Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Xiao-Hong Liu
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Wei-Song Cai
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, China.
| |
Collapse
|
28
|
Liu S, Fu W, Fu J, Chen G, He Y, Zheng T, Ma T. Electroacupuncture alleviates intestinal inflammation via a distinct neuro-immune signal pathway in the treatment of postoperative ileus. Biomed Pharmacother 2024; 173:116387. [PMID: 38471276 DOI: 10.1016/j.biopha.2024.116387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Revised: 02/29/2024] [Accepted: 03/06/2024] [Indexed: 03/14/2024] Open
Abstract
BACKGROUND The induction of intestinal inflammation as a result of abdominal surgery is an essential factor in postoperative ileus (POI) development. Electroacupuncture (EA) at ST36 has been demonstrated to relieve intestinal inflammation and restore gastrointestinal dysmotility in POI. This study aims to elucidate the neuroimmune pathway involved in the anti-inflammatory properties of EA in POI. METHODS After intestinal manipulation (IM) was performed to induce POI, intestinal inflammation and motility were assessed 24 h post-IM, by evaluating gastrointestinal transit (GIT), cytokines expression, and leukocyte infiltration. Experimental surgery, pharmacological intervention, and genetic knockout mice were used to elucidate the neuroimmune mechanisms of EA. RESULTS EA at ST36 significantly improved GIT and reduced the expression of pro-inflammatory cytokines and leukocyte infiltration in the intestinal muscularis following IM in mice. The anti-inflammatory effectiveness of EA treatment was abolished by sub-diaphragmatic vagotomy, whereas splenectomy did not hinder the anti-inflammatory benefits of EA treatment. The hexamethonium chloride (HEX) administration contributes to a notable reduction in the EA capacity to suppress inflammation and enhance motility dysfunction, and EA is ineffective in α7 nicotinic acetylcholine receptor (α7nAChR) knockout mice. CONCLUSIONS EA at ST36 prevents intestinal inflammation and dysmotility through a neural circuit that requires vagal innervation but is independent of the spleen. Further findings revealed that the process involves enteric neurons mediating the vagal signal and requires the presence of α7nAChR. These findings suggest that utilizing EA at ST36 may represent a possible therapeutic approach for POI and other immune-related gastrointestinal diseases.
Collapse
Affiliation(s)
- Shuchang Liu
- Department of General Surgery, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin 300052, China
| | - Wei Fu
- Department of General Surgery, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin 300052, China
| | - Jingnan Fu
- Department of General Surgery, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin 300052, China; Department of Minimally Invasive Surgery, Characteristics Medical Center of Chinese People Armed Police Force, Tianjin 300162, China
| | - Guibing Chen
- Department of General Surgery, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin 300052, China; Department of Gastrointestinal Surgery, Clinical Medical College and The First Affilliated Hospital of Chengdu Medical College, Chengdu, Sichuan 610500, China
| | - Yuxin He
- Department of General Surgery, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin 300052, China
| | - Ting Zheng
- Department of General Surgery, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin 300052, China
| | - Tao Ma
- Department of General Surgery, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin 300052, China.
| |
Collapse
|
29
|
Pilloni G, Best P, Kister I, Charvet L. Heart Rate Variability (HRV) serves as an objective correlate of distress and symptom burden in multiple sclerosis. Int J Clin Health Psychol 2024; 24:100454. [PMID: 38525015 PMCID: PMC10958478 DOI: 10.1016/j.ijchp.2024.100454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Accepted: 03/08/2024] [Indexed: 03/26/2024] Open
Abstract
Background Autonomic nervous system (ANS) dysfunction is frequently seen in people living with multiple sclerosis (MS). Heart rate variability (HRV) is an easy and objective index for evaluating ANS functioning, and it has been previously used to explore the association between ANS and the experience of symptom burden in other chronic diseases. Given ANS functioning can be influenced by physical and psychological factors, this study investigated whether emotional distress and/or the presence of ANS dysfunction is associated with symptom severity in people living with MS. Methods Participants with MS and healthy controls (HC) with no history of cardiac conditions were recruited to self-collect HR data sampled from a chest strap HR monitor (PolarH10). Short-term HR signal was collected for five minutes, and time and frequency HRV analyses were performed and compared between groups. HRV values were then compared to self-reported distress (Kessler Psychological Distress Scale) and MS participants' self-reported measures of symptom burden (SymptoMScreen). Results A total of n = 23 adults with MS (51 ± 12 years, 65 % female, median Patient Determined Disease Steps [PDDS]: 3.0) and n = 23 HCs (43 ± 18 years, 40 % female) completed the study procedures. All participants were able to complete the chest strap placement and HR data capture independently. Participants with MS, compared to the HC participants, had a significantly lower parasympathetic activation as shown by lower values of the root mean square of successive differences between normal heartbeats (RMSSD: 21.86 ± 9.84 vs. 43.13 ± 20.98 ms, p = 0.002) and of high-frequency (HF) power band (HF-HRV: 32.69 ± 12.01 vs. 42.39 ± 7.96 nu, p = 0.016), indicating an overall lower HRV in the MS group. Among individuals with MS, HF-HRV was significantly correlated with the severity of self-reported MS symptoms (r = -0.548, p = 0.010). Participants with MS also reported higher levels of distress compared to HC participants (18.32 ± 6.05 vs. 15.00 ± 4.61, p = 0.050), and HRV correlated with the severity of distress in MS participants (r = -0.569, p = 0.007). A significant mediation effect was also observed, with emotional distress fully mediating the association between HRV and symptom burden. Conclusions These findings suggest the potential for ANS dysfunction, as measured by HRV (i.e., lower value of HF power), to be utilized as an objective marker of symptom burden in people living with MS. Moreover, it is apparent that the relationship between HRV and symptom burden is mediated by emotional distress.
Collapse
Affiliation(s)
- Giuseppina Pilloni
- Department of Neurology, New York University Grossman School of Medicine, 222 E 41st Street, 10th floor, New York, NY 10017, United States
| | - Pamela Best
- Department of Neurology, New York University Grossman School of Medicine, 222 E 41st Street, 10th floor, New York, NY 10017, United States
| | - Ilya Kister
- Department of Neurology, New York University Grossman School of Medicine, 222 E 41st Street, 10th floor, New York, NY 10017, United States
| | - Leigh Charvet
- Department of Neurology, New York University Grossman School of Medicine, 222 E 41st Street, 10th floor, New York, NY 10017, United States
| |
Collapse
|
30
|
Cao Y, Li R, Bai L. Vagal sensory pathway for the gut-brain communication. Semin Cell Dev Biol 2024; 156:228-243. [PMID: 37558522 DOI: 10.1016/j.semcdb.2023.07.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 06/07/2023] [Accepted: 07/20/2023] [Indexed: 08/11/2023]
Abstract
The communication between the gut and brain is crucial for regulating various essential physiological functions, such as energy balance, fluid homeostasis, immune response, and emotion. The vagal sensory pathway plays an indispensable role in connecting the gut to the brain. Recently, our knowledge of the vagal gut-brain axis has significantly advanced through molecular genetic studies, revealing a diverse range of vagal sensory cell types with distinct peripheral innervations, response profiles, and physiological functions. Here, we review the current understanding of how vagal sensory neurons contribute to gut-brain communication. First, we highlight recent transcriptomic and genetic approaches that have characterized different vagal sensory cell types. Then, we focus on discussing how different subtypes encode numerous gut-derived signals and how their activities are translated into physiological and behavioral regulations. The emerging insights into the diverse cell types and functional properties of vagal sensory neurons have paved the way for exciting future directions, which may provide valuable insights into potential therapeutic targets for disorders involving gut-brain communication.
