1
|
Feng J, Cen Q, Cui Y, Hu X, Li M, Wang L, Wei J, Sun N, Wang J, Zhang A. Lactobacillus rhamnosus: An emerging probiotic with therapeutic potential for depression. Pharmacol Res 2025; 211:107541. [PMID: 39653301 DOI: 10.1016/j.phrs.2024.107541] [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: 08/31/2024] [Revised: 11/18/2024] [Accepted: 12/06/2024] [Indexed: 12/16/2024]
Abstract
Depression, a complex psychological disorder, involves multiple biological pathways in its pathogenesis. In recent years, the gut-brain axis theory has provided novel insights into the pathogenesis of depression, particularly the crucial role of the gut microbiota in mood regulation. While there remains no universal consensus on the most efficacious strains for depression treatment, Lactobacillus rhamnosus has risen to prominence within the realm of probiotics for its potential to positively modulate depressive symptoms. This review preliminarily explores the clinical significance of Lactobacillus rhamnosus in the treatment of depression and summarizes the potential mechanisms by which Lactobacillus rhamnosus treats depression, including its regulation of gut microbiota, alterations in gene expression, improvement of intestinal barrier function, maintenance of neurotransmitter balance, suppression of inflammatory responses, modulation of the immune system, coping with oxidative stress, and optimization of metabolic processes. Future research needs to further explore these mechanisms and combine them with clinical research results to optimize treatment plans and provide more effective treatment options for patients with depression.
Collapse
Affiliation(s)
- Jing Feng
- Department of Rehabilitation Medicine, Shanghai Fourth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Qiuyu Cen
- Department of Rehabilitation Medicine, Shanghai Fourth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yanru Cui
- Department of Rehabilitation Medicine, Shanghai Fourth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xiaomin Hu
- Department of Rehabilitation Medicine, General Hospital of Western Theater Command, Chengdu, China
| | - Min Li
- Department of Rehabilitation Medicine, Shanghai Fourth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Linjie Wang
- Department of Rehabilitation Medicine, General Hospital of Western Theater Command, Chengdu, China
| | - Juanfang Wei
- College of Physical Education and Health, Geely University of China, Chengdu, China
| | - Nianyi Sun
- Department of Rehabilitation Medicine, Shanghai Fourth People's Hospital, Tongji University School of Medicine, Shanghai, China.
| | - Junyu Wang
- Department of Rehabilitation Medicine, Shanghai Fourth People's Hospital, Tongji University School of Medicine, Shanghai, China.
| | - Anren Zhang
- Department of Rehabilitation Medicine, Shanghai Fourth People's Hospital, Tongji University School of Medicine, Shanghai, China.
| |
Collapse
|
2
|
He Q, Liu Q, Liang B, Zhao X, Huang W, Gao S, Chen C, Zhou F, Wang Z. Esketamine use for primary intelligent analgesia in adults with severe burns: A double-blind randomized trial with effects on analgesic efficacy, gastrointestinal function and mental state. Burns 2024; 50:107187. [PMID: 39317541 DOI: 10.1016/j.burns.2024.06.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 05/02/2024] [Accepted: 06/13/2024] [Indexed: 09/26/2024]
Abstract
BACKGROUND Opioid consumption for analgesia in burn patients is enormous. Non-opioid analgesics for burn pain management may result in opioid sparing, reducing opioid-related adverse reactions and drug tolerance or addiction. METHODS A dual-center, randomized controlled trial assessed Esketamine for the perioperative period in patients with severe [20-50 % total body surface area (TBSA)] and extensive (≥ 50 % TBSA) burns, comparing analgesia with standard anesthesia. Sixty patients were randomly allocated (1:1 ratio) to two arms. In the Treatment Arm, patients received intra-operative Esketamine and postoperative intravenous primary intelligent analgesia pump with Esketamine. Patients in the Control Arm received the same intervention as Treatment Arm without Esketamine. The primary endpoint was subjective analgesic efficacy (SAE) evaluated on Day 28 or the day before hospital discharge. Secondary outcomes included the postoperative Numeric Pain Rating (NPR) Scale at rest (NPRr) and during movement (NPRm) and opioid consumption. Gastrointestinal dysfunction Scores (GIDS) and serum markers of intestinal injury [intestinal fatty acid-binding protein 2 (iFabp2) and apolipoproteinA2 (ApoA2)] were measured in the 1st and 4th post-injury weeks. Depression and sleep quality were assessed by relevant questionnaires. RESULTS Fifty-five patients were included in the analysis. Esketamine-treated Arm recorded a better analgesic efficacy than the Control Arm (proportion of patients with Grade 1 or 2 SAE scores, 67.9 % vs. 40.7 %, p = 0.022). Esketamine-treated patients had lower NPRm values (p = 0.033) and lower daily opioid consumption (p = 0.033) when compared with Controls. Esketamine-treated patients showed comparable gastrointestinal recovery to those in the Control Arm. The overall sleep quality might be improved in the Treatment Arm. CONCLUSIONS Esketamine use is safe for perioperative primary intelligent analgesia of severe burns, resulting in improved resting pain control and lower opioid requirements. TRIAL REGISTRATION The trial was registered at the Chinese Clinical Trial Registry (www.chictr.org.cn/) (ChiCTR2000034069).
Collapse
Affiliation(s)
- Qiulan He
- Department of Anesthesiology, First Affiliated Hospital, Sun Yat-sen University, No. 58, Zhongshan 2nd Road, Yuexiu District, Guangzhou City, Guangdong Province, China.
| | - Qi Liu
- Department of Anesthesiology, First Affiliated Hospital, Sun Yat-sen University, No. 58, Zhongshan 2nd Road, Yuexiu District, Guangzhou City, Guangdong Province, China.
| | - Bing Liang
- Department of Anesthesiology, Guangzhou Red Cross Hospital, No. 396 Tongfu Middle Road, Haizhu District, Guangzhou City, Guangdong Province, China.
| | - Xu Zhao
- Department of Anesthesiology, First Affiliated Hospital, Sun Yat-sen University, No. 58, Zhongshan 2nd Road, Yuexiu District, Guangzhou City, Guangdong Province, China.
| | - Wenqi Huang
- Department of Anesthesiology, First Affiliated Hospital, Sun Yat-sen University, No. 58, Zhongshan 2nd Road, Yuexiu District, Guangzhou City, Guangdong Province, China.
| | - Shaowei Gao
- Department of Anesthesiology, First Affiliated Hospital, Sun Yat-sen University, No. 58, Zhongshan 2nd Road, Yuexiu District, Guangzhou City, Guangdong Province, China.
| | - Caiyun Chen
- Department of Anesthesiology, First Affiliated Hospital, Sun Yat-sen University, No. 58, Zhongshan 2nd Road, Yuexiu District, Guangzhou City, Guangdong Province, China.
| | - Fei Zhou
- Department of Burn Surgery, First Affiliated Hospital, Sun Yat-sen University, No. 396 Tongfu Middle Road, Haizhu District, Guangzhou City, Guangdong Province, China.
| | - Zhongxing Wang
- Department of Anesthesiology, First Affiliated Hospital, Sun Yat-sen University, No. 58, Zhongshan 2nd Road, Yuexiu District, Guangzhou City, Guangdong Province, China.
| |
Collapse
|
3
|
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
|
4
|
Li X, He E, Chen G, Cao X, Zhao L, Xu X, Fu Z, Qiu H. Intergenerational neurotoxicity of polystyrene nanoplastics in offspring mice is mediated by dysfunctional microbe-gut-brain axis. ENVIRONMENT INTERNATIONAL 2024; 192:109026. [PMID: 39321539 DOI: 10.1016/j.envint.2024.109026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 08/28/2024] [Accepted: 09/19/2024] [Indexed: 09/27/2024]
Abstract
Nanoplastics (NPs) are ubiquitous in daily life, posing potential risks to the environment and human. While their negative effects on parental organisms have been extensively studied, intergenerational effects are still in the early stages of investigation. Here, we aimed to investigate the impact of maternal exposure to an environmentally relevant level of polystyrene NPs (PSNPs, 100 nm) during gestation and lactation (∼32 days, 50 μg/mouse/day) on neurotoxicity mediated by the microbe-gut-brain axis in offspring mice. Maternal PSNPs exposure significantly increased brain TNF-α level and microglia by 1.43 and 1.48 folds respectively, compared to control, accompanied by nuclear pyknosis and cell vacuolization in cortex and hippocampus. Targeted neurotransmitter metabolomics analysis revealed dysregulation in dopamine and serotonin metabolism. Specifically, dopamine levels increased significantly from 0.007 ng/L to 0.015 ng/L, while N-acetylseroton and 3,4-dihydroxyphenylacetic acid decreased significantly from 0.002 and 0.929 ng/L to 0.001 and 0.680 ng/L, respectively. Through a combination of 16S rRNA sequencing and biochemical analysis, we discovered that maternal PSNPs exposure led to a depletion of anti-inflammatory bacteria and an enrichment of pro-inflammatory bacteria resulting in intestinal barrier damage, elevated levels of lipopolysaccharide in blood, and subsequent activation of neuroinflammation. Meanwhile, gut bacteria dysbiosis interfered with communication between gut and brain by dysregulating neurotransmitter synthesis, as evidenced by significant associations between neurotransmitter-related bacteria (Akkermansia, Family_XIII_AD3011_group, Lachnoclostridium) and dopamine/serotonin related metabolites. Furthermore, transcriptional alterations in dopamine and serotonin related pathways were observed in the enteric nervous system, suggesting abnormal signal transduction from gut to brain contributes to neurotoxicity. This study provides new insights into NPs-induced neurotoxicity within the context of microbe-gut-brain axis and highlights the risk of cerebral dysfunction in offspring with maternal NPs exposure.
Collapse
Affiliation(s)
- Xing Li
- School of Environmental Science and Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Erkai He
- School of Geographic Sciences, East China Normal University, Shanghai 200241, China
| | - Guangquan Chen
- Department of Obstetrics and Gynecology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai 201204, China
| | - Xinde Cao
- School of Environmental Science and Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Ling Zhao
- School of Environmental Science and Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Xiaoyun Xu
- School of Environmental Science and Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Zhuozhong Fu
- School of Geographic Sciences, East China Normal University, Shanghai 200241, China
| | - Hao Qiu
- School of Environmental Science and Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China.
| |
Collapse
|
5
|
Qu S, Yu Z, Zhou Y, Wang S, Jia M, Chen T, Zhang X. Gut microbiota modulates neurotransmitter and gut-brain signaling. Microbiol Res 2024; 287:127858. [PMID: 39106786 DOI: 10.1016/j.micres.2024.127858] [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: 02/08/2024] [Revised: 06/16/2024] [Accepted: 07/22/2024] [Indexed: 08/09/2024]
Abstract
Neurotransmitters, including 5-hydroxytryptamine (5-HT), dopamine (DA), gamma-aminobutyric acid (GABA), and glutamate, are essential transductors in the Gut-Brain Axis (GBA), playing critical roles both peripherally and centrally. Accumulating evidence suggests that the gut microbiota modulates intestinal neurotransmitter metabolism and gut-to-brain signaling, shedding light on the crucial role of the gut microbiota in brain function and the pathogenesis of various neuropsychiatric diseases, such as major depression disorder (MDD), anxiety, addiction and Parkinson's disease (PD). Despite the exciting findings, the mechanisms underlying the modulation of neurotransmitter metabolism and function by the gut microbiota are still being elucidated. In this review, we aim to provide a comprehensive overview of the existing knowledge about the role of the gut microbiota in neurotransmitter metabolism and function in animal and clinical experiments. Moreover, we will discuss the potential mechanisms through which gut microbiota-derived neurotransmitters contribute to the pathogenesis of neuropsychiatric diseases, thus highlighting a novel therapeutic target for these conditions.
Collapse
Affiliation(s)
- Shiyan Qu
- Department of Psychiatry, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410000, China; National Clinic Research Center for Mental Disorders, Changsha, Hunan 410000, China; National Technology Institute on Mental Disorders, Changsha, Hunan 410000, China; Hunan Key Laboratory of Psychiatry and Mental Health, Changsha, Hunan 410000, China; Mental Health Institute, Second Xiangya Hospital, Central South University, Changsha 410000, China
| | - Zijin Yu
- Department of Psychiatry, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410000, China; National Clinic Research Center for Mental Disorders, Changsha, Hunan 410000, China; National Technology Institute on Mental Disorders, Changsha, Hunan 410000, China; Hunan Key Laboratory of Psychiatry and Mental Health, Changsha, Hunan 410000, China; Mental Health Institute, Second Xiangya Hospital, Central South University, Changsha 410000, China
| | - Yaxuan Zhou
- Department of Psychiatry, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410000, China; National Clinic Research Center for Mental Disorders, Changsha, Hunan 410000, China; National Technology Institute on Mental Disorders, Changsha, Hunan 410000, China; Hunan Key Laboratory of Psychiatry and Mental Health, Changsha, Hunan 410000, China; Mental Health Institute, Second Xiangya Hospital, Central South University, Changsha 410000, China
| | - Shiyi Wang
- Department of Psychiatry, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410000, China; National Clinic Research Center for Mental Disorders, Changsha, Hunan 410000, China; National Technology Institute on Mental Disorders, Changsha, Hunan 410000, China; Hunan Key Laboratory of Psychiatry and Mental Health, Changsha, Hunan 410000, China; Mental Health Institute, Second Xiangya Hospital, Central South University, Changsha 410000, China
| | - Minqi Jia
- Department of Psychiatry, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410000, China; National Clinic Research Center for Mental Disorders, Changsha, Hunan 410000, China; National Technology Institute on Mental Disorders, Changsha, Hunan 410000, China; Hunan Key Laboratory of Psychiatry and Mental Health, Changsha, Hunan 410000, China; Mental Health Institute, Second Xiangya Hospital, Central South University, Changsha 410000, China
| | - Ti Chen
- Clinical Laboratory, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410000, China
| | - Xiaojie Zhang
- Department of Psychiatry, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410000, China; National Clinic Research Center for Mental Disorders, Changsha, Hunan 410000, China; National Technology Institute on Mental Disorders, Changsha, Hunan 410000, China; Hunan Key Laboratory of Psychiatry and Mental Health, Changsha, Hunan 410000, China; Mental Health Institute, Second Xiangya Hospital, Central South University, Changsha 410000, China.
| |
Collapse
|
6
|
Vashishth S, Ambasta RK, Kumar P. Deciphering the microbial map and its implications in the therapeutics of neurodegenerative disorder. Ageing Res Rev 2024; 100:102466. [PMID: 39197710 DOI: 10.1016/j.arr.2024.102466] [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: 07/31/2024] [Revised: 08/19/2024] [Accepted: 08/19/2024] [Indexed: 09/01/2024]
Abstract
Every facet of biological anthropology, including development, ageing, diseases, and even health maintenance, is influenced by gut microbiota's significant genetic and metabolic capabilities. With current advancements in sequencing technology and with new culture-independent approaches, researchers can surpass older correlative studies and develop mechanism-based studies on microbiome-host interactions. The microbiota-gut-brain axis (MGBA) regulates glial functioning, making it a possible target for the improvement of development and advancement of treatments for neurodegenerative diseases (NDDs). The gut-brain axis (GBA) is accountable for the reciprocal communication between the gastrointestinal and central nervous system, which plays an essential role in the regulation of physiological processes like controlling hunger, metabolism, and various gastrointestinal functions. Lately, studies have discovered the function of the gut microbiome for brain health-different microbiota through different pathways such as immunological, neurological and metabolic pathways. Additionally, we review the involvement of the neurotransmitters and the gut hormones related to gut microbiota. We also explore the MGBA in neurodegenerative disorders by focusing on metabolites. Further, targeting the blood-brain barrier (BBB), intestinal barrier, meninges, and peripheral immune system is investigated. Lastly, we discuss the therapeutics approach and evaluate the pre-clinical and clinical trial data regarding using prebiotics, probiotics, paraprobiotics, fecal microbiota transplantation, personalised medicine, and natural food bioactive in NDDs. A comprehensive study of the GBA will felicitate the creation of efficient therapeutic approaches for treating different NDDs.
Collapse
Affiliation(s)
- Shrutikirti Vashishth
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Delhi Technological University (Formerly DCE), Delhi 110042, India
| | - Rashmi K Ambasta
- Department of Medicine, School of Medicine, VUMC, Vanderbilt University, TN, USA
| | - Pravir Kumar
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Delhi Technological University (Formerly DCE), Delhi 110042, India.
| |
Collapse
|
7
|
Filippini F, Galli T. Unveiling defects of secretion mechanisms in Parkinson's disease. J Biol Chem 2024; 300:107603. [PMID: 39059489 PMCID: PMC11378209 DOI: 10.1016/j.jbc.2024.107603] [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: 01/30/2024] [Revised: 07/11/2024] [Accepted: 07/15/2024] [Indexed: 07/28/2024] Open
Abstract
Neurodegenerative diseases are characterized by progressive dysfunction and loss of specific sets of neurons. While extensive research has focused on elucidating the genetic and epigenetic factors and molecular mechanisms underlying these disorders, emerging evidence highlights the critical role of secretion in the pathogenesis, possibly even onset, and progression of neurodegenerative diseases, suggesting the occurrence of non-cell-autonomous mechanisms. Secretion is a fundamental process that regulates intercellular communication, supports cellular homeostasis, and orchestrates various physiological functions in the body. Defective secretion can impair the release of neurotransmitters and other signaling molecules, disrupting synaptic transmission and compromising neuronal survival. It can also contribute to the accumulation, misfolding, and aggregation of disease-associated proteins, leading to neurotoxicity and neuronal dysfunction. In this review, we discuss the implications of defective secretion in the context of Parkinson's disease, emphasizing its role in protein aggregation, synaptic dysfunction, extracellular vesicle secretion, and neuroinflammation. We propose a multiple-hit model whereby protein accumulation and secretory defects must be combined for the onset and progression of the disease.
Collapse
Affiliation(s)
- Francesca Filippini
- Department of Cell Biology, Yale University School of Medicine, New Haven, Connecticut, USA; Department of Neuroscience, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Thierry Galli
- Université Paris Cité, Institute of Psychiatry and Neuroscience of Paris, INSERM U1266, Membrane Traffic in Healthy & Diseased Brain, Paris, France; Groupe Hospitalier Universitaire Paris Psychiatrie & Neurosciences, Paris, France.
| |
Collapse
|
8
|
Wang X, Wang J, Han R, Yu C, Shen F. Neural circuit mechanisms of acupuncture effect: where are we now? Front Neurol 2024; 15:1399925. [PMID: 38938783 PMCID: PMC11208484 DOI: 10.3389/fneur.2024.1399925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 05/24/2024] [Indexed: 06/29/2024] Open
Abstract
Recently, there has been increasing attention on the impact of acupuncture on the dysregulated neural circuits in different disease. This has led to new understandings of how acupuncture works. This review presents a comprehensive analysis of research that have examined the impact of acupuncture on abnormal neural circuits associated with pain, anxiety, Parkinson's disease, addiction disorders, cognitive problems, and gastrointestinal disorders. These studies have shown that acupuncture's therapeutic effects are mediated by specific brain areas and neurons involved in neural circuit mechanisms, emphasising its wide-ranging influence. The positive impacts of acupuncture can be ascribed to its ability to modify the functioning of neurocircuits in various physiological conditions. Nevertheless, contemporary studies on acupuncture neural circuits frequently overlook the comprehensive circuit mechanism including the periphery, central nervous system, and target organ. Additionally, the scope of diseases studied is restricted. Future study should focus on broadening the range of diseases studied and exploring the neural circuit mechanisms of these diseases in depth in order to enhance our understanding of acupuncture's neurobiological impacts.
Collapse
Affiliation(s)
- Xuesong Wang
- College of Acupuncture-Moxibustion and Tuina, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Jia Wang
- Department of Acupuncture and Moxibustion, Wuhan Hospital of Integrated Traditional Chinese and Western Medicine, Wuhan, China
| | - Rui Han
- Department of Child Rehabilitation Medicine, Qujing Hospital of Maternity and Childcare, Qujing, China
| | - Chaochao Yu
- Department of Tuina, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, China
- The Fourth Clinical College of Guangzhou University of Chinese Medicine, Shenzhen, China
- Department of Rehabilitation, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Feng Shen
- College of Acupuncture and Orthopedics, Hubei University of Chinese Medicine, Wuhan, Hubei, China
| |
Collapse
|
9
|
D'Aquila PS. Dopamine, activation of ingestion and evaluation of response efficacy: a focus on the within-session time-course of licking burst number. Psychopharmacology (Berl) 2024; 241:1111-1124. [PMID: 38702473 PMCID: PMC11106101 DOI: 10.1007/s00213-024-06600-1] [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: 02/07/2024] [Accepted: 04/24/2024] [Indexed: 05/06/2024]
Abstract
RATIONALE Evidence on the effect of dopamine D1-like and D2-like receptor antagonists on licking microstructure and the forced swimming response led us to suggest that (i) dopamine on D1-like receptors plays a role in activating reward-directed responses and (ii) the level of response activation is reboosted based on a process of evaluation of response efficacy requiring dopamine on D2-like receptors. A main piece of evidence in support of this hypothesis is the observation that the dopamine D2-like receptor antagonist raclopride induces a within-session decrement of burst number occurring after the contact with the reward. The few published studies with a detailed analysis of the time-course of this measure were conducted in our laboratory. OBJECTIVES The aim of this review is to recapitulate and discuss the evidence in support of the analysis of the within-session burst number as a behavioural substrate for the study of the mechanisms governing ingestion, behavioural activation and the related evaluation processes, and its relevance in the analysis of drug effects on ingestion. CONCLUSIONS The evidence gathered so far suggests that the analysis of the within-session time-course of burst number provides an important behavioural substrate for the study of the mechanisms governing ingestion, behavioural activation and the related evaluation processes, and might provide decisive evidence in the analysis of the effects of drugs on ingestion. However, further evidence from independent sources is necessary to validate the use and the proposed interpretation of this measure.
Collapse
Affiliation(s)
- Paolo S D'Aquila
- Dipartimento di Scienze Biomediche, Università di Sassari, Viale S. Pietro 43/b, Sassari, 07100, Italy.
| |
Collapse
|
10
|
Sun H, Li C, Shi Y, Wang Y, Li J, Fan L, Yu Y, Ji X, Gao X, Hou K, Li Y. Investigating the L-Glu-NMDA receptor-H 2S-NMDA receptor pathway that regulates gastric function in rats' nucleus ambiguus. Front Pharmacol 2024; 15:1389873. [PMID: 38751777 PMCID: PMC11094298 DOI: 10.3389/fphar.2024.1389873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Accepted: 04/15/2024] [Indexed: 05/18/2024] Open
Abstract
Background In previous investigations, we explored the regulation of gastric function by hydrogen sulfide (H2S) and L-glutamate (L-Glu) injections in the nucleus ambiguus (NA). We also determined that both H2S and L-Glu have roles to play in the physiological activities of the body, and that NA is an important nucleus for receiving visceral sensations. The purpose of this study was to explore the potential pathway link between L-Glu and H2S, resulting in the regulation of gastric function. Methods Physiological saline (PS), L-glutamate (L-Glu, 2 nmol), NaHS (2 nmol), D-2-amino-5-phopho-novalerate (D-AP5, 2 nmol) + L-Glu (2 nmol), aminooxyacetic acid (AOAA, 2 nmol) + L-Glu (2 nmol), D-AP5 (2 nmol) + NaHS (2 nmol) were injected into the NA. A balloon was inserted into the stomach to observe gastric pressure and for recording the changes of gastric smooth muscle contraction curve. The gastric fluid was collected by esophageal perfusion and for recording the change of gastric pH value. Results Injecting L-Glu in NA was found to significantly inhibit gastric motility and promote gastric acid secretion in rats (p < 0.01). On the other hand, injecting the PS, pre-injection N-methyl-D-aspartate (NMDA) receptor blocker D-AP5, cystathionine beta-synthase (CBS) inhibitor AOAA and re-injection L-Glu did not result in significant changes (p > 0.05). The same injection NaHS significantly inhibit gastric motility and promote gastric acid secretion in rats (p < 0.01), but is eliminated by injection D-AP5 (p > 0.05). Conclusion The results indicate that both exogenous L-Glu and H2S injected in NA regulate gastric motility and gastric acid secretion through NMDA receptors. This suggests that NA has an L-Glu-NMDA receptor-CBS-H2S pathway that regulates gastric function.