Collapse
Affiliation(s)
- Yiyun Cao
- Chinese Institute for Brain Research, Beijing 102206, China
| | - Rui Li
- Chinese Institute for Brain Research, Beijing 102206, China; State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing, 100875, China
| | - Ling Bai
- Chinese Institute for Brain Research, Beijing 102206, China.
| |
Collapse
|
31
|
Coverdell TC, Abbott SBG, Campbell JN. Molecular cell types as functional units of the efferent vagus nerve. Semin Cell Dev Biol 2024; 156:210-218. [PMID: 37507330 PMCID: PMC10811285 DOI: 10.1016/j.semcdb.2023.07.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 07/20/2023] [Accepted: 07/20/2023] [Indexed: 07/30/2023]
Abstract
The vagus nerve vitally connects the brain and body to coordinate digestive, cardiorespiratory, and immune functions. Its efferent neurons, which project their axons from the brainstem to the viscera, are thought to comprise "functional units" - neuron populations dedicated to the control of specific vagal reflexes or organ functions. Previous research indicates that these functional units differ from one another anatomically, neurochemically, and physiologically but have yet to define their identity in an experimentally tractable way. However, recent work with genetic technology and single-cell genomics suggests that genetically distinct subtypes of neurons may be the functional units of the efferent vagus. Here we review how these approaches are revealing the organizational principles of the efferent vagus in unprecedented detail.
Collapse
Affiliation(s)
- Tatiana C Coverdell
- Biomedical Sciences Graduate Program, University of Virginia, Charlottesville, VA 22903, USA
| | - Stephen B G Abbott
- Department of Pharmacology, University of Virginia, Charlottesville, VA 22903, USA
| | - John N Campbell
- Department of Biology, University of Virginia, Charlottesville, VA 22903, USA.
| |
Collapse
|
32
|
Wang W, Zhou L, Hu Q, Gao Y, Wei Y, Tang X, Hu Y, Xu L, Liu H, Wang Z, Chen T, Li C, Wu H, Wang J, Zhang T. Correlative relationship between body mass index and heart rate variability in psychiatric disorders. Eur Arch Psychiatry Clin Neurosci 2024:10.1007/s00406-024-01768-1. [PMID: 38470538 DOI: 10.1007/s00406-024-01768-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 01/19/2024] [Indexed: 03/14/2024]
Abstract
OBJECTIVE Indicators of heart rate variability (HRV) have been used to assess the autonomic activity. However, the influence of obesity on HRV in these patients remains to be determined. This study aimed to examine how obesity (measured with the body mass index [BMI]) affects HRV and determine whether the effect varies among different psychiatric disorders. We recruited 3159 consecutive patients, including 1744 with schizophrenia, 966 with mood disorders, and 449 with anxiety disorders. Patients were divided into four groups based on BMI: underweight (< 18.5), normal weight (18.5-23.9), overweight (24-27.9), and obese (≥ 28). The cardiovascular status was assessed using several time- and frequency-based HRV indicators, measured via electrocardiogram signals recorded for 5 min. The mean BMI of the participants was 23.6 ± 4.0. The patients in the overweight and obese groups were 29.4% and 13.6% of the total, respectively. The HRV indicators were higher in underweight and normal-weight patients than in the overweight and obese ones. After stratification based on the psychiatric diagnosis, the patients with mood disorders showed lower HRV than those with schizophrenia or anxiety disorder in the normal-weight group. In contrast, in the overweight and obese groups the patients with mood disorders showed higher HRV than those with the other disorders. The HRV variables were significantly associated with BMI, and higher BMI was associated with higher heart rates and lower HRV. These results indicate that weight gain in psychiatric disorders is associated with an imbalance in autonomic nerve activity. However, the relationship between autonomic activity, weight gain, and psychiatric disorders warrants further investigation.
Collapse
Affiliation(s)
- WenZheng Wang
- Shanghai Mental Health Center, Shanghai Jiaotong University School of Medicine, Shanghai Engineering Research Center of Intelligent Psychological Evaluation and Intervention, Shanghai Key Laboratory of Psychotic Disorders, Shanghai, 200030, China
| | - LinLin Zhou
- Shanghai Mental Health Center, Shanghai Jiaotong University School of Medicine, Shanghai Engineering Research Center of Intelligent Psychological Evaluation and Intervention, Shanghai Key Laboratory of Psychotic Disorders, Shanghai, 200030, China
| | - Qiang Hu
- Department of Psychiatry, Zhen Jiang Mental Health Center, Zhenjiang, People's Republic of China
| | - YuQing Gao
- Shanghai Mental Health Center, Shanghai Jiaotong University School of Medicine, Shanghai Engineering Research Center of Intelligent Psychological Evaluation and Intervention, Shanghai Key Laboratory of Psychotic Disorders, Shanghai, 200030, China
| | - YanYan Wei
- Shanghai Mental Health Center, Shanghai Jiaotong University School of Medicine, Shanghai Engineering Research Center of Intelligent Psychological Evaluation and Intervention, Shanghai Key Laboratory of Psychotic Disorders, Shanghai, 200030, China
| | - XiaoChen Tang
- Shanghai Mental Health Center, Shanghai Jiaotong University School of Medicine, Shanghai Engineering Research Center of Intelligent Psychological Evaluation and Intervention, Shanghai Key Laboratory of Psychotic Disorders, Shanghai, 200030, China
| | - YeGang Hu
- Shanghai Mental Health Center, Shanghai Jiaotong University School of Medicine, Shanghai Engineering Research Center of Intelligent Psychological Evaluation and Intervention, Shanghai Key Laboratory of Psychotic Disorders, Shanghai, 200030, China
| | - LiHua Xu
- Shanghai Mental Health Center, Shanghai Jiaotong University School of Medicine, Shanghai Engineering Research Center of Intelligent Psychological Evaluation and Intervention, Shanghai Key Laboratory of Psychotic Disorders, Shanghai, 200030, China
| | - HaiChun Liu
- Department of Automation, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - ZiXuan Wang
- Shanghai Xinlianxin Psychological Counseling Center, Shanghai, China
| | - Tao Chen
- Big Data Research Lab, University of Waterloo, Waterloo, ON, Canada
- Labor and Worklife Program, Harvard University, Massachusetts, USA
| | - ChunBo Li
- Shanghai Mental Health Center, Shanghai Jiaotong University School of Medicine, Shanghai Engineering Research Center of Intelligent Psychological Evaluation and Intervention, Shanghai Key Laboratory of Psychotic Disorders, Shanghai, 200030, China
| | - HaiSu Wu
- Shanghai Mental Health Center, Shanghai Jiaotong University School of Medicine, Shanghai Engineering Research Center of Intelligent Psychological Evaluation and Intervention, Shanghai Key Laboratory of Psychotic Disorders, Shanghai, 200030, China.
| | - JiJun Wang
- Shanghai Mental Health Center, Shanghai Jiaotong University School of Medicine, Shanghai Engineering Research Center of Intelligent Psychological Evaluation and Intervention, Shanghai Key Laboratory of Psychotic Disorders, Shanghai, 200030, China.
- Center for Excellence in Brain Science and Intelligence Technology (CEBSIT), Chinese Academy of Science, Beijing, People's Republic of China.
- Institute of Psychology and Behavioral Science, Shanghai Jiao Tong University, Shanghai, People's Republic of China.
| | - TianHong Zhang
- Shanghai Mental Health Center, Shanghai Jiaotong University School of Medicine, Shanghai Engineering Research Center of Intelligent Psychological Evaluation and Intervention, Shanghai Key Laboratory of Psychotic Disorders, Shanghai, 200030, China.
| |
Collapse
|
33
|
Teckentrup V, Kroemer NB. Mechanisms for survival: vagal control of goal-directed behavior. Trends Cogn Sci 2024; 28:237-251. [PMID: 38036309 DOI: 10.1016/j.tics.2023.11.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 11/01/2023] [Accepted: 11/02/2023] [Indexed: 12/02/2023]
Abstract
Survival is a fundamental physiological drive, and neural circuits have evolved to prioritize actions that meet the energy demands of the body. This fine-tuning of goal-directed actions based on metabolic states ('allostasis') is deeply rooted in our brain, and hindbrain nuclei orchestrate the vital communication between the brain and body through the vagus nerve. Despite mounting evidence for vagal control of allostatic behavior in animals, its broader function in humans is still contested. Based on stimulation studies, we propose that the vagal afferent pathway supports transitions between survival modes by gating the integration of ascending bodily signals, thereby regulating reward-seeking. By reconceptualizing vagal signals as catalysts for goal-directed behavior, our perspective opens new avenues for theory-driven translational work in mental disorders.
Collapse
Affiliation(s)
- Vanessa Teckentrup
- Department of Psychiatry and Psychotherapy, Tübingen Center for Mental Health, University of Tübingen, 72076 Tübingen, Germany; School of Psychology and Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin 2, Ireland
| | - Nils B Kroemer
- Department of Psychiatry and Psychotherapy, Tübingen Center for Mental Health, University of Tübingen, 72076 Tübingen, Germany; Section of Medical Psychology, Department of Psychiatry and Psychotherapy, Faculty of Medicine, University of Bonn, 53127 Bonn, Germany; German Center for Mental Health (DZPG), 72076 Tübingen, Germany.
| |
Collapse
|
34
|
Shi X, Zhao L, Luo H, Deng H, Wang X, Ren G, Zhang L, Tao Q, Liang S, Liu N, Huang X, Zhang X, Yang X, Sun J, Qin W, Kang X, Han Y, Pan Y, Fan D. Transcutaneous Auricular Vagal Nerve Stimulation Is Effective for the Treatment of Functional Dyspepsia: A Multicenter, Randomized Controlled Study. Am J Gastroenterol 2024; 119:521-531. [PMID: 37787432 DOI: 10.14309/ajg.0000000000002548] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2023] [Accepted: 09/22/2023] [Indexed: 10/04/2023]
Abstract
INTRODUCTION Vagal nerve stimulation (VNS) can be used to modulate gastrointestinal motility, inflammation, and nociception. However, it remains unclear whether VNS is effective in adult patients with functional dyspepsia (FD). We investigated the effectiveness of transcutaneous auricular VNS (taVNS) in patients with FD. METHODS Consecutive patients with FD meeting Rome IV criteria with modified FD Symptom Diary score ≥10 were enrolled. Patients were randomly allocated to 10-Hz taVNS (V10 group), 25-Hz taVNS (V25 group), or sham group, with 30 minutes of treatment twice a day for 4 weeks. The primary outcome was the response rate at week 4, defined as the proportion of patients whose modified FD Symptom Diary score was reduced ≥5 when compared with the baseline. Secondary outcomes included adequate relief rate and adverse events. RESULTS A total of 300 patients were randomized to V10 (n = 101), V25 (n = 99), and sham groups (n = 100). After 4 weeks of treatment, V10 and V25 groups had a higher response rate (81.2% vs 75.9% vs 47%, both P < 0.001) and adequate relief rate (85.1% vs 80.8% vs 67%, both P < 0.05) compared with the sham group. There was no significant difference between V10 and V25 in response rate and adequate relief rate (both P > 0.05). The efficacy of taVNS (both 10 and 25 Hz) lasted at week 8 and week 12 during follow-up period. Adverse events were all mild and comparable among the 3 groups (1%-3%). DISCUSSION Our study firstly showed that 4-week taVNS (both 10 and 25 Hz) was effective and safe for the treatment of adult FD ( clinicaltrials.gov number: NCT04668534).
Collapse
Affiliation(s)
- Xin Shi
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, China
| | - Lina Zhao
- Department of Radiotherapy, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Hui Luo
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, China
| | - Hui Deng
- Engineering Research Center of Molecular and Neuro Imaging of Ministry of Education, Intelligent Non-Invasive Neuromodulation and Transformation Joint Laboratory, School of Life Science and Technology, Xidian University, Xi'an, China
| | - Xiangping Wang
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, China
| | - Gui Ren
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, China
| | - Linhui Zhang
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, China
| | - Qin Tao
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, China
| | - Shuhui Liang
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, China
| | - Na Liu
- Department of Gastroenterology, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Xiaojun Huang
- Department of Gastroenterology, Lanzhou University Second Hospital, Lanzhou, China
| | - Xiaoyin Zhang
- Department of Gastroenterology, National Clinical Research Center of Infectious Disease, The Third People's Hospital of Shenzhen, The Second Affiliated Hospital of Southern University of Science and Technology, Shenzhen, China
| | - Xuejuan Yang
- Engineering Research Center of Molecular and Neuro Imaging of Ministry of Education, Intelligent Non-Invasive Neuromodulation and Transformation Joint Laboratory, School of Life Science and Technology, Xidian University, Xi'an, China
| | - Jinbo Sun
- Engineering Research Center of Molecular and Neuro Imaging of Ministry of Education, Intelligent Non-Invasive Neuromodulation and Transformation Joint Laboratory, School of Life Science and Technology, Xidian University, Xi'an, China
| | - Wei Qin
- Engineering Research Center of Molecular and Neuro Imaging of Ministry of Education, Intelligent Non-Invasive Neuromodulation and Transformation Joint Laboratory, School of Life Science and Technology, Xidian University, Xi'an, China
| | - Xiaoyu Kang
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, China
| | - Ying Han
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, China
| | - Yanglin Pan
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, China
| | - Daiming Fan
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, China
| |
Collapse
|
35
|
Nabar S, Fernandez J, Prakash V, Safder S. Gastrointestinal manifestations seen in pediatric patients diagnosed with small fiber neuropathy. J Pediatr Gastroenterol Nutr 2024; 78:583-591. [PMID: 38504414 DOI: 10.1002/jpn3.12099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 08/22/2023] [Accepted: 09/23/2023] [Indexed: 03/21/2024]
Abstract
OBJECTIVES Small fiber neuropathy (SFN) affects the fibers involved in cutaneous and visceral pain and temperature sensation and are a crucial part of the autonomic nervous system. Autonomic dysfunction secondary to SFN and autoimmune receptor antibodies is being increasingly recognized, and gastrointestinal (GI) manifestations include constipation, early satiety, nausea, vomiting, and diarrhea. Enteric nervous system involvement may be a possible explanation of abnormal GI motility patterns seen in these patients. METHODS Children suspected to have SFN based on symptoms underwent skin biopsy at the Child Neurology clinic at Arnold Palmer Hospital for Children, which was processed at Therapath™ Neuropathology. SFN was diagnosed using epidermal nerve fiber density values that were below 5th percentile from the left distal leg (calf) as reported per Therapath™ laboratory. RESULTS Twenty-six patients were diagnosed with SFN. Retrospective chart review was performed, including demographic data, clinical characteristics, and evaluation. A majority of patients were white adolescent females. Autonomic dysfunction, including orthostasis and temperature dysregulation were seen in 61.5% of patients (p = 0.124). Somatosensory symptoms, including pain or numbness were seen in 85% of patients (p < 0.001). GI symptoms were present in 85% of patients (p < 0.001) with constipation being the most common symptom seen in 50% of patients. This correlated with the motility testing results. CONCLUSIONS Pediatric patients with SFN commonly have GI symptoms, which may be the main presenting symptom. It is important to recognize and look for symptoms of small fiber neuropathy in children with refractory GI symptoms that may explain multisystemic complaints often seen in these patients.