Collapse
|
11
|
Zong J, Yang Y, Wang H, Zhang H, Yang X, Yang X. The two-directional prospective association between inflammatory bowel disease and neurodegenerative disorders: a systematic review and meta-analysis based on longitudinal studies. Front Immunol 2024; 15:1325908. [PMID: 38720896 PMCID: PMC11076839 DOI: 10.3389/fimmu.2024.1325908] [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: 10/22/2023] [Accepted: 04/10/2024] [Indexed: 05/12/2024] Open
Abstract
Objective Previous studies reported possible connections between inflammatory bowel disease (IBD) and several neurodegenerative disorders. However, the comprehensive relationships between IBD and various neurodegenerative disorders were not summarized. We executed a meta-analysis of longitudinal studies to provide an estimate of the strength of the two-directional prospective association between IBD and neurodegenerative disorders. Methods We accomplished a thorough bibliographic search of PubMed, Web of Science, Embase, PsycINFO, and Cochrane Library databases until June 2023 to locate relevant longitudinal studies. The extracted data were then analyzed via meta-analysis using either a fixed or random effects model. Results The final analysis encompassed 27 studies. Individuals with IBD faced an increased risk of developing four neurodegenerative disorders than the general public, namely, Alzheimer's disease (hazard ratio[HR] = 1.35, 95% confidence interval [CI]: 1.03-1.77, P=0.031), dementia (HR =1.24, 95% CI: 1.13-1.36, P<0.001), multiple sclerosis (HR =2.07, 95% CI:1.42-3.02, P<0.001) and Parkinson's disease (HR =1.23, 95% CI:1.10-1.38, P<0.001). Two articles reported an increased incidence of amyotrophic lateral sclerosis or multiple system atrophy in IBD patients. Three studies investigated the prospective association between multiple sclerosis and IBD, revealing an elevated risk of the latter in patients with the former. (HR=1.87, 95% CI:1.66-2.10, P<0.001). Interpretation These findings verified the two-directional relationship between the brain-gut axis, specifically demonstrating a heightened risk of various neurodegenerative diseases among IBD patients. It may be profitable to prepare screening strategies for IBD patients to find neurodegenerative diseases during the long-term course of treatment for IBD with a view to potential earlier diagnosis and treatment of neurodegenerative diseases, reducing public health and social burden. Systematic Review Registration PROSPERO (CRD42023437553).
Collapse
Affiliation(s)
- Jiahao Zong
- Department of Gastroenterology, Qilu Hospital of Shandong University, Jinan, China
- Shandong Provincial Clinical Research Center for Digestive Disease, Shandong, China
- Laboratory of Translational Gastroenterology, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Yue Yang
- Department of General Surgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hui Wang
- Department of Gastroenterology, Qilu Hospital of Shandong University, Jinan, China
- Shandong Provincial Clinical Research Center for Digestive Disease, Shandong, China
- Laboratory of Translational Gastroenterology, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Huipeng Zhang
- Department of Gastroenterology, Qilu Hospital of Shandong University, Jinan, China
- Shandong Provincial Clinical Research Center for Digestive Disease, Shandong, China
- Laboratory of Translational Gastroenterology, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Xiaorong Yang
- Department of Gastroenterology, Qilu Hospital of Shandong University, Jinan, China
- Shandong Provincial Clinical Research Center for Digestive Disease, Shandong, China
- Laboratory of Translational Gastroenterology, Qilu Hospital of Shandong University, Jinan, Shandong, China
- Clinical Epidemiology Unit, Clinical Research Center of Shandong University, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Xiaoyun Yang
- Department of Gastroenterology, Qilu Hospital of Shandong University, Jinan, China
- Shandong Provincial Clinical Research Center for Digestive Disease, Shandong, China
- Laboratory of Translational Gastroenterology, Qilu Hospital of Shandong University, Jinan, Shandong, China
- Clinical Epidemiology Unit, Clinical Research Center of Shandong University, Qilu Hospital of Shandong University, Jinan, Shandong, China
| |
Collapse
|
12
|
Liu J, Zhang S, Emadi S, Guo T, Chen L, Feng B. Morphological, molecular, and functional characterization of mouse glutamatergic myenteric neurons. Am J Physiol Gastrointest Liver Physiol 2024; 326:G279-G290. [PMID: 38193160 PMCID: PMC11211033 DOI: 10.1152/ajpgi.00200.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 12/26/2023] [Accepted: 01/02/2024] [Indexed: 01/10/2024]
Abstract
The enteric nervous system (ENS) functions largely independently of the central nervous system (CNS). Glutamate, the dominant neurotransmitter in the CNS and sensory afferents, is not a primary neurotransmitter in the ENS. Only a fraction (∼2%) of myenteric neurons in the mouse distal colon and rectum (colorectum) are positive for vesicular glutamate transporter type 2 (VGLUT2), the structure and function of which remain undetermined. Here, we systematically characterized VGLUT2-positive enteric neurons (VGLUT2-ENs) through sparse labeling with adeno-associated virus, single-cell mRNA sequencing (scRNA-seq), and GCaMP6f calcium imaging. Our results reveal that the majority of VGLUT2-ENs (29 of 31, 93.5%) exhibited Dogiel type I morphology with a single aborally projecting axon; most axons (26 of 29, 89.7%) are between 4 and 10 mm long, each traversing 19 to 34 myenteric ganglia. These anatomical features exclude the VGLUT2-ENs from being intrinsic primary afferent or motor neurons. The scRNA-seq conducted on 52 VGLUT2-ENs suggests different expression profiles from conventional descending interneurons. Ex vivo GCaMP6f recordings from flattened colorectum indicate that almost all VGLUT2-EN (181 of 215, 84.2%) are indirectly activated by colorectal stretch via nicotinic cholinergic neural transmission. In conclusion, VGLUT2-ENs are a functionally unique group of enteric neurons with single aborally projecting long axons that traverse multiple myenteric ganglia and are activated indirectly by colorectal mechanical stretch. This knowledge will provide a solid foundation for subsequent studies on the potential interactions of VGLUT2-EN with extrinsic colorectal afferents via glutamatergic neurotransmission.NEW & NOTEWORTHY We reveal that VGLUT2-positive enteric neurons (EN), although constituting a small fraction of total EN, are homogeneously expressed in the myenteric ganglia, with a slight concentration at the intermediate region between the colon and rectum. Through anatomic, molecular, and functional analyses, we demonstrated that VGLUT2-ENs are activated indirectly by noxious circumferential colorectal stretch via nicotinic cholinergic transmission, suggesting their participation in mechanical visceral nociception.
Collapse
Affiliation(s)
- Jia Liu
- Department of Biomedical Engineering, University of Connecticut, Storrs, Connecticut, United States
| | - Shaopeng Zhang
- Department of Biomedical Engineering, University of Connecticut, Storrs, Connecticut, United States
| | - Sharareh Emadi
- Department of Biomedical Engineering, University of Connecticut, Storrs, Connecticut, United States
| | - Tiantian Guo
- Department of Biomedical Engineering, University of Connecticut, Storrs, Connecticut, United States
| | - Longtu Chen
- Department of Biomedical Engineering, University of Connecticut, Storrs, Connecticut, United States
| | - Bin Feng
- Department of Biomedical Engineering, University of Connecticut, Storrs, Connecticut, United States
| |
Collapse
|
13
|
Wu X, Xiao X, Fang H, He C, Wang H, Wang M, Lan P, Wang F, Du Q, Yang H. Elucidating shared biomarkers in gastroesophageal reflux disease and idiopathic pulmonary fibrosis: insights into novel therapeutic targets and the role of angelicae sinensis radix. Front Pharmacol 2024; 15:1348708. [PMID: 38414734 PMCID: PMC10897002 DOI: 10.3389/fphar.2024.1348708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Accepted: 01/31/2024] [Indexed: 02/29/2024] Open
Abstract
Background: The etiological underpinnings of gastroesophageal reflux disease (GERD) and idiopathic pulmonary fibrosis (IPF) remain elusive, coupled with a scarcity of effective therapeutic interventions for IPF. Angelicae sinensis radix (ASR, also named Danggui) is a Chinese herb with potential anti-fibrotic properties, that holds promise as a therapeutic agent for IPF. Objective: This study seeks to elucidate the causal interplay and potential mechanisms underlying the coexistence of GERD and IPF. Furthermore, it aims to investigate the regulatory effect of ASR on this complex relationship. Methods: A two-sample Mendelian randomization (TSMR) approach was employed to delineate the causal connection between gastroesophageal reflux disease and IPF, with Phennoscanner V2 employed to mitigate confounding factors. Utilizing single nucleotide polymorphism (SNPs) and publicly available microarray data, we analyzed potential targets and mechanisms related to IPF in GERD. Network pharmacology and molecular docking were employed to explore the targets and efficacy of ASR in treating GERD-related IPF. External datasets were subsequently utilized to identify potential diagnostic biomarkers for GERD-related IPF. Results: The IVW analysis demonstrated a positive causal relationship between GERD and IPF (IVW: OR = 1.002, 95%CI: 1.001, 1.003; p < 0.001). Twenty-five shared differentially expressed genes (DEGs) were identified. GO functional analysis revealed enrichment in neural, cellular, and brain development processes, concentrated in chromosomes and plasma membranes, with protein binding and activation involvement. KEGG analysis unveiled enrichment in proteoglycan, ERBB, and neuroactive ligand-receptor interaction pathways in cancer. Protein-protein interaction (PPI) analysis identified seven hub genes. Network pharmacology analysis demonstrated that 104 components of ASR targeted five hub genes (PDE4B, DRD2, ERBB4, ESR1, GRM8), with molecular docking confirming their excellent binding efficiency. GRM8 and ESR1 emerged as potential diagnostic biomarkers for GERD-related IPF (ESR1: AUCGERD = 0.762, AUCIPF = 0.725; GRM8: AUCGERD = 0.717, AUCIPF = 0.908). GRM8 and ESR1 emerged as potential diagnostic biomarkers for GERD-related IPF, validated in external datasets. Conclusion: This study establishes a causal link between GERD and IPF, identifying five key targets and two potential diagnostic biomarkers for GERD-related IPF. ASR exhibits intervention efficacy and favorable binding characteristics, positioning it as a promising candidate for treating GERD-related IPF. The potential regulatory mechanisms may involve cell responses to fibroblast growth factor stimulation and steroidal hormone-mediated signaling pathways.
Collapse
Affiliation(s)
- Xuanyu Wu
- Hospital of Chengdu University of Traditional Chinese Medicine, School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiang Xiao
- Hospital of Chengdu University of Traditional Chinese Medicine, School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Hanyu Fang
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
- Department of Traditional Chinese Medicine for Pulmonary Diseases, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China
| | - Cuifang He
- Hospital of Chengdu University of Traditional Chinese Medicine, School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Hanyue Wang
- Hospital of Chengdu University of Traditional Chinese Medicine, School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Miao Wang
- Hospital of Chengdu University of Traditional Chinese Medicine, School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Peishu Lan
- Hospital of Chengdu University of Traditional Chinese Medicine, School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Fei Wang
- Hospital of Chengdu University of Traditional Chinese Medicine, School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Quanyu Du
- Hospital of Chengdu University of Traditional Chinese Medicine, School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Han Yang
- Hospital of Chengdu University of Traditional Chinese Medicine, School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
14
|
Gupta S, Dinesh S, Sharma S. Bridging the Mind and Gut: Uncovering the Intricacies of Neurotransmitters, Neuropeptides, and their Influence on Neuropsychiatric Disorders. Cent Nerv Syst Agents Med Chem 2024; 24:2-21. [PMID: 38265387 DOI: 10.2174/0118715249271548231115071021] [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: 07/11/2023] [Revised: 08/31/2023] [Accepted: 10/04/2023] [Indexed: 01/25/2024]
Abstract
BACKGROUND The gut-brain axis (GBA) is a bidirectional signaling channel that facilitates communication between the gastrointestinal tract and the brain. Recent research on the gut-brain axis demonstrates that this connection enables the brain to influence gut function, which in turn influences the brain and its cognitive functioning. It is well established that malfunctioning of this axis adversely affects both systems' ability to operate effectively. OBJECTIVE Dysfunctions in the GBA have been associated with disorders of gut motility and permeability, intestinal inflammation, indigestion, constipation, diarrhea, IBS, and IBD, as well as neuropsychiatric and neurodegenerative disorders like depression, anxiety, schizophrenia, autism, Alzheimer's, and Parkinson's disease. Multiple research initiatives have shown that the gut microbiota, in particular, plays a crucial role in the GBA by participating in the regulation of a number of key neurochemicals that are known to have significant effects on the mental and physical well-being of an individual. METHODS Several studies have investigated the relationship between neuropsychiatric disorders and imbalances or disturbances in the metabolism of neurochemicals, often leading to concomitant gastrointestinal issues and modifications in gut flora composition. The interaction between neurological diseases and gut microbiota has been a focal point within this research. The novel therapeutic interventions in neuropsychiatric conditions involving interventions such as probiotics, prebiotics, and dietary modifications are outlined in this review. RESULTS The findings of multiple studies carried out on mice show that modulating and monitoring gut microbiota can help treat symptoms of such diseases, which raises the possibility of the use of probiotics, prebiotics, and even dietary changes as part of a new treatment strategy for neuropsychiatric disorders and their symptoms. CONCLUSION The bidirectional communication between the gut and the brain through the gut-brain axis has revealed profound implications for both gastrointestinal and neurological health. Malfunctions in this axis have been connected to a range of disorders affecting gut function as well as cognitive and neuropsychiatric well-being. The emerging understanding of the role of gut microbiota in regulating key neurochemicals opens up possibilities for novel treatment approaches for conditions like depression, anxiety, and neurodegenerative diseases.