Collapse
Affiliation(s)
- Shruti Nabar
- Center for Digestive Health and Nutrition, Orlando Health Arnold Palmer Hospital for Children, Orlando, Florida, USA
| | - Jenelle Fernandez
- Division of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, Morsani College of Medicine, University of South Florida, Tampa, Florida, USA
| | - Vikram Prakash
- Orlando Health Arnold Palmer Hospital for Children Neurology, Orlando, Florida, USA
| | - Shaista Safder
- Center for Digestive Health and Nutrition, Orlando Health Arnold Palmer Hospital for Children, Orlando, Florida, USA
| |
Collapse
|
36
|
Suprunowicz M, Tomaszek N, Urbaniak A, Zackiewicz K, Modzelewski S, Waszkiewicz N. Between Dysbiosis, Maternal Immune Activation and Autism: Is There a Common Pathway? Nutrients 2024; 16:549. [PMID: 38398873 PMCID: PMC10891846 DOI: 10.3390/nu16040549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 02/05/2024] [Accepted: 02/15/2024] [Indexed: 02/25/2024] Open
Abstract
Autism spectrum disorder (ASD) is a neuropsychiatric condition characterized by impaired social interactions and repetitive stereotyped behaviors. Growing evidence highlights an important role of the gut-brain-microbiome axis in the pathogenesis of ASD. Research indicates an abnormal composition of the gut microbiome and the potential involvement of bacterial molecules in neuroinflammation and brain development disruptions. Concurrently, attention is directed towards the role of short-chain fatty acids (SCFAs) and impaired intestinal tightness. This comprehensive review emphasizes the potential impact of maternal gut microbiota changes on the development of autism in children, especially considering maternal immune activation (MIA). The following paper evaluates the impact of the birth route on the colonization of the child with bacteria in the first weeks of life. Furthermore, it explores the role of pro-inflammatory cytokines, such as IL-6 and IL-17a and mother's obesity as potentially environmental factors of ASD. The purpose of this review is to advance our understanding of ASD pathogenesis, while also searching for the positive implications of the latest therapies, such as probiotics, prebiotics or fecal microbiota transplantation, targeting the gut microbiota and reducing inflammation. This review aims to provide valuable insights that could instruct future studies and treatments for individuals affected by ASD.
Collapse
Affiliation(s)
| | | | | | | | - Stefan Modzelewski
- Department of Psychiatry, Medical University of Bialystok, pl. Wołodyjowskiego 2, 15-272 Białystok, Poland; (M.S.); (N.T.); (A.U.); (K.Z.); (N.W.)
| | | |
Collapse
|
37
|
Bauer KC, Trehan R, Ruf B, Myojin Y, Benmebarek MR, Ma C, Seifert M, Nur A, Qi J, Huang P, Soliman M, Green BL, Wabitsch S, Springer DA, Rodriguez-Matos FJ, Ghabra S, Gregory SN, Matta J, Dawson B, Golino J, Xie C, Dzutsev A, Trinchieri G, Korangy F, Greten TF. The Gut Microbiome Controls Liver Tumors via the Vagus Nerve. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.23.576951. [PMID: 38328040 PMCID: PMC10849697 DOI: 10.1101/2024.01.23.576951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2024]
Abstract
Liver cancer ranks amongst the deadliest cancers. Nerves have emerged as an understudied regulator of tumor progression. The parasympathetic vagus nerve influences systemic immunity via acetylcholine (ACh). Whether cholinergic neuroimmune interactions influence hepatocellular carcinoma (HCC) remains uncertain. Liver denervation via hepatic vagotomy (HV) significantly reduced liver tumor burden, while pharmacological enhancement of parasympathetic tone promoted tumor growth. Cholinergic disruption in Rag1KO mice revealed that cholinergic regulation requires adaptive immunity. Further scRNA-seq and in vitro studies indicated that vagal ACh dampens CD8+ T cell activity via muscarinic ACh receptor (AChR) CHRM3. Depletion of CD8+ T cells abrogated HV outcomes and selective deletion of Chrm3 on CD8 + T cells inhibited liver tumor growth. Beyond tumor-specific outcomes, vagotomy improved cancer-associated fatigue and anxiety-like behavior. As microbiota transplantation from HCC donors was sufficient to impair behavior, we investigated putative microbiota-neuroimmune crosstalk. Tumor, rather than vagotomy, robustly altered fecal bacterial composition, increasing Desulfovibrionales and Clostridial taxa. Strikingly, in tumor-free mice, vagotomy permitted HCC-associated microbiota to activate hepatic CD8+ T cells. These findings reveal that gut bacteria influence behavior and liver anti-tumor immunity via a dynamic and pharmaceutically targetable, vagus-liver axis.
Collapse
|
38
|
Tucker JAL, Bornath DPD, McCarthy SF, Hazell TJ. Leptin and energy balance: exploring Leptin's role in the regulation of energy intake and energy expenditure. Nutr Neurosci 2024; 27:87-95. [PMID: 36583502 DOI: 10.1080/1028415x.2022.2161135] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Leptin is a tonic appetite-regulating hormone, which is integral for the long-term regulation of energy balance. The current evidence suggests that the typical orexigenic or anorexigenic response of many of these appetite-regulating hormones, most notably ghrelin and cholecystokinin (CCK), require leptin to function whereas glucagon-like peptide-1 (GLP-1) is required for leptin to function, and these responses are altered when leptin injection or gene therapy is administered in combination with these same hormones or respective agonists. The appetite-regulatory pathway is complex, thus peptide tyrosine tyrosine (PYY), brain-derived neurotrophic factor (BDNF), orexin-A (OXA), and amylin also maintain ties to leptin, however these are less well understood. While reviews to date have focused on the existing relationships between leptin and the various neuropeptide modulators of appetite within the central nervous system (CNS) or it's role in thermogenesis, no review paper has synthesised the information regarding the interactions between appetite-regulating hormones and how leptin as a chronic regulator of energy balance can influence the acute appetite-regulatory response. Current evidence suggests that potential relationships exist between leptin and the circulating peripheral appetite hormones ghrelin, GLP-1, CCK, OXA and amylin to exhibit either synergistic or opposing effects on appetite inhibition. Though more research is warranted, leptin appears to be integral in both energy intake and energy expenditure. More specifically, functional leptin receptors appear to play an essential role in these processes.
Collapse
Affiliation(s)
- Jessica A L Tucker
- Department of Kinesiology and Physical Education, Wilfrid Laurier University, Waterloo, Canada
| | - Derek P D Bornath
- Department of Kinesiology and Physical Education, Wilfrid Laurier University, Waterloo, Canada
| | - Seth F McCarthy
- Department of Kinesiology and Physical Education, Wilfrid Laurier University, Waterloo, Canada
| | - Tom J Hazell
- Department of Kinesiology and Physical Education, Wilfrid Laurier University, Waterloo, Canada
| |
Collapse
|
39
|
Chandra R, Sokratian A, Chavez KR, King S, Swain SM, Snyder JC, West AB, Liddle RA. Gut mucosal cells transfer α-synuclein to the vagus nerve. JCI Insight 2023; 8:e172192. [PMID: 38063197 PMCID: PMC10795834 DOI: 10.1172/jci.insight.172192] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 10/27/2023] [Indexed: 12/18/2023] Open
Abstract
Epidemiological and histopathological findings have raised the possibility that misfolded α-synuclein protein might spread from the gut to the brain and increase the risk of Parkinson's disease. Although past experimental studies in mouse models have relied on gut injections of exogenous recombinant α-synuclein fibrils to study gut-to-brain α-synuclein transfer, the possible origins of misfolded α-synuclein within the gut have remained elusive. We recently demonstrated that sensory cells of intestinal mucosa express α-synuclein. Here, we employed mouse intestinal organoids expressing human α-synuclein to observe the transfer of α-synuclein protein from epithelial cells in organoids to cocultured nodose neurons devoid of α-synuclein. In mice expressing human α-synuclein, but no mouse α-synuclein, α-synuclein fibril-templating activity emerged in α-synuclein-seeded fibril aggregation assays in intestine, vagus nerve, and dorsal motor nucleus. In newly engineered transgenic mice that restrict pathological human α-synuclein expression to intestinal epithelial cells, α-synuclein fibril-templating activity transfered to the vagus nerve and dorsal motor nucleus. Subdiaphragmatic vagotomy prior to induction of α-synuclein expression in intestinal epithelial cells effectively protected the hindbrain from emergence of α-synuclein fibril-templating activity. Overall, these findings highlight a potential non-neuronal source of fibrillar α-synuclein protein that might arise in gut mucosal cells.
Collapse
Affiliation(s)
| | | | | | | | | | - Joshua C. Snyder
- Department of Surgery, and
- Department of Cell Biology, Duke University, Durham, North Carolina, USA
| | - Andrew B. West
- Department of Pharmacology and Cancer Biology
- Duke Institute for Brain Sciences, Durham, North Carolina, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, Maryland, USA
| | - Rodger A. Liddle
- Department of Medicine
- Duke Institute for Brain Sciences, Durham, North Carolina, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, Maryland, USA
- Department of Veterans Affairs, Chevy Chase, Maryland, USA
| |
Collapse
|
40
|
Dirr EW, Jiracek LG, Baekey DM, J Martyniuk C, Otto KJ, Zubcevic J. Subdiaphragmatic vagal nerve stimulation attenuates the development of hypertension and alters nucleus of the solitary tract transcriptional networks in the spontaneously hypertensive rat. Physiol Genomics 2023; 55:606-617. [PMID: 37746712 PMCID: PMC11178265 DOI: 10.1152/physiolgenomics.00016.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 08/18/2023] [Accepted: 09/18/2023] [Indexed: 09/26/2023] Open
Abstract
Augmented vagal signaling may be therapeutic in hypertension. Most studies to date have used stimulation of the cervical vagal branches. Here, we investigated the effects of chronic intermittent electric stimulation of the ventral subdiaphragmatic vagal nerve branch (sdVNS) on long-term blood pressure, immune markers, and gut microbiota in the spontaneously hypertensive rat (SHR), a rodent model of hypertension characterized by vagal dysfunction, gut dysbiosis, and low-grade inflammation. We evaluated the effects of sdVNS on transcriptional networks in the nucleus of the solitary tract (NTS), a major cardioregulatory brain region with direct gut vagal projections. Male juvenile SHRs were implanted with radiotelemetry transmitters and vagal nerve cuffs for chronic intermittent electric sdVNS, applied three times per day for 7 consecutive weeks followed by 1 wk of no stimulation. Blood pressure was measured once a week using telemetry in the sdVNS group as well as age-matched sham-stimulated SHR controls. At the endpoint, colonic and circulating inflammatory markers, corticosterone, and circulating catecholamines were investigated. Bacterial 16 s sequencing measured gut bacterial abundance and composition. RNA sequencing evaluated the effects of sdVNS on transcriptional networks in the NTS. SHRs that received sdVNS exhibited attenuated development of hypertension compared with sham animals. No changes in peripheral inflammatory markers, corticosterone, or catecholamines and no major differences in gut bacterial diversity and composition were observed following sdVNS, apart from decreased abundance of Defluviitaleaceale bacterium detected in sdVNS SHRs compared with sham animals. RNA sequencing revealed significant sdVNS-dependent modulation of select NTS transcriptional networks, including catecholaminergic and corticosteroid networks.NEW & NOTEWORTHY We show that stimulation of the ventral subdiaphragmatic vagal nerve branch may be a promising potential approach to treating hypertension. The data are especially encouraging given that rodents received only 30 min per day of intermittent stimulation therapy and in view of the potential of long-term blood pressure effects that are not stimulus-locked.
Collapse
Affiliation(s)
- Elliott W Dirr
- Department of Physiology and Pharmacology, University of Toledo, Toledo, Ohio, United States
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, Florida, United States
| | - Ladan G Jiracek
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, Florida, United States
| | - David M Baekey
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, Florida, United States
| | - Christopher J Martyniuk
- Department of Physiological Sciences, University of Florida, Gainesville, Florida, United States
| | - Kevin J Otto
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, Florida, United States
- Department of Neuroscience, University of Florida, Gainesville, Florida, United States
- Department of Neurology, University of Florida, Gainesville, Florida, United States
- Department of Materials Science and Engineering, University of Florida, Gainesville, Florida, United States
- Department of Electrical and Computer Engineering, University of Florida, Gainesville, Florida, United States
| | - Jasenka Zubcevic
- Department of Physiology and Pharmacology, University of Toledo, Toledo, Ohio, United States
| |
Collapse
|
41
|
Akshay A, Gasim R, Ali TE, Kumar YS, Hassan A. Unlocking the Gut-Cardiac Axis: A Paradigm Shift in Cardiovascular Health. Cureus 2023; 15:e51039. [PMID: 38264397 PMCID: PMC10805229 DOI: 10.7759/cureus.51039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/24/2023] [Indexed: 01/25/2024] Open
Abstract
The gut-cardiac axis represents an emerging area of research focusing on the relationship between gut health and cardiovascular function. This narrative review examines the Gut-Cardiac Axis, emphasizing its emerging role in cardiovascular health and disease management. Traditionally viewed as a component of the digestive system, the gut is now recognized for its significant influence on cardiac health. The gut microbiota, its metabolites, and gut-related inflammation are key factors affecting heart structure and function. This review highlights how dietary and nutritional interventions can effectively modulate the gut-cardiac axis, leading to personalized strategies for optimizing cardiovascular health. We discuss the clinical relevance of the gut-cardiac axis, particularly its role in providing diagnostic and prognostic markers for cardiovascular diseases. This exploration of the gut-cardiac axis marks a significant shift in cardiology, integrating gut health into cardiovascular risk assessment and treatment strategies. The review provides an in-depth overview of current research and its potential to impact cardiovascular medicine significantly. We emphasize the importance of this research in advancing patient care and improving cardiac outcomes, underlining the potential of the gut-cardiac axis to transform cardiovascular health management.
Collapse
Affiliation(s)
| | - Rayan Gasim
- Internal Medicine, University of Khartoum, Khartoum, SDN
| | - Thowaiba E Ali
- Medicine and Surgery, University of Tennessee, Chattanooga, USA
| | | | | |
Collapse
|
42
|
Dorsey AF, Miller EM. Revisiting geophagy: An evolved sickness behavior to microbiome-mediated gastrointestinal inflammation. Evol Anthropol 2023; 32:325-335. [PMID: 37661330 DOI: 10.1002/evan.22004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 07/19/2023] [Accepted: 08/21/2023] [Indexed: 09/05/2023]
Abstract
Geophagy, the consumption of clay or similar substances, is known as an evolved behavior that protects vulnerable populations, such as pregnant women and children, against gastrointestinal injury. However, perplexing questions remain, like the presence of geophagy in the absence of overt gastrointestinal infection and the potential causal relationship between geophagy and iron deficiency anemia. In this review, we hypothesize that geophagy is an inflammation-mediated sickness behavior regulated via the vagus nerve. We further hypothesize that the gut microbiome plays a critical role in mediating the relationship between inflammation and geophagy. By including inflammation and the microbiome within the existing protection hypothesis, we can explain how subclinical gastrointestinal states induce geophagy. Furthermore, we can explain how gastrointestinal inflammation is responsible for both geophagy and iron-deficiency anemia, explaining why the two phenomena frequently co-occur. Ultimately, defining geophagy as a sickness behavior allows us to integrate the gut-brain axis into geophagy research.