Collapse
Affiliation(s)
- Saumya Gupta
- Department of Bioinformatics, BioNome, Bengaluru, India
| | - Susha Dinesh
- Department of Bioinformatics, BioNome, Bengaluru, India
| | - Sameer Sharma
- Department of Bioinformatics, BioNome, Bengaluru, India
| |
Collapse
|
15
|
Kamble SR, Dandekar MP. Implication of microbiota gut-brain axis in the manifestation of obsessive-compulsive disorder: Preclinical and clinical evidence. Eur J Pharmacol 2023; 957:176014. [PMID: 37619786 DOI: 10.1016/j.ejphar.2023.176014] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 08/21/2023] [Accepted: 08/22/2023] [Indexed: 08/26/2023]
Abstract
Recent research has highlighted the key role of gut microbiota in the development of psychiatric disorders. The adverse impact of stress, anxiety, and depression has been well documented on the commensal gut microflora. Thus, therapeutic benefits of gut microbiota-based interventions may not be avoided in central nervous system (CNS) disorders. In this review, we outline the current state of knowledge of gut microbiota with respect to obsessive-compulsive disorder (OCD). We discuss how OCD-generated changes corresponding to the key neurotransmitters, hypothalamic-pituitary-adrenal axis, and immunological and inflammatory pathways are connected with the modifications of the microbiota-gut-brain axis. Notably, administration of few probiotics such as Lactobacillus rhamnosus (ATCC 53103), Lactobacillus helveticus R0052, Bifidobacterium longum R0175, Saccharomyces boulardii, and Lactobacillus casei Shirota imparted positive effects in the management of OCD symptoms. Taken together, we suggest that the gut microbiota-directed therapeutics may open new treatment approaches for the management of OCD.
Collapse
Affiliation(s)
- Sonali R Kamble
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Manoj P Dandekar
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India.
| |
Collapse
|
16
|
Yu Z, Chen W, Zhang L, Chen Y, Chen W, Meng S, Lu L, Han Y, Shi J. Gut-derived bacterial LPS attenuates incubation of methamphetamine craving via modulating microglia. Brain Behav Immun 2023; 111:101-115. [PMID: 37004759 DOI: 10.1016/j.bbi.2023.03.027] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Revised: 03/16/2023] [Accepted: 03/28/2023] [Indexed: 04/04/2023] Open
Abstract
BACKGROUND The microbiota-gut-brain axis plays a critical role in the pathophysiology of neuropsychiatric disorders, and the compositions of gut microbiota are altered by addictive drugs. However, the role of gut microbiota in the incubation of methamphetamine (METH) craving remains poorly understood. METHODS 16S rRNA gene sequencing was performed to assess the richness and diversity of gut microbiota in METH self-administration model. Hematoxylin and eosin staining was performed to evaluate the integrity of intestinal barrier. Immunofluorescence and three-dimensional reconstruction were performed to assess the morphologic changes of microglia. Serum levels of lipopolysaccharide (LPS) were determined using the rat enzyme-linked immunosorbent assay kits. Quantitative real-time PCR was performed to assess transcript levels of dopamine receptor, glutamate ionotropic AMPA receptor 3 and brain-derived neurotrophic factor. RESULTS METH self-administration induced gut microbiota dysbiosis, intestinal barrier damage and microglia activation in the nucleus accumbens core (NAcc), which was partially recovered after prolonged withdrawal. Microbiota depletion via antibiotic treatment increased LPS levels and induced a marked change in the microglial morphology in the NAcc, as indicated by the decreases in the lengths and numbers of microglial branches. Depleting the gut microbiota also prevented the incubation of METH craving and increased the population of Klebsiella oxytoca. Furthermore, Klebsiella oxytoca treatment or exogenous administration of the gram-negative bacterial cell wall component LPS increased serum and central LPS levels, induced microglial morphological changes and reduced the dopamine receptor transcription in the NAcc. Both treatments and NAcc microinjections of gut-derived bacterial LPS significantly decreased METH craving after prolonged withdrawal. CONCLUSIONS These data suggest that LPS from gut gram-negative bacteria may enter circulating blood, activate microglia in the brain and consequently decrease METH craving after withdrawal, which may have important implications for novel strategies to prevent METH addiction and relapse.
Collapse
Affiliation(s)
- Zhoulong Yu
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing 100191, China; National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence Research, Peking University, Beijing 100191, China
| | - Wenjun Chen
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence Research, Peking University, Beijing 100191, China; Department of Neurobiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Libo Zhang
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence Research, Peking University, Beijing 100191, China; Peking University Shenzhen Hospital, Shenzhen 518036, China
| | - Yun Chen
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence Research, Peking University, Beijing 100191, China
| | - Wenxi Chen
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence Research, Peking University, Beijing 100191, China
| | - Shiqiu Meng
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence Research, Peking University, Beijing 100191, China
| | - Lin Lu
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing 100191, China; Peking-Tsinghua Center for Life Sciences and PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing 100871, China
| | - Ying Han
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence Research, Peking University, Beijing 100191, China.
| | - Jie Shi
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence Research, Peking University, Beijing 100191, China; Peking University Shenzhen Hospital, Shenzhen 518036, China; The Key Laboratory for Neuroscience of the Ministry of Education and Health, Peking University, Beijing 100191, China; The State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing 100191, China.
| |
Collapse
|
17
|
Dalziel JE, Zobel G, Dewhurst H, Hurst C, Olson T, Rodriguez-Sanchez R, Mace L, Parkar N, Thum C, Hannaford R, Fraser K, MacGibbon A, Bassett SA, Dekker J, Anderson RC, Young W. A Diet Enriched with Lacticaseibacillus rhamnosus HN001 and Milk Fat Globule Membrane Alters the Gut Microbiota and Decreases Amygdala GABA a Receptor Expression in Stress-Sensitive Rats. Int J Mol Sci 2023; 24:10433. [PMID: 37445611 DOI: 10.3390/ijms241310433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Revised: 06/12/2023] [Accepted: 06/19/2023] [Indexed: 07/15/2023] Open
Abstract
Brain signalling pathways involved in subclinical anxiety and depressed mood can be modulated via the gut brain axis (GBA), providing the potential for diet and dietary components to affect mood. We investigated behavioural, physiological and gut microbiome responses to the Lacticaseibacillus rhamnosus strain HN001 (LactoB HN001™), which has been shown to reduce postpartum anxiety and depression, and a milk fat globule membrane-enriched product, Lipid 70 (SurestartTM MFGM Lipid 70), which has been implicated in memory in stress-susceptible Wistar Kyoto rats. We examined behaviour in the open field, elevated plus maze and novel object recognition tests in conjunction with the expression of host genes in neuro-signalling pathways, and we also assessed brain lipidomics. Treatment-induced alterations in the caecal microbiome and short-chain fatty acid (SCFA) profiles were also assessed. Neither ingredient induced behavioural changes or altered the brain lipidome (separately or when combined). However, with regard to brain gene expression, the L. rhamnosus HN001 + Lipid 70 combination produced a synergistic effect, reducing GABAA subunit expression in the amygdala (Gabre, Gat3, Gabrg1) and hippocampus (Gabrd). Treatment with L. rhamnosus HN001 alone altered expression of the metabotropic glutamate receptor (Grm4) in the amygdala but produced only minor changes in gut microbiota composition. In contrast, Lipid 70 alone did not alter brain gene expression but produced a significant shift in the gut microbiota profile. Under the conditions used, there was no observed effect on rat behaviour for the ingredient combination. However, the enhancement of brain gene expression by L. rhamnosus HN001 + Lipid 70 implicates synergistic actions on region-specific neural pathways associated with fear, anxiety, depression and memory. A significant shift in the gut microbiota profile also occurred that was mainly attributable to Lipid 70.
Collapse
Affiliation(s)
- Julie E Dalziel
- Smart Foods & Bioproducts, AgResearch, Palmerston North 4442, New Zealand
| | - Gosia Zobel
- Ethical Agriculture, AgResearch, Hamilton 3240, New Zealand
| | - Hilary Dewhurst
- Smart Foods & Bioproducts, AgResearch, Palmerston North 4442, New Zealand
| | - Charlotte Hurst
- Smart Foods & Bioproducts, AgResearch, Palmerston North 4442, New Zealand
| | - Trent Olson
- Smart Foods & Bioproducts, AgResearch, Palmerston North 4442, New Zealand
| | | | - Louise Mace
- Smart Foods & Bioproducts, AgResearch, Palmerston North 4442, New Zealand
| | - Nabil Parkar
- Smart Foods & Bioproducts, AgResearch, Palmerston North 4442, New Zealand
| | - Caroline Thum
- Smart Foods & Bioproducts, AgResearch, Palmerston North 4442, New Zealand
| | - Rina Hannaford
- Digital Agriculture, AgResearch, Palmerston North 4442, New Zealand
| | - Karl Fraser
- Smart Foods & Bioproducts, AgResearch, Palmerston North 4442, New Zealand
| | - Alastair MacGibbon
- Fonterra Research and Development Centre Co., Ltd., Palmerston North 4442, New Zealand
| | - Shalome A Bassett
- Fonterra Research and Development Centre Co., Ltd., Palmerston North 4442, New Zealand
| | - James Dekker
- Fonterra Research and Development Centre Co., Ltd., Palmerston North 4442, New Zealand
| | - Rachel C Anderson
- Smart Foods & Bioproducts, AgResearch, Palmerston North 4442, New Zealand
| | - Wayne Young
- Smart Foods & Bioproducts, AgResearch, Palmerston North 4442, New Zealand
| |
Collapse
|
18
|
Anchesi I, Schepici G, Chiricosta L, Gugliandolo A, Salamone S, Caprioglio D, Pollastro F, Mazzon E. Δ 8-THC Induces Up-Regulation of Glutamatergic Pathway Genes in Differentiated SH-SY5Y: A Transcriptomic Study. Int J Mol Sci 2023; 24:ijms24119486. [PMID: 37298437 DOI: 10.3390/ijms24119486] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 05/18/2023] [Accepted: 05/28/2023] [Indexed: 06/12/2023] Open
Abstract
Cannabinoids, natural or synthetic, have antidepressant, anxiolytic, anticonvulsant, and anti-psychotic properties. Cannabidiol (CBD) and delta-9-tetrahydrocannabinol (Δ9-THC) are the most studied cannabinoids, but recently, attention has turned towards minor cannabinoids. Delta-8-tetrahydrocannabinol (Δ8-THC), an isomer of Δ9-THC, is a compound for which, to date, there is no evidence of its role in the modulation of synaptic pathways. The aim of our work was to evaluate the effects of Δ8-THC on differentiated SH-SY5Y human neuroblastoma cells. Using next generation sequencing (NGS), we investigated whether Δ8-THC could modify the transcriptomic profile of genes involved in synapse functions. Our results showed that Δ8-THC upregulates the expression of genes involved in the glutamatergic pathway and inhibits gene expression at cholinergic synapses. Conversely, Δ8-THC did not modify the transcriptomic profile of genes involved in the GABAergic and dopaminergic pathways.
Collapse
Affiliation(s)
- Ivan Anchesi
- IRCCS Centro Neurolesi "Bonino-Pulejo", Via Provinciale Palermo, Contrada Casazza, 98124 Messina, Italy
| | - Giovanni Schepici
- IRCCS Centro Neurolesi "Bonino-Pulejo", Via Provinciale Palermo, Contrada Casazza, 98124 Messina, Italy
| | - Luigi Chiricosta
- IRCCS Centro Neurolesi "Bonino-Pulejo", Via Provinciale Palermo, Contrada Casazza, 98124 Messina, Italy
| | - Agnese Gugliandolo
- IRCCS Centro Neurolesi "Bonino-Pulejo", Via Provinciale Palermo, Contrada Casazza, 98124 Messina, Italy
| | - Stefano Salamone
- Department of Pharmaceutical Sciences, University of Eastern Piedmont, Largo Donegani 2, 28100 Novara, Italy
- PlantaChem S.r.l.s., Via Amico Canobio 4/6, 28100 Novara, Italy
| | - Diego Caprioglio
- Department of Pharmaceutical Sciences, University of Eastern Piedmont, Largo Donegani 2, 28100 Novara, Italy
- PlantaChem S.r.l.s., Via Amico Canobio 4/6, 28100 Novara, Italy
| | - Federica Pollastro
- Department of Pharmaceutical Sciences, University of Eastern Piedmont, Largo Donegani 2, 28100 Novara, Italy
- PlantaChem S.r.l.s., Via Amico Canobio 4/6, 28100 Novara, Italy
| | - Emanuela Mazzon
- IRCCS Centro Neurolesi "Bonino-Pulejo", Via Provinciale Palermo, Contrada Casazza, 98124 Messina, Italy
| |
Collapse
|
19
|
Osorio N, Martineau M, Fortea M, Rouget C, Penalba V, Lee CJ, Boesmans W, Rolli-Derkinderen M, Patel AV, Mondielli G, Conrod S, Labat-Gest V, Papin A, Sasabe J, Sweedler JV, Vanden Berghe P, Delmas P, Mothet JP. d-Serine agonism of GluN1-GluN3 NMDA receptors regulates the activity of enteric neurons and coordinates gut motility. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.19.537136. [PMID: 37131687 PMCID: PMC10153202 DOI: 10.1101/2023.04.19.537136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
The enteric nervous system (ENS) is a complex network of diverse molecularly defined classes of neurons embedded in the gastrointestinal wall and responsible for controlling the major functions of the gut. As in the central nervous system, the vast array of ENS neurons is interconnected by chemical synapses. Despite several studies reporting the expression of ionotropic glutamate receptors in the ENS, their roles in the gut remain elusive. Here, by using an array of immunohistochemistry, molecular profiling and functional assays, we uncover a new role for d-serine (d-Ser) and non-conventional GluN1-GluN3 N-methyl d-aspartate receptors (NMDARs) in regulating ENS functions. We demonstrate that d-Ser is produced by serine racemase (SR) expressed in enteric neurons. By using both in situ patch clamp recording and calcium imaging, we show that d-Ser alone acts as an excitatory neurotransmitter in the ENS independently of the conventional GluN1-GluN2 NMDARs. Instead, d-Ser directly gates the non-conventional GluN1-GluN3 NMDARs in enteric neurons from both mouse and guinea-pig. Pharmacological inhibition or potentiation of GluN1-GluN3 NMDARs had opposite effects on mouse colonic motor activities, while genetically driven loss of SR impairs gut transit and fluid content of pellet output. Our results demonstrate the existence of native GluN1-GluN3 NMDARs in enteric neurons and open new perspectives on the exploration of excitatory d-Ser receptors in gut function and diseases.
Collapse
Affiliation(s)
- Nancy Osorio
- Laboratoire de Neurosciences Cognitives (LNC), Aix-Marseille-Université, CNRS, UMR 7291, Marseille, France
- Centre de Recherche en Neurophysiologie et Neuroscience de Marseille, UMR 7286, CNRS, Université Aix-Marseille, Marseille, France
| | | | - Marina Fortea
- Laboratory for Enteric NeuroScience (LENS), Translational Research Center for Gastrointestinal Disorders (TARGID), University of Leuven, Leuven, Belgium
| | | | - Virginie Penalba
- Laboratoire de Neurosciences Cognitives (LNC), Aix-Marseille-Université, CNRS, UMR 7291, Marseille, France
- Centre de Recherche en Neurophysiologie et Neuroscience de Marseille, UMR 7286, CNRS, Université Aix-Marseille, Marseille, France
| | - Cindy J. Lee
- Department of Chemistry and the Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Werend Boesmans
- Laboratory for Enteric NeuroScience (LENS), Translational Research Center for Gastrointestinal Disorders (TARGID), University of Leuven, Leuven, Belgium
| | | | - Amit V. Patel
- Department of Chemistry and the Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Grégoire Mondielli
- Centre de Recherche en Neurophysiologie et Neuroscience de Marseille, UMR 7286, CNRS, Université Aix-Marseille, Marseille, France
| | - Sandrine Conrod
- Centre de Recherche en Neurophysiologie et Neuroscience de Marseille, UMR 7286, CNRS, Université Aix-Marseille, Marseille, France
| | | | - Amandine Papin
- Laboratoire de Neurosciences Cognitives (LNC), Aix-Marseille-Université, CNRS, UMR 7291, Marseille, France
| | - Jumpei Sasabe
- Department of Pharmacology, Keio University School of Medicine, Tokyo, Japan
| | - Jonathan V. Sweedler
- Department of Chemistry and the Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Pieter Vanden Berghe
- Laboratory for Enteric NeuroScience (LENS), Translational Research Center for Gastrointestinal Disorders (TARGID), University of Leuven, Leuven, Belgium
| | - Patrick Delmas
- Laboratoire de Neurosciences Cognitives (LNC), Aix-Marseille-Université, CNRS, UMR 7291, Marseille, France
- Centre de Recherche en Neurophysiologie et Neuroscience de Marseille, UMR 7286, CNRS, Université Aix-Marseille, Marseille, France
| | - Jean-Pierre Mothet
- Neurocentre Magendie, INSERM UMR U862, Bordeaux, France
- Centre de Recherche en Neurophysiologie et Neuroscience de Marseille, UMR 7286, CNRS, Université Aix-Marseille, Marseille, France
- Université Paris-Saclay, École Normale Supérieure Paris-Saclay, Centre National de la Recherche Scientifique, CentraleSupélec, LuMIn UMR9024, Gif-sur-Yvette 91190, France
| |
Collapse
|
20
|
Binh Tran TD, Nguyen H, Sodergren E, Addiction CFSNO, Dickson PE, Wright SN, Philip VM, Weinstock GM, Chesler EJ, Zhou Y, Bubier JA. Microbial glutamate metabolism predicts intravenous cocaine self-administration in diversity outbred mice. Neuropharmacology 2023; 226:109409. [PMID: 36592885 PMCID: PMC9943525 DOI: 10.1016/j.neuropharm.2022.109409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Revised: 12/23/2022] [Accepted: 12/27/2022] [Indexed: 01/01/2023]
Abstract
The gut microbiome is thought to play a critical role in the onset and development of psychiatric disorders, including depression and substance use disorder (SUD). To test the hypothesis that the microbiome affects addiction predisposing behaviors and cocaine intravenous self-administration (IVSA) and to identify specific microbes involved in the relationship, we performed 16S rRNA gene sequencing on feces from 228 diversity outbred mice. Twelve open field measures, two light-dark assay measures, one hole board and novelty place preference measure significantly differed between mice that acquired cocaine IVSA (ACQ) and those that failed to acquire IVSA (FACQ). We found that ACQ mice are more active and exploratory and display decreased fear than FACQ mice. The microbial abundances that differentiated ACQ from FACQ mice were an increased abundance of Barnesiella, Ruminococcus, and Robinsoniella and decreased Clostridium IV in ACQ mice. There was a sex-specific correlation between ACQ and microbial abundance, a reduced Lactobacillus abundance in ACQ male mice, and a decreased Blautia abundance in female ACQ mice. The abundance of Robinsoniella was correlated, and Clostridium IV inversely correlated with the number of doses of cocaine self-administered during acquisition. Functional analysis of the microbiome composition of a subset of mice suggested that gut-brain modules encoding glutamate metabolism genes are associated with the propensity to self-administer cocaine. These findings establish associations between the microbiome composition and glutamate metabolic potential and the ability to acquire cocaine IVSA thus indicating the potential translational impact of targeting the gut microbiome or microbial metabolites for treatment of SUD. This article is part of the Special Issue on "Microbiome & the Brain: Mechanisms & Maladies".
Collapse
Affiliation(s)
- Thi Dong Binh Tran
- The Jackson Laboratory Genomic Medicine, 10 Discovery Way, Farmington, CT, USA
| | - Hoan Nguyen
- The Jackson Laboratory Genomic Medicine, 10 Discovery Way, Farmington, CT, USA
| | - Erica Sodergren
- The Jackson Laboratory Genomic Medicine, 10 Discovery Way, Farmington, CT, USA
| | | | - Price E Dickson
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine Marshall University, Huntington, WV, USA
| | - Susan N Wright
- Division of Neuroscience and Behavior, National Institute on Drug Abuse, National Institutes of Health, Three White Flint North, Room 08C08 MSC 6018, Bethesda, MD, 20892, USA
| | - Vivek M Philip
- The Jackson Laboratory Mammalian Genetics, 600 Main St, Bar Harbor, ME, USA
| | - George M Weinstock
- The Jackson Laboratory Genomic Medicine, 10 Discovery Way, Farmington, CT, USA
| | - Elissa J Chesler
- The Jackson Laboratory Mammalian Genetics, 600 Main St, Bar Harbor, ME, USA
| | - Yanjiao Zhou
- Department of Medicine, University of Connecticut Health Center, 263 Farmington Avenue, Farmington, CT, USA
| | - Jason A Bubier
- The Jackson Laboratory Mammalian Genetics, 600 Main St, Bar Harbor, ME, USA.
| |
Collapse
|
21
|
Miri S, Yeo J, Abubaker S, Hammami R. Neuromicrobiology, an emerging neurometabolic facet of the gut microbiome? Front Microbiol 2023; 14:1098412. [PMID: 36733917 PMCID: PMC9886687 DOI: 10.3389/fmicb.2023.1098412] [Citation(s) in RCA: 50] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 01/03/2023] [Indexed: 01/18/2023] Open
Abstract
The concept of the gut microbiome is emerging as a metabolic interactome influenced by diet, xenobiotics, genetics, and other environmental factors that affect the host's absorption of nutrients, metabolism, and immune system. Beyond nutrient digestion and production, the gut microbiome also functions as personalized polypharmacy, where bioactive metabolites that our microbes excrete or conjugate may reach systemic circulation and impact all organs, including the brain. Appreciable evidence shows that gut microbiota produce diverse neuroactive metabolites, particularly neurotransmitters (and their precursors), stimulating the local nervous system (i.e., enteric and vagus nerves) and affecting brain function and cognition. Several studies have demonstrated correlations between the gut microbiome and the central nervous system sparking an exciting new research field, neuromicrobiology. Microbiome-targeted interventions are seen as promising adjunctive treatments (pre-, pro-, post-, and synbiotics), but the mechanisms underlying host-microbiome interactions have yet to be established, thus preventing informed evidence-based therapeutic applications. In this paper, we review the current state of knowledge for each of the major classes of microbial neuroactive metabolites, emphasizing their biological effects on the microbiome, gut environment, and brain. Also, we discuss the biosynthesis, absorption, and transport of gut microbiota-derived neuroactive metabolites to the brain and their implication in mental disorders.