Collapse
Affiliation(s)
- Achsah F Dorsey
- Department of Anthropology, University of Massachusetts, Amherst, Massachusetts, USA
| | - Elizabeth M Miller
- Department of Anthropology, University of South Florida, Tampa, Florida, USA
| |
Collapse
|
43
|
Huang J, Huang W, Yi J, Deng Y, Li R, Chen J, Shi J, Qiu Y, Wang T, Chen X, Zhang X, Xiang AP. Mesenchymal stromal cells alleviate depressive and anxiety-like behaviors via a lung vagal-to-brain axis in male mice. Nat Commun 2023; 14:7406. [PMID: 37973914 PMCID: PMC10654509 DOI: 10.1038/s41467-023-43150-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Accepted: 11/01/2023] [Indexed: 11/19/2023] Open
Abstract
Major depressive disorder (MDD) is one of the most common and disabling mental disorders, and current strategies remain inadequate. Although mesenchymal stromal cells (MSCs) have shown beneficial effects in experimental models of depression, underlying mechanisms remain elusive. Here, using murine depression models, we demonstrated that MSCs could alleviate depressive and anxiety-like behaviors not due to a reduction in proinflammatory cytokines, but rather activation of dorsal raphe nucleus (DRN) 5-hydroxytryptamine (5-HT) neurons. Mechanistically, peripheral delivery of MSCs activated pulmonary innervating vagal sensory neurons, which projected to the nucleus tractus solitarius, inducing the release of 5-HT in DRN. Furthermore, MSC-secreted brain-derived neurotrophic factor activated lung sensory neurons through tropomyosin receptor kinase B (TrkB), and inhalation of a TrkB agonist also achieved significant therapeutic effects in male mice. This study reveals a role of peripheral MSCs in regulating central nervous system function and demonstrates a potential "lung vagal-to-brain axis" strategy for MDD.
Collapse
Affiliation(s)
- Jing Huang
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China
- National-Local Joint Engineering Research Center for Stem Cells and Regenerative Medicine, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China
| | - Weijun Huang
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China
- National-Local Joint Engineering Research Center for Stem Cells and Regenerative Medicine, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China
| | - Junzhe Yi
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China
- National-Local Joint Engineering Research Center for Stem Cells and Regenerative Medicine, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China
| | - Yiwen Deng
- Key Laboratory of Medical Transformation of Jiujiang, Jiujiang University, Jiujiang, Jiangxi, 332005, China
| | - Ruijie Li
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China
- National-Local Joint Engineering Research Center for Stem Cells and Regenerative Medicine, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China
| | - Jieying Chen
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China
- National-Local Joint Engineering Research Center for Stem Cells and Regenerative Medicine, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China
| | - Jiahao Shi
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China
- National-Local Joint Engineering Research Center for Stem Cells and Regenerative Medicine, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China
| | - Yuan Qiu
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China
- National-Local Joint Engineering Research Center for Stem Cells and Regenerative Medicine, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China
- Department of Histoembryology and Cell Biology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China
| | - Tao Wang
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China
- National-Local Joint Engineering Research Center for Stem Cells and Regenerative Medicine, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China
- Department of Histoembryology and Cell Biology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China
| | - Xiaoyong Chen
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China
- National-Local Joint Engineering Research Center for Stem Cells and Regenerative Medicine, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China
- Department of Histoembryology and Cell Biology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China
| | - Xiaoran Zhang
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China.
- National-Local Joint Engineering Research Center for Stem Cells and Regenerative Medicine, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China.
- Department of Histoembryology and Cell Biology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China.
| | - Andy Peng Xiang
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China.
- National-Local Joint Engineering Research Center for Stem Cells and Regenerative Medicine, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China.
- Department of Histoembryology and Cell Biology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China.
| |
Collapse
|
44
|
De Looze K, Tanasescu R, Vonck K. The gut-microbiota-brain axis: An introduction to a special issue on its role in neurological disorders. Eur J Neurol 2023; 30:3411-3416. [PMID: 37817414 DOI: 10.1111/ene.16080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Accepted: 09/15/2023] [Indexed: 10/12/2023]
Affiliation(s)
- Katrien De Looze
- Department of Internal Medicine, Ghent University Hospital, Ghent, Belgium
| | - Radu Tanasescu
- Department of Neurology, Queen's Medical Centre, Nottingham University Hospitals NHS Trust, Nottingham, UK
- Academic Clinical Neurology, Mental Health, and Clinical Neurosciences Academic Unit, School of Medicine, University of Nottingham, Nottingham, UK
| | - Kristl Vonck
- Department of Neurology, 4Brain, Ghent University Hospital, Ghent, Belgium
| |
Collapse
|
45
|
Wang J, Liu G, Xu K, Ai K, Huang W, Zhang J. The role of neurotransmitters in mediating the relationship between brain alterations and depressive symptoms in patients with inflammatory bowel disease. Hum Brain Mapp 2023; 44:5357-5371. [PMID: 37530546 PMCID: PMC10543356 DOI: 10.1002/hbm.26439] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 06/07/2023] [Accepted: 07/10/2023] [Indexed: 08/03/2023] Open
Abstract
A growing body of evidence from neuroimaging studies suggests that inflammatory bowel disease (IBD) is associated with functional and structural alterations in the central nervous system and that it has a potential link to emotional symptoms, such as anxiety and depression. However, the neurochemical underpinnings of depression symptoms in IBD remain unclear. We hypothesized that changes in cortical gamma-aminobutyric acid (GABA+) and glutamine (Glx) concentrations are related to cortical thickness and resting-state functional connectivity in IBD as compared to healthy controls. To test this, we measured whole-brain cortical thickness and functional connectivity within the medial prefrontal cortex (mPFC), as well as the concentrations of neurotransmitters in the same brain region. We used the edited magnetic resonance spectroscopy (MRS) with the MEGA-PRESS sequence at a 3 T scanner to quantitate the neurotransmitter levels in the mPFC. Subjects with IBD (N = 37) and healthy control subjects (N = 32) were enrolled in the study. Compared with healthy controls, there were significantly decreased GABA+ and Glx concentrations in the mPFC of patients with IBD. The cortical thickness of patients with IBD was thin in two clusters that included the right medial orbitofrontal cortex and the right posterior cingulate cortex. A seed-based functional connectivity analysis indicated that there was higher connectivity of the mPFC with the left precuneus cortex (PC) and the posterior cingulate cortex, and conversely, lower connectivity in the left frontal pole was observed. The functional connectivity between the mPFC and the left PC was negatively correlated with the IBD questionnaire score (r = -0.388, p = 0.018). GABA+ concentrations had a negative correlation with the Hamilton Depression Scale (HAMD) score (r = -0.497, p = 0.002). Glx concentration was negatively correlated with the HAMD score (r = -0.496, p = 0.002) and positively correlated with the Short-Form McGill Pain Questionnaire score (r = 0.330, p = 0.046, uncorrected). There was a significant positive correlation between the ratio of Glx to GABA+ and the HAMD score (r = 0.428, p = 0.008). Mediation analysis revealed that GABA+ significantly mediated the main effect of the relationship between the structural and functional alterations and the severity of depression in patients with IBD. Our study provides initial evidence of neurochemistry that can be used to identify potential mechanisms underlying the modulatory effects of GABA+ on the development of depression in patients with IBD.