Collapse
Affiliation(s)
- Saba Miri
- School of Nutrition Sciences, Faculty of Health Sciences, University of Ottawa, Ottawa, ON, Canada
| | - JuDong Yeo
- School of Nutrition Sciences, Faculty of Health Sciences, University of Ottawa, Ottawa, ON, Canada
| | - Sarah Abubaker
- School of Nutrition Sciences, Faculty of Health Sciences, University of Ottawa, Ottawa, ON, Canada
| | - Riadh Hammami
- School of Nutrition Sciences, Faculty of Health Sciences, University of Ottawa, Ottawa, ON, Canada
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| |
Collapse
|
22
|
Gao S, Shan L, Shi Y, Zhao Y, Mu Q, Cui Y, Chai X, Wang Y. Exploration of the variations of amino acids in Massa Medicata Fermentata and their effects on gastrointestinal diseases. Lebensm Wiss Technol 2022. [DOI: 10.1016/j.lwt.2022.114309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
23
|
Shobatake R, Ota H, Takahashi N, Ueno S, Sugie K, Takasawa S. The Impact of Intermittent Hypoxia on Metabolism and Cognition. Int J Mol Sci 2022; 23:12957. [PMID: 36361741 PMCID: PMC9654766 DOI: 10.3390/ijms232112957] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 10/15/2022] [Accepted: 10/23/2022] [Indexed: 11/29/2022] Open
Abstract
Intermittent hypoxia (IH), one of the primary pathologies of sleep apnea syndrome (SAS), exposes cells throughout the body to repeated cycles of hypoxia/normoxia that result in oxidative stress and systemic inflammation. Since SAS is epidemiologically strongly correlated with type 2 diabetes/insulin resistance, obesity, hypertension, and dyslipidemia included in metabolic syndrome, the effects of IH on gene expression in the corresponding cells of each organ have been studied intensively to clarify the molecular mechanism of the association between SAS and metabolic syndrome. Dementia has recently been recognized as a serious health problem due to its increasing incidence, and a large body of evidence has shown its strong correlation with SAS and metabolic disorders. In this narrative review, we first outline the effects of IH on the expression of genes related to metabolism in neuronal cells, pancreatic β cells, hepatocytes, adipocytes, myocytes, and renal cells (mainly based on the results of our experiments). Next, we discuss the literature regarding the mechanisms by which metabolic disorders and IH develop dementia to understand how IH directly and indirectly leads to the development of dementia.
Collapse
Affiliation(s)
- Ryogo Shobatake
- Department of Neurology, Nara Medical University, 840 Shijo-cho, Kashihara 634-8522, Japan
- Department of Neurology, Nara City Hospital, 1-50-1 Higashikidera-cho, Nara 630-8305, Japan
- Department of Biochemistry, Nara Medical University, 840 Shijo-cho, Kashihara 634-8521, Japan
| | - Hiroyo Ota
- Department Respiratory Medicine, Nara Medical University, 840 Shijo-cho, Kashihara 634-8522, Japan
| | - Nobuyuki Takahashi
- Department of Neurology, Nara City Hospital, 1-50-1 Higashikidera-cho, Nara 630-8305, Japan
| | - Satoshi Ueno
- Department of Neurology, Nara Medical University, 840 Shijo-cho, Kashihara 634-8522, Japan
| | - Kazuma Sugie
- Department of Neurology, Nara Medical University, 840 Shijo-cho, Kashihara 634-8522, Japan
| | - Shin Takasawa
- Department of Biochemistry, Nara Medical University, 840 Shijo-cho, Kashihara 634-8521, Japan
| |
Collapse
|
24
|
Krakovski MA, Arora N, Jain S, Glover J, Dombrowski K, Hernandez B, Yadav H, Sarma AK. Diet-microbiome-gut-brain nexus in acute and chronic brain injury. Front Neurosci 2022; 16:1002266. [PMID: 36188471 PMCID: PMC9523267 DOI: 10.3389/fnins.2022.1002266] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 08/29/2022] [Indexed: 11/13/2022] Open
Abstract
In recent years, appreciation for the gut microbiome and its relationship to human health has emerged as a facilitator of maintaining healthy physiology and a contributor to numerous human diseases. The contribution of the microbiome in modulating the gut-brain axis has gained significant attention in recent years, extensively studied in chronic brain injuries such as Epilepsy and Alzheimer’s Disease. Furthermore, there is growing evidence that gut microbiome also contributes to acute brain injuries like stroke(s) and traumatic brain injury. Microbiome-gut-brain communications are bidirectional and involve metabolite production and modulation of immune and neuronal functions. The microbiome plays two distinct roles: it beneficially modulates immune system and neuronal functions; however, abnormalities in the host’s microbiome also exacerbates neuronal damage or delays the recovery from acute injuries. After brain injury, several inflammatory changes, such as the necrosis and apoptosis of neuronal tissue, propagates downward inflammatory signals to disrupt the microbiome homeostasis; however, microbiome dysbiosis impacts the upward signaling to the brain and interferes with recovery in neuronal functions and brain health. Diet is a superlative modulator of microbiome and is known to impact the gut-brain axis, including its influence on acute and neuronal injuries. In this review, we discussed the differential microbiome changes in both acute and chronic brain injuries, as well as the therapeutic importance of modulation by diets and probiotics. We emphasize the mechanistic studies based on animal models and their translational or clinical relationship by reviewing human studies.
Collapse
Affiliation(s)
| | - Niraj Arora
- Department of Neurology, University of Missouri, Columbia, MO, United States
| | - Shalini Jain
- Department of Neurosurgery and Brain Repair, University of South Florida, Tampa, FL, United States
| | - Jennifer Glover
- Department of Neurosurgery and Brain Repair, University of South Florida, Tampa, FL, United States
| | - Keith Dombrowski
- Department of Neurosurgery and Brain Repair, University of South Florida, Tampa, FL, United States
| | - Beverly Hernandez
- Clinical Nutrition Services, Tampa General Hospital, Tampa, FL, United States
| | - Hariom Yadav
- Department of Neurosurgery and Brain Repair, University of South Florida, Tampa, FL, United States
- USF Center for Microbiome Research, Microbiomes Institute, University of South Florida, Tampa, FL, United States
- *Correspondence: Hariom Yadav,
| | - Anand Karthik Sarma
- Wake Forest University School of Medicine, Winston-Salem, NC, United States
- Department of Neurology, Atrium Health Wake Forest Baptist, Winston-Salem, NC, United States
- Anand Karthik Sarma,
| |
Collapse
|
25
|
Bile Reflux Gastritis: Insights into Pathogenesis, Relevant Factors, Carcinomatous Risk, Diagnosis, and Management. Gastroenterol Res Pract 2022; 2022:2642551. [PMID: 36134174 PMCID: PMC9484982 DOI: 10.1155/2022/2642551] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 08/22/2022] [Indexed: 12/03/2022] Open
Abstract
Bile reflux gastritis (BRG), a kind of gastrointestinal disorder in clinical practice, is characterized by regurgitation and inflammation. However, lack of guidelines leads to simple cognition and even ignorance of this disease for clinicians. Primarily, making the pathogenesis of BRG clear contributes to a correct and general understanding of this disease for physicians. Next, although recently there has been an increasing awareness among researchers in terms of the relevant factors for BRG, further studies involving large samples are still required to certify the relationship between them explicitly. Besides, researches have established that BRG is closely associated with the development of precancerous lesions and gastric cancer. Till now, there is still no golden standard for diagnosis of BRG. Nevertheless, advances in techniques, especially extensive applications of endoscopy and chemical analysis of reflux contents, have improved our ability to identify the occurrence of this disease as well as distinguishing physiological reflux from pathological reflux. Finally, it is fortunate for patients that more and more importance has been attached to the treatment of BRG. From lifestyle modification to drug therapy to surgery, all of them with the view of realizing symptomatic relief are employed for patients with BRG. In this review, we briefly evaluate this disorder based on the best available evidence, offering an overview of its complicated pathogenesis, diverse relevant factors, potential carcinomatous risk, modern diagnostic investigations, and effective therapeutic plans.
Collapse
|
26
|
Qasim H, Nasr M, Mohammad A, Hor M, Baradeiya AM. Dysbiosis and Migraine Headaches in Adults With Celiac Disease. Cureus 2022; 14:e28346. [PMID: 36168375 PMCID: PMC9506300 DOI: 10.7759/cureus.28346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/24/2022] [Indexed: 11/09/2022] Open
Abstract
One of the most significant illnesses associated with gluten is celiac disease, which encompasses many conditions. It is generally recognized that neurological manifestations can occur either at the time of the disease onset or as the illness continues to develop. One of the main clinical presentations of celiac disease is headache, either in the form of migraine or in an unspecific form. Migraine pathophysiology is intricate and still poorly understood. Several mechanisms involving the gut-brain axis have been proposed to explain this association. These include the interaction of chronic inflammation with inflammatory and vasoactive mediators, the modulation of the intestinal immune environment of the microbiota, and the dysfunction of the autonomic nervous system. However, further research is required to fully comprehend the fundamental mechanisms and pathways at play. This review aims to give a narrative summary of the literature on celiac disease's neurological symptoms, particularly migraines, and to assess any potential associations to dysbiosis, an imbalance in the microbiome.
Collapse
|
27
|
Li YR, Li Y, Jin Y, Xu M, Fan HW, Zhang Q, Tan GH, Chen J, Li YQ. Involvement of nitrergic neurons in colonic motility in a rat model of ulcerative colitis. World J Gastroenterol 2022; 28:3854-3868. [PMID: 36157548 PMCID: PMC9367233 DOI: 10.3748/wjg.v28.i29.3854] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 04/27/2022] [Accepted: 07/06/2022] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND The mechanisms underlying gastrointestinal (GI) dysmotility with ulcerative colitis (UC) have not been fully elucidated. The enteric nervous system (ENS) plays an essential role in the GI motility. As a vital neurotransmitter in the ENS, the gas neurotransmitter nitric oxide (NO) may impact the colonic motility. In this study, dextran sulfate sodium (DSS)-induced UC rat model was used for investigating the effects of NO by examining the effects of rate-limiting enzyme nitric oxide synthase (NOS) changes on the colonic motility as well as the role of the ENS in the colonic motility during UC. AIM To reveal the relationship between the effects of NOS expression changes in NOS-containing nitrergic neurons and the colonic motility in a rat UC model. METHODS Male rats (n = 8/each group) were randomly divided into a control (CG), a UC group (EG1), a UC + thrombin derived polypeptide 508 trifluoroacetic acid (TP508TFA; an NOS agonist) group (EG2), and a UC + NG-monomethyl-L-arginine monoacetate (L-NMMA; an NOS inhibitor) group (EG3). UC was induced by administering 5.5% DSS in drinking water without any other treatment (EG1), while the EG2 and EG3 were gavaged with TP508 TFA and L-NMMA, respectively. The disease activity index (DAI) and histological assessment were recorded for each group, whereas the changes in the proportion of colonic nitrergic neurons were counted using immunofluorescence histochemical staining, Western blot, and enzyme linked immunosorbent assay, respectively. In addition, the contractile tension changes in the circular and longitudinal muscles of the rat colon were investigated in vitro using an organ bath system. RESULTS The proportion of NOS-positive neurons within the colonic myenteric plexus (MP), the relative expression of NOS, and the NOS concentration in serum and colonic tissues were significantly elevated in EG1, EG2, and EG3 compared with CG rats. In UC rats, stimulation with agonists and inhibitors led to variable degrees of increase or decrease for each indicator in the EG2 and EG3. When the rats in EGs developed UC, the mean contraction tension of the colonic smooth muscle detected in vitro was higher in the EG1, EG2, and EG3 than in the CG group. Compared with the EG1, the contraction amplitude and mean contraction tension of the circular and longitudinal muscles of the colon in the EG2 and EG3 were enhanced and attenuated, respectively. Thus, during UC, regulation of the expression of NOS within the MP improved the intestinal motility, thereby favoring the recovery of intestinal functions. CONCLUSION In UC rats, an increased number of nitrergic neurons in the colonic MP leads to the attenuation of colonic motor function. To intervene NOS activity might modulate the function of nitrergic neurons in the colonic MP and prevent colonic motor dysfunction. These results might provide clues for a novel approach to alleviate diarrhea symptoms of UC patients.