Collapse
Affiliation(s)
- Jun Wang
- Department of Magnetic ResonanceLanzhou University Second HospitalLanzhouChina
- Second Clinical SchoolLanzhou UniversityLanzhouChina
- Gansu Province Clinical Research Center for Functional and Molecular ImagingLanzhou University Second HospitalLanzhouChina
| | - Guangyao Liu
- Department of Magnetic ResonanceLanzhou University Second HospitalLanzhouChina
- Gansu Province Clinical Research Center for Functional and Molecular ImagingLanzhou University Second HospitalLanzhouChina
| | - Kun Xu
- Department of Magnetic ResonanceLanzhou University Second HospitalLanzhouChina
- Second Clinical SchoolLanzhou UniversityLanzhouChina
- Gansu Province Clinical Research Center for Functional and Molecular ImagingLanzhou University Second HospitalLanzhouChina
| | - Kai Ai
- Deparment of Clinical and Technical Support, Philips HealthcareXi'anChina
| | - Wenjing Huang
- Department of Magnetic ResonanceLanzhou University Second HospitalLanzhouChina
- Second Clinical SchoolLanzhou UniversityLanzhouChina
- Gansu Province Clinical Research Center for Functional and Molecular ImagingLanzhou University Second HospitalLanzhouChina
| | - Jing Zhang
- Department of Magnetic ResonanceLanzhou University Second HospitalLanzhouChina
- Second Clinical SchoolLanzhou UniversityLanzhouChina
- Gansu Province Clinical Research Center for Functional and Molecular ImagingLanzhou University Second HospitalLanzhouChina
| |
Collapse
|
46
|
Zheng Y, Bonfili L, Wei T, Eleuteri AM. Understanding the Gut-Brain Axis and Its Therapeutic Implications for Neurodegenerative Disorders. Nutrients 2023; 15:4631. [PMID: 37960284 PMCID: PMC10648099 DOI: 10.3390/nu15214631] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 10/25/2023] [Accepted: 10/28/2023] [Indexed: 11/15/2023] Open
Abstract
The gut-brain axis (GBA) is a complex bidirectional communication network connecting the gut and brain. It involves neural, immune, and endocrine communication pathways between the gastrointestinal (GI) tract and the central nervous system (CNS). Perturbations of the GBA have been reported in many neurodegenerative disorders (NDDs), such as Alzheimer's disease (AD), Parkinson's disease (PD), and amyotrophic lateral sclerosis (ALS), among others, suggesting a possible role in disease pathogenesis. The gut microbiota is a pivotal component of the GBA, and alterations in its composition, known as gut dysbiosis, have been associated with GBA dysfunction and neurodegeneration. The gut microbiota might influence the homeostasis of the CNS by modulating the immune system and, more directly, regulating the production of molecules and metabolites that influence the nervous and endocrine systems, making it a potential therapeutic target. Preclinical trials manipulating microbial composition through dietary intervention, probiotic and prebiotic supplementation, and fecal microbial transplantation (FMT) have provided promising outcomes. However, its clear mechanism is not well understood, and the results are not always consistent. Here, we provide an overview of the major components and communication pathways of the GBA, as well as therapeutic approaches targeting the GBA to ameliorate NDDs.
Collapse
Affiliation(s)
- Yadong Zheng
- School of Biosciences and Veterinary Medicine, University of Camerino, 62032 Camerino, MC, Italy; (Y.Z.); (L.B.)
- School of Food and Biological Engineering, Zhengzhou University of Light Industry, Zhengzhou 450002, China
| | - Laura Bonfili
- School of Biosciences and Veterinary Medicine, University of Camerino, 62032 Camerino, MC, Italy; (Y.Z.); (L.B.)
| | - Tao Wei
- School of Food and Biological Engineering, Zhengzhou University of Light Industry, Zhengzhou 450002, China
| | - Anna Maria Eleuteri
- School of Biosciences and Veterinary Medicine, University of Camerino, 62032 Camerino, MC, Italy; (Y.Z.); (L.B.)
| |
Collapse
|
47
|
Sahn B, Pascuma K, Kohn N, Tracey KJ, Markowitz JF. Transcutaneous auricular vagus nerve stimulation attenuates inflammatory bowel disease in children: a proof-of-concept clinical trial. Bioelectron Med 2023; 9:23. [PMID: 37849000 PMCID: PMC10583463 DOI: 10.1186/s42234-023-00124-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Accepted: 09/11/2023] [Indexed: 10/19/2023] Open
Abstract
BACKGROUND Vagus nerve stimulation is an investigational anti-inflammatory therapy targeting the nervous system to modulate immune activity. This study evaluated the efficacy and safety of transcutaneous auricular VNS (ta-VNS) in patients with pediatric-onset Crohn's disease (CD) or ulcerative colitis (UC). METHODS Participants were 10-21 years of age with mild/moderate CD or UC and fecal calprotectin (FC) > 200 ug/g within 4 weeks of study entry. Subjects were randomized to receive either ta-VNS targeting the cymba conchae of the external left ear, or sham stimulation, of 5 min duration once daily for a 2-week period, followed by a cross over to the alternative stimulation for an additional 2 weeks. At week 4, all subjects received ta-VNS of 5 min duration twice daily until week 16. Primary study endpoints were clinical remission, and a ≥ 50% reduction in FC level from baseline to week 16. Heart rate variability measurements and patient-reported outcome questionnaires were completed during interval and week 16 assessments. RESULTS Twenty-two subjects were enrolled and analyzed (10 CD, 12 UC). Six of 10 with CD had a wPCDAI > 12.5 and 6/12 with UC had a PUCAI > 10 at baseline, correlating to mild to moderate symptom activity. Among the 12 subjects with active symptomatic disease indices at baseline, clinical remission was achieved in 3/6 (50%) with CD and 2/6 (33%) with UC at week 16. Despite all subjects having FC levels ≥ 200 within 4 weeks of enrollment, five subjects (4 UC, 1 CD) had FC levels < 200 at the baseline visit and were excluded from the FC analysis. Of the remaining 17, median baseline FC was 907 µg/g (IQR 411-2,120). At week 16, 11/17 (64.7%) of those with baseline FC ≥ 200 had a ≥ 50% reduction in FC (95% CI 38.3-85.8). In the UC subjects, there was an 81% median reduction in FC vs baseline (833 µg/g; p = 0.03) while in the CD subjects, median reduction in FC at 16 weeks was 51% (357 µg/g; p = 0.09). There were no safety concerns. CONCLUSION Noninvasive ta-VNS attenuated signs and symptoms in a pediatric cohort with mild to moderate inflammatory bowel disease. TRIAL REGISTRATION NCT03863704-Date of registration 3/4/2019.
Collapse
Affiliation(s)
- Benjamin Sahn
- Division of Pediatric Gastroenterology, Liver Diseases, & Nutrition, Steven & Alexandra Cohen Children's Medical Center, Northwell Health, 1991 Marcus Ave, Suite M100, New Hyde Park, NY, 11042, USA.
- Feinstein Institutes for Medical Research, Manhasset, NY, USA.
| | - Kristine Pascuma
- Division of Pediatric Gastroenterology, Liver Diseases, & Nutrition, Steven & Alexandra Cohen Children's Medical Center, Northwell Health, 1991 Marcus Ave, Suite M100, New Hyde Park, NY, 11042, USA
| | - Nina Kohn
- Feinstein Institutes for Medical Research, Manhasset, NY, USA
- Biostatistics Unit, Office of Academic Affairs, New Hyde Park, NY, USA
| | - Kevin J Tracey
- Feinstein Institutes for Medical Research, Manhasset, NY, USA
| | - James F Markowitz
- Division of Pediatric Gastroenterology, Liver Diseases, & Nutrition, Steven & Alexandra Cohen Children's Medical Center, Northwell Health, 1991 Marcus Ave, Suite M100, New Hyde Park, NY, 11042, USA
| |
Collapse
|
48
|
Yan Q, Chen J, Ren X, Song Y, Xu J, Xuan S, Jiang X, Kuang Z, Tang Z. Vagus Nerve Stimulation Relives Irritable Bowel Syndrome and the Associated Depression via α7nAChR-mediated Anti-inflammatory Pathway. Neuroscience 2023; 530:26-37. [PMID: 37625687 DOI: 10.1016/j.neuroscience.2023.08.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 08/10/2023] [Accepted: 08/18/2023] [Indexed: 08/27/2023]
Abstract
OBJECTIVES The present study is designed to investigate the role of vagus nerve in the treatments of irritable bowel syndrome (IBS) and the associated central nervous system disorders. METHODS An IBS animal model was established by giving acetic acid and chronic-acute stress (AA-CAS) treatment in adult male Wistar rats. Subdiaphragmatic vagotomy (SDV) and vagus nerve stimulation (VNS) were performed to intervene the excitability of vagus nerve. Permeability of blood brain barrier (BBB) was measured and agonist and antagonist of α7 nicotinic acetylcholine receptor (α7nAChR) were used to explore the relevant mechanisms. RESULTS AA-CAS treatment resulted in abnormal fecal output, increased visceral sensitivity, depressive-like behaviors, and overexpression of inflammatory mediators, all of which were reversed by VNS treatment. The effects of VNS could also be observed when α7nAChR agonist was applied. Whereas α7nAChR antagonist (methyllycaconitine, MLA) reversed VNS's effects. Interestingly, VNS also reduced the increased permeability of blood brain barrier (BBB) following AA-CAS treatment in IBS rats. SDV treatment only show temporary efficacy on AA-CAS-induced symptoms and had no effect on the permeability of BBB. CONCLUSION The intestinal abnormalities and depressive symptoms in IBS rats can be improved by VNS treatment. This positive effect of VNS was achieved through α7nAChR-mediated inflammatory pathway and may also be associated with the decreased of BBB permeability.