Collapse
Affiliation(s)
- Yan-Rong Li
- Department of Human Anatomy, Basic Medical College, Guangxi Medical University, Nanning 530000, Guangxi Zhuang Autonomous Region, China
- Department of Gastroenterology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou 121000, Liaoning Province, China
| | - Yan Li
- Department of Human Anatomy, Basic Medical College, Zunyi Medical University, Zunyi 563006, Guizhou Province, China
| | - Yuan Jin
- Department of Human Anatomy, Basic Medical College, Zunyi Medical University, Zunyi 563006, Guizhou Province, China
| | - Mang Xu
- Department of Anatomy, Basic Medical College, Dali University, Dali 671000, Yunnan Province, China
| | - Hong-Wei Fan
- Department of Anatomy, Histology and Embryology, Xuzhou Medical University, Xuzhou 221000, Jiangsu Province, China
| | - Qian Zhang
- Department of Human Anatomy, Basic Medical College, Zunyi Medical University, Zunyi 563006, Guizhou Province, China
| | - Guo-He Tan
- Key Lab of Longevity and Aging-related Diseases of Chinese Ministry of Education, Guangxi Medical University, Nanning 530000, Guangxi Zhuang Autonomous Region, China
- School of Basic Medical Sciences and Center for Translational Medicine, Guangxi Medical University, Nanning 530000, Guangxi Zhuang Autonomous Region, China
| | - Jing Chen
- Department of Anatomy, Histology and Embryology and K. K. Leung Brain Research Centre, The Fourth Military Medical University, Xi'an 710032, Shaanxi Province, China
| | - Yun-Qing Li
- Department of Human Anatomy, Basic Medical College, Guangxi Medical University, Nanning 530000, Guangxi Zhuang Autonomous Region, China
- Department of Human Anatomy, Basic Medical College, Zunyi Medical University, Zunyi 563006, Guizhou Province, China
- Department of Anatomy, Histology and Embryology and K. K. Leung Brain Research Centre, The Fourth Military Medical University, Xi'an 710032, Shaanxi Province, China
- Department of Human Anatomy, College of Preclinical Medical Sciences, Zhengzhou University, Zhengzhou 450001, Henan Province, China
- Key Laboratory of Brain Science Research and Transformation in Tropical Environment of Hainan Province, Hainan Medical University, Haikou 571199, Hainan Province, China
| |
Collapse
|
28
|
Yu F, Hou ZS, Luo HR, Cui XF, Xiao J, Kim YB, Li JL, Feng WR, Tang YK, Li HX, Su SY, Song CY, Wang MY, Xu P. Zinc alters behavioral phenotypes, neurotransmitter signatures, and immune homeostasis in male zebrafish (Danio rerio). THE SCIENCE OF THE TOTAL ENVIRONMENT 2022; 828:154099. [PMID: 35240190 DOI: 10.1016/j.scitotenv.2022.154099] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Revised: 02/14/2022] [Accepted: 02/19/2022] [Indexed: 06/14/2023]
Abstract
Anthropogenic activities discharge zinc into aquatic ecosystems, and the effects of long-term and low-concentration zinc exposure on fish behavior are unclear. We evaluated the behavior and physiology of male zebrafish (Danio rerio) after a 6-week exposure to 1.0 or 1.5 ppm (mg/L) zinc chloride. The exposure caused anxiety-like behaviors and altered the social preferences in both exposure groups. Analysis of transcriptional changes suggested that in the brain, zinc exerted heterogenetic effects on immune and neurotransmitter functions. Exposure to 1.0 ppm zinc chloride resulted in constitutive immune dyshomeostasis, while exposure to 1.5 ppm zinc chloride impaired the neurotransmitter glutamate. In the intestine, zinc dysregulated self-renewal of intestinal cells, a potential loss of defense function. Moreover, exposure to 1.5 ppm zinc chloride suppressed intestinal immune functions and dysregulated tyrosine metabolism. These behavioral alterations suggested that the underlying mechanisms were distinct and concentration-specific. Overall, environmental levels of zinc can alter male zebrafish behaviors by dysregulating neurotransmitter and immunomodulation signatures.
Collapse
Affiliation(s)
- Fan Yu
- Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi 214081, China.
| | - Zhi-Shuai Hou
- Key Laboratory of Mariculture (Ocean University of China), Ministry of Education (KLMME), Ocean University of China, Qingdao 266003, China; Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215, USA
| | - Hong-Rui Luo
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China
| | - Xue-Fan Cui
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China
| | - Jun Xiao
- Guangxi Key Laboratory of Aquatic Genetic Breeding and Healthy Aquaculture, Guangxi Academy of Fishery Sciences, Nanning 530021, China
| | - Young-Bum Kim
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215, USA
| | - Jian-Lin Li
- Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi 214081, China
| | - Wen-Rong Feng
- Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi 214081, China
| | - Yong-Kai Tang
- Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi 214081, China
| | - Hong-Xia Li
- Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi 214081, China
| | - Sheng-Yan Su
- Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi 214081, China
| | - Chang-You Song
- Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi 214081, China
| | - Mei-Yao Wang
- Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi 214081, China
| | - Pao Xu
- Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi 214081, China.
| |
Collapse
|
29
|
Montanari M, Martella G, Bonsi P, Meringolo M. Autism Spectrum Disorder: Focus on Glutamatergic Neurotransmission. Int J Mol Sci 2022; 23:ijms23073861. [PMID: 35409220 PMCID: PMC8998955 DOI: 10.3390/ijms23073861] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 03/24/2022] [Accepted: 03/29/2022] [Indexed: 12/16/2022] Open
Abstract
Disturbances in the glutamatergic system have been increasingly documented in several neuropsychiatric disorders, including autism spectrum disorder (ASD). Glutamate-centered theories of ASD are based on evidence from patient samples and postmortem studies, as well as from studies documenting abnormalities in glutamatergic gene expression and metabolic pathways, including changes in the gut microbiota glutamate metabolism in patients with ASD. In addition, preclinical studies on animal models have demonstrated glutamatergic neurotransmission deficits and altered expression of glutamate synaptic proteins. At present, there are no approved glutamatergic drugs for ASD, but several ongoing clinical trials are currently focusing on evaluating in autistic patients glutamatergic pharmaceuticals already approved for other conditions. In this review, we provide an overview of the literature concerning the role of glutamatergic neurotransmission in the pathophysiology of ASD and as a potential target for novel treatments.
Collapse
Affiliation(s)
- Martina Montanari
- Laboratory of Neurophysiology and Plasticity, IRCCS Fondazione Santa Lucia, 00179 Rome, Italy; (M.M.); (G.M.)
- Department of Systems Neuroscience, University Tor Vergata, 00133 Rome, Italy
| | - Giuseppina Martella
- Laboratory of Neurophysiology and Plasticity, IRCCS Fondazione Santa Lucia, 00179 Rome, Italy; (M.M.); (G.M.)
| | - Paola Bonsi
- Laboratory of Neurophysiology and Plasticity, IRCCS Fondazione Santa Lucia, 00179 Rome, Italy; (M.M.); (G.M.)
- Correspondence: (P.B.); (M.M.)
| | - Maria Meringolo
- Laboratory of Neurophysiology and Plasticity, IRCCS Fondazione Santa Lucia, 00179 Rome, Italy; (M.M.); (G.M.)
- Correspondence: (P.B.); (M.M.)
| |
Collapse
|
30
|
Alibeik N, Pishgar E, Bozorgmehr R, Aghaaliakbari F, Rahimian N. Potential role of gut microbiota in patients with COVID-19, its relationship with lung axis, central nervous system (CNS) axis, and improvement with probiotic therapy. IRANIAN JOURNAL OF MICROBIOLOGY 2022; 14:1-9. [PMID: 35611351 PMCID: PMC9085538 DOI: 10.18502/ijm.v14i1.8794] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Coronavirus Disease 2019 (COVID-19) is a pandemic disease caused by a new corona virus. COVID-19 affects different people in different ways. COVID-19 could affect the gastrointestinal system via gut microbiota impairment. Gut microbiota could affect lung health through a relationship between gut and lung microbiota, which is named gut-lung axis. Gut microbiota impairment plays a role in pathogenesis of various pulmonary disease states, so GI diseases were found to be associated with respiratory diseases. Moreover, most infected people will develop mild to moderate gastrointestinal (GI) symptoms such as diarrhea, vomiting, and stomachache, which is caused by impairment in gut microbiota. Therefore, the current study aimed to review potential role of gut microbiota in patients with COVID-19, its relation with lung axis, Central Nervous System (CNS) axis and improvement with probiotic therapy. Also, this review can be a guide for potential role of gut microbiota in patients with COVID-19.
Collapse
Affiliation(s)
- Nazanin Alibeik
- Department of Internal Medicine, School of Medicine, Firoozgar Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Elham Pishgar
- Gastrointestinal and Liver Diseases Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Ramin Bozorgmehr
- Department of Surgery, School of Medicine, Shahid Madani Hospital, Alborz University of Medical Sciences, Karaj, Iran
| | - Farshad Aghaaliakbari
- Department of Internal Medicine, School of Medicine, Firoozgar Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Neda Rahimian
- Department of Internal Medicine, School of Medicine, Firoozgar Hospital, Iran University of Medical Sciences, Tehran, Iran
- Endocrine Research Center, Institute of Endocrinology and Metabolism, Iran University of Medical Sciences (IUMS), Tehran, Iran
| |
Collapse
|
31
|
Shobatake R, Ota H, Takahashi N, Ueno S, Sugie K, Takasawa S. Anorexigenic Effects of Intermittent Hypoxia on the Gut-Brain Axis in Sleep Apnea Syndrome. Int J Mol Sci 2021; 23:364. [PMID: 35008784 PMCID: PMC8745445 DOI: 10.3390/ijms23010364] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 12/23/2021] [Accepted: 12/28/2021] [Indexed: 02/07/2023] Open
Abstract
Sleep apnea syndrome (SAS) is a breathing disorder characterized by recurrent episodes of upper-airway collapse, resulting in intermittent hypoxia (IH) during sleep. Experimental studies with animals and cellular models have indicated that IH leads to attenuation of glucose-induced insulin secretion from pancreatic β cells and to enhancement of insulin resistance in peripheral tissues and cells, such as the liver (hepatocytes), adipose tissue (adipocytes), and skeletal muscles (myocytes), both of which could lead to obesity. Although obesity is widely recognized as a major factor in SAS, it is controversial whether the development of SAS could contribute directly to obesity, and the effect of IH on the expression of appetite regulatory genes remains elusive. Appetite is regulated appropriately by both the hypothalamus and the gut as a gut-brain axis driven by differential neural and hormonal signals. In this review, we summarized the recent epidemiological findings on the relationship between SAS and feeding behavior and focused on the anorexigenic effects of IH on the gut-brain axis by the IH-induced up-regulation of proopiomelanocortin and cocaine- and amphetamine-regulated transcript in neuronal cells and the IH-induced up-regulation of peptide YY, glucagon-like peptide-1 and neurotensin in enteroendocrine cells and their molecular mechanisms.
Collapse
Affiliation(s)
- Ryogo Shobatake
- Department of Neurology, Nara Medical University, 840 Shijo-cho, Kashihara 634-8522, Japan; (S.U.); (K.S.)
- Department of Neurology, Nara City Hospital, 1-50-1 Higashikidera-cho, Nara 630-8305, Japan;
- Department of Biochemistry, Nara Medical University, 840 Shijo-cho, Kashihara 634-8521, Japan;
| | - Hiroyo Ota
- Department Respiratory Medicine, Nara Medical University, 840 Shijo-cho, Kashihara 634-8522, Japan;
| | - Nobuyuki Takahashi
- Department of Neurology, Nara City Hospital, 1-50-1 Higashikidera-cho, Nara 630-8305, Japan;
| | - Satoshi Ueno
- Department of Neurology, Nara Medical University, 840 Shijo-cho, Kashihara 634-8522, Japan; (S.U.); (K.S.)
| | - Kazuma Sugie
- Department of Neurology, Nara Medical University, 840 Shijo-cho, Kashihara 634-8522, Japan; (S.U.); (K.S.)
| | - Shin Takasawa
- Department of Biochemistry, Nara Medical University, 840 Shijo-cho, Kashihara 634-8521, Japan;
| |
Collapse
|
32
|
Lin A, Shih CT, Chu HF, Chen CW, Cheng YT, Wu CC, Yang CCH, Tsai YC. Lactobacillus fermentum PS150 promotes non-rapid eye movement sleep in the first night effect of mice. Sci Rep 2021; 11:16313. [PMID: 34381098 PMCID: PMC8357945 DOI: 10.1038/s41598-021-95659-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 07/06/2021] [Indexed: 12/30/2022] Open
Abstract
The first night effect (FNE) is a type of sleep disturbance caused by an unfamiliar environment, which leads to difficulty falling asleep and reduced sleep duration. Previously, we reported that Lactobacillus fermentum PS150 (PS150) improves sleep conditions in a pentobarbital-induced sleep mouse model. In this study, we aimed to evaluate the effect of PS150 on the FNE in mice. Briefly, mice were implanted with electrodes and orally administered PS150 for four weeks, and then the FNE was induced by cage changing. Analysis of polysomnographic signals revealed that intervention with PS150 restored non-rapid eye movement (NREM) sleep length under the FNE. Compared to diphenhydramine, a commonly used sleep aid, PS150 had no unwanted side effects, such as rapid eye movement (REM) sleep deprivation and fragmented sleep. Moreover, temporal analysis revealed that PS150 efficiently reduced both sleep latency and time spent restoring normal levels of REM sleep. Taken together, these results suggest that PS150 efficiently ameliorates sleep disturbance caused by the FNE. Additionally, V3–V4 16S rRNA sequencing revealed significant increases in Erysipelotrichia, Actinobacteria, and Coriobacteriia in fecal specimens of the PS150-treated group, indicating that PS150 induces gut microbiota remodeling.