Collapse
Affiliation(s)
- Qizhi Yan
- Shaoxing People's Hospital, Shaoxing 312000, China
| | - Jiawei Chen
- Shaoxing People's Hospital, Shaoxing 312000, China
| | - Xiuying Ren
- Shaoxing People's Hospital, Shaoxing 312000, China
| | - Yibo Song
- Shaoxing People's Hospital, Shaoxing 312000, China
| | - Jian Xu
- Shaoxing People's Hospital, Shaoxing 312000, China
| | - Shaoyan Xuan
- Shaoxing People's Hospital, Shaoxing 312000, China
| | - Xi Jiang
- Zhejiang University Mingzhou Hospital, Ningbo 315000, China
| | - Zhijian Kuang
- Zhejiang University Mingzhou Hospital, Ningbo 315000, China
| | - Zhihua Tang
- Shaoxing People's Hospital, Shaoxing 312000, China.
| |
Collapse
|
49
|
Zhou Y, Xie L, Schröder J, Schuster IS, Nakai M, Sun G, Sun YBY, Mariño E, Degli-Esposti MA, Marques FZ, Grubman A, Polo JM, Mackay CR. Dietary Fiber and Microbiota Metabolite Receptors Enhance Cognition and Alleviate Disease in the 5xFAD Mouse Model of Alzheimer's Disease. J Neurosci 2023; 43:6460-6475. [PMID: 37596052 PMCID: PMC10506626 DOI: 10.1523/jneurosci.0724-23.2023] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 07/27/2023] [Accepted: 07/28/2023] [Indexed: 08/20/2023] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder with poorly understood etiology. AD has several similarities with other "Western lifestyle" inflammatory diseases, where the gut microbiome and immune pathways have been associated. Previously, we and others have noted the involvement of metabolite-sensing GPCRs and their ligands, short-chain fatty acids (SCFAs), in protection of numerous Western diseases in mouse models, such as Type I diabetes and hypertension. Depletion of GPR43, GPR41, or GPR109A accelerates disease, whereas high SCFA yielding diets protect in mouse models. Here, we extended the concept that metabolite-sensing receptors and SCFAs may be a more common protective mechanism against Western diseases by studying their role in AD pathogenesis in the 5xFAD mouse model. Both male and female mice were included. Depletion of GPR41 and GPR43 accelerated cognitive decline and impaired adult hippocampal neurogenesis in 5xFAD and WT mice. Lack of fiber/SCFAs accelerated a memory deficit, whereas diets supplemented with high acetate and butyrate (HAMSAB) delayed cognitive decline in 5xFAD mice. Fiber intake impacted on microglial morphology in WT mice and microglial clustering phenotype in 5xFAD mice. Lack of fiber impaired adult hippocampal neurogenesis in both W and AD mice. Finally, maternal dietary fiber intake significantly affects offspring's cognitive functions in 5xFAD mice and microglial transcriptome in both WT and 5xFAD mice, suggesting that SCFAs may exert their effect during pregnancy and lactation. Together, metabolite-sensing GPCRs and SCFAs are essential for protection against AD, and reveal a new strategy for disease prevention.Significance Statement Alzheimer's disease (AD) is one of the most common neurodegenerative diseases; currently, there is no cure for AD. In our study, short-chain fatty acids and metabolite receptors play an important role in cognitive function and pathology in AD mouse model as well as in WT mice. SCFAs also impact on microglia transcriptome, and immune cell recruitment. Out study indicates the potential of specialized diets (supplemented with high acetate and butyrate) releasing high amounts of SCFAs to protect against disease.
Collapse
Affiliation(s)
- Yichen Zhou
- Department of Microbiology, Monash University, Clayton, Victoria, Australia, 3800
| | - Liang Xie
- Department of Microbiology, Monash University, Clayton, Victoria, Australia, 3800
- Hypertension Research Laboratory, School of Biological Sciences, Monash University, Clayton, Victoria, Australia, 3800
| | - Jan Schröder
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia, 3800
- Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Clayton, Victoria, Australia, 3800
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia, 3800
| | - Iona S Schuster
- Department of Microbiology, Monash University, Clayton, Victoria, Australia, 3800
- Center for Experimental Immunology, Lions Eye Institute, Nedlands, Western Australia, Australia, 6009
| | - Michael Nakai
- Hypertension Research Laboratory, School of Biological Sciences, Monash University, Clayton, Victoria, Australia, 3800
| | - Guizhi Sun
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia, 3800
- Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Clayton, Victoria, Australia, 3800
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia, 3800
| | - Yu B Y Sun
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia, 3800
- Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Clayton, Victoria, Australia, 3800
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia, 3800
| | - Eliana Mariño
- Department of Biochemistry & Molecular Biology, Monash University, Clayton, Victoria, Australia, 3800
| | - Mariapia A Degli-Esposti
- Department of Microbiology, Monash University, Clayton, Victoria, Australia, 3800
- Center for Experimental Immunology, Lions Eye Institute, Nedlands, Western Australia, Australia, 6009
| | - Francine Z Marques
- Hypertension Research Laboratory, School of Biological Sciences, Monash University, Clayton, Victoria, Australia, 3800
- Heart Failure Research Laboratory, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia, 6009
| | - Alexandra Grubman
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia, 3800
- Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Clayton, Victoria, Australia, 3800
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia, 3800
| | - Jose M Polo
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia, 3800
- Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Clayton, Victoria, Australia, 3800
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia, 3800
| | - Charles R Mackay
- Department of Microbiology, Monash University, Clayton, Victoria, Australia, 3800
- School of Pharmaceutical Sciences, Shandong Analysis and Test Center, Qilu University of Technology (Shandong Academy of Sciences), Jinan, China, 6009
| |
Collapse
|
50
|
Chandra R, Sokratian A, Chavez KR, King S, Swain SM, Snyder JC, West AB, Liddle RA. Gut mucosal cells transfer α-synuclein to the vagus nerve. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.14.553305. [PMID: 37645945 PMCID: PMC10461984 DOI: 10.1101/2023.08.14.553305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/31/2023]
Abstract
Epidemiological and histopathological findings have raised the possibility that misfolded α-synuclein protein might spread from the gut to the brain and increase the risk of Parkinson's disease (PD). While past experimental studies in mouse models have relied on gut injections of exogenous recombinant α-synuclein fibrils to study gut to brain α-synuclein transfer, the possible origins of misfolded α-synuclein within the gut have remained elusive. We recently demonstrated that sensory cells of the gut mucosa express α-synuclein. In this study, we employed mouse intestinal organoids expressing human α-synuclein to observe the transfer of α-synuclein protein from gut epithelial cells in organoids co-cultured with vagal nodose neurons that are otherwise devoid of α-synuclein expression. In intact mice that express pathological human α-synuclein, but no mouse α-synuclein, α-synuclein fibril templating activity emerges in α-synuclein seeded fibril aggregation assays in tissues from the gut, vagus nerve, and dorsal motor nucleus. In newly engineered transgenic mice that restrict pathological human α-synuclein expression to intestinal epithelial cells, α-synuclein fibril-templating activity transfers to the vagus nerve and to the dorsal motor nucleus. Subdiaphragmatic vagotomy prior to the induction of α-synuclein expression in the gut epithelial cells effectively protects the hindbrain from the emergence of α-synuclein fibril templating activity. Overall, these findings highlight a novel potential non-neuronal source of fibrillar α-synuclein protein that might arise in gut mucosal cells.
Collapse
|