Collapse
Affiliation(s)
- Alexander Lin
- Institute of Biochemistry and Molecular Biology, National Yang-Ming University, 155, Section 2, Linong Street, Beitou District, Taipei, 11221, Taiwan.,Chung Mei Biopharma Co., Ltd., Taichung, Taiwan
| | | | - Hsu-Feng Chu
- Biomedical Industry Ph.D. Program, National Yang-Ming University, Taipei, Taiwan
| | - Chieh-Wen Chen
- Institute of Brain Science, National Yang-Ming University, No. 155, Section 2, Linong Street, Beitou District, Taipei, 11221, Taiwan.,Sleep Research Center, National Yang-Ming University, Taipei, Taiwan
| | - Yu-Ting Cheng
- Institute of Brain Science, National Yang-Ming University, No. 155, Section 2, Linong Street, Beitou District, Taipei, 11221, Taiwan.,Sleep Research Center, National Yang-Ming University, Taipei, Taiwan
| | | | - Cheryl C H Yang
- Institute of Brain Science, National Yang-Ming University, No. 155, Section 2, Linong Street, Beitou District, Taipei, 11221, Taiwan. .,Sleep Research Center, National Yang-Ming University, Taipei, Taiwan. .,Brain Research Center, National Yang-Ming University, Taipei, Taiwan.
| | - Ying-Chieh Tsai
- Institute of Biochemistry and Molecular Biology, National Yang-Ming University, 155, Section 2, Linong Street, Beitou District, Taipei, 11221, Taiwan.
| |
Collapse
|
33
|
Ghaisas S, Harischandra DS, Palanisamy B, Proctor A, Jin H, Dutta S, Sarkar S, Langley M, Zenitsky G, Anantharam V, Kanthasamy A, Phillips GJ, Kanthasamy A. Chronic Manganese Exposure and the Enteric Nervous System: An in Vitro and Mouse in Vivo Study. ENVIRONMENTAL HEALTH PERSPECTIVES 2021; 129:87005. [PMID: 34410835 PMCID: PMC8375672 DOI: 10.1289/ehp7877] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/02/2023]
Abstract
BACKGROUND Chronic environmental exposure to manganese (Mn) can cause debilitating damage to the central nervous system. However, its potential toxic effects on the enteric nervous system (ENS) have yet to be assessed. OBJECTIVE We examined the effect of Mn on the ENS using both cell and animal models. METHOD Rat enteric glial cells (EGCs) and mouse primary enteric cultures were exposed to increasing concentrations of Mn and cell viability and mitochondrial health were assessed using various morphological and functional assays. C57BL/6 mice were exposed daily to a sublethal dose of Mn (15mg/kg/d) for 30 d. Gut peristalsis, enteric inflammation, gut microbiome profile, and fecal metabolite composition were assessed at the end of exposure. RESULTS EGC mitochondria were highly susceptible to Mn neurotoxicity, as evidenced by lower mitochondrial mass, adenosine triphosphate-linked respiration, and aconitase activity as well as higher mitochondrial superoxide, upon Mn exposure. Minor differences were seen in the mouse model: specifically, longer intestinal transit times and higher levels of colonic inflammation. CONCLUSION Based on our findings from this study, Mn preferentially induced mitochondrial dysfunction in a rat EGC line and in vivo resulted in inflammation in the ENS. https://doi.org/10.1289/EHP7877.
Collapse
Affiliation(s)
- Shivani Ghaisas
- Department of Biomedical Sciences, Iowa Center for Advanced Neurotoxicology, Iowa State University, Ames, Iowa, USA
| | - Dilshan S Harischandra
- Department of Biomedical Sciences, Iowa Center for Advanced Neurotoxicology, Iowa State University, Ames, Iowa, USA
| | - Bharathi Palanisamy
- Department of Biomedical Sciences, Iowa Center for Advanced Neurotoxicology, Iowa State University, Ames, Iowa, USA
| | - Alexandra Proctor
- Department of Veterinary Microbiology & Preventive Medicine, College of Veterinary Medicine, Iowa State University, Ames, Iowa, USA
| | - Huajun Jin
- Department of Biomedical Sciences, Iowa Center for Advanced Neurotoxicology, Iowa State University, Ames, Iowa, USA
| | - Somak Dutta
- Department of Statistics, Iowa State University, Ames, Iowa, USA
| | - Souvarish Sarkar
- Department of Biomedical Sciences, Iowa Center for Advanced Neurotoxicology, Iowa State University, Ames, Iowa, USA
| | - Monica Langley
- Department of Biomedical Sciences, Iowa Center for Advanced Neurotoxicology, Iowa State University, Ames, Iowa, USA
| | - Gary Zenitsky
- Department of Biomedical Sciences, Iowa Center for Advanced Neurotoxicology, Iowa State University, Ames, Iowa, USA
| | - Vellareddy Anantharam
- Department of Biomedical Sciences, Iowa Center for Advanced Neurotoxicology, Iowa State University, Ames, Iowa, USA
| | - Arthi Kanthasamy
- Department of Biomedical Sciences, Iowa Center for Advanced Neurotoxicology, Iowa State University, Ames, Iowa, USA
| | - Gregory J Phillips
- Department of Veterinary Microbiology & Preventive Medicine, College of Veterinary Medicine, Iowa State University, Ames, Iowa, USA
| | - Anumantha Kanthasamy
- Department of Biomedical Sciences, Iowa Center for Advanced Neurotoxicology, Iowa State University, Ames, Iowa, USA
| |
Collapse
|
34
|
Gros M, Gros B, Mesonero JE, Latorre E. Neurotransmitter Dysfunction in Irritable Bowel Syndrome: Emerging Approaches for Management. J Clin Med 2021; 10:jcm10153429. [PMID: 34362210 PMCID: PMC8347293 DOI: 10.3390/jcm10153429] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 07/13/2021] [Accepted: 07/28/2021] [Indexed: 02/06/2023] Open
Abstract
Irritable bowel syndrome (IBS) is a functional gastrointestinal disorder whose aetiology is still unknown. Most hypotheses point out the gut-brain axis as a key factor for IBS. The axis is composed of different anatomic and functional structures intercommunicated through neurotransmitters. However, the implications of key neurotransmitters such as norepinephrine, serotonin, glutamate, GABA or acetylcholine in IBS are poorly studied. The aim of this review is to evaluate the current evidence about neurotransmitter dysfunction in IBS and explore the potential therapeutic approaches. IBS patients with altered colorectal motility show augmented norepinephrine and acetylcholine levels in plasma and an increased sensitivity of central serotonin receptors. A decrease of colonic mucosal serotonin transporter and a downregulation of α2 adrenoceptors are also correlated with visceral hypersensitivity and an increase of 5-hydroxyindole acetic acid levels, enhanced expression of high affinity choline transporter and lower levels of GABA. Given these neurotransmitter dysfunctions, novel pharmacological approaches such as 5-HT3 receptor antagonists and 5-HT4 receptor agonists are being explored for IBS management, for their antiemetic and prokinetic effects. GABA-analogous medications are being considered to reduce visceral pain. Moreover, agonists and antagonists of muscarinic receptors are under clinical trials. Targeting neurotransmitter dysfunction could provide promising new approaches for IBS management.
Collapse
Affiliation(s)
- Mónica Gros
- Centro de Salud Univérsitas, Hospital Clínico Universitario Lozano Blesa, 50009 Zaragoza, Spain;
- Instituto de Investigación Sanitaria de Aragón (IIS Aragón), 50009 Zaragoza, Spain; (B.G.); (J.E.M.)
| | - Belén Gros
- Instituto de Investigación Sanitaria de Aragón (IIS Aragón), 50009 Zaragoza, Spain; (B.G.); (J.E.M.)
- Servicio de Urgencias, Hospital Universitario Miguel Servet, 50009 Zaragoza, Spain
| | - José Emilio Mesonero
- Instituto de Investigación Sanitaria de Aragón (IIS Aragón), 50009 Zaragoza, Spain; (B.G.); (J.E.M.)
- Departamento de Farmacología, Fisiología y Medicina Legal y Forense, Facultad de Veterinaria, Universidad de Zaragoza, 50009 Zaragoza, Spain
- Instituto Agroalimentario de Aragón—IA2—(Universidad de Zaragoza—CITA), 50013 Zaragoza, Spain
| | - Eva Latorre
- Instituto de Investigación Sanitaria de Aragón (IIS Aragón), 50009 Zaragoza, Spain; (B.G.); (J.E.M.)
- Instituto Agroalimentario de Aragón—IA2—(Universidad de Zaragoza—CITA), 50013 Zaragoza, Spain
- Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Ciencias, Universidad de Zaragoza, 50009 Zaragoza, Spain
- Correspondence:
| |
Collapse
|
35
|
Onaolapo AY, Onaolapo OJ. Glutamate and depression: Reflecting a deepening knowledge of the gut and brain effects of a ubiquitous molecule. World J Psychiatry 2021; 11:297-315. [PMID: 34327123 PMCID: PMC8311508 DOI: 10.5498/wjp.v11.i7.297] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Revised: 04/13/2021] [Accepted: 05/10/2021] [Indexed: 02/06/2023] Open
Abstract
The versatility of glutamate as the brain’s foremost excitatory neurotransmitter and modulator of neurotransmission and function is considered common knowledge. Years of research have continued to uncover glutamate’s effects and roles in several neurological and neuropsychiatric disorders, including depression. It had been considered that a deeper understanding of the roles of glutamate in depression might open a new door to understanding the pathological basis of the disorder, improve the approach to patient management, and lead to the development of newer drugs that may benefit more patients. This review examines our current understanding of the roles of endogenous and exogenous sources of glutamate and the glutamatergic system in the aetiology, progression and management of depression. It also examines the relationships that link the gut-brain axis, glutamate and depression; as it emphasizes how the gut-brain axis could impact depression pathogenesis and management via changes in glutamate homeostasis. Finally, we consider what the likely future of glutamate-based therapies and glutamate-based therapeutic manipulations in depression are, and if with them, we are now on the final chapter of understanding the neurochemical milieu of depressive disorders.
Collapse
Affiliation(s)
- Adejoke Yetunde Onaolapo
- Behavioural Neuroscience Unit, Neurobiology Subdivision, Department of Anatomy, Ladoke Akintola University of Technology, Oyo State 234, Nigeria
| | - Olakunle James Onaolapo
- Behavioural Neuroscience Unit, Neuropharmacology Subdivision, Department of Pharmacology, Ladoke Akintola University of Technology, Oyo State 234, Nigeria
| |
Collapse
|
36
|
Traini C, Idrizaj E, Garella R, Faussone-Pellegrini MS, Baccari MC, Vannucchi MG. Otilonium Bromide treatment prevents nitrergic functional and morphological changes caused by chronic stress in the distal colon of a rat IBS model. J Cell Mol Med 2021; 25:6988-7000. [PMID: 34109728 PMCID: PMC8278105 DOI: 10.1111/jcmm.16710] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 05/07/2021] [Accepted: 05/11/2021] [Indexed: 12/14/2022] Open
Abstract
Irritable bowel syndrome (IBS) is a highly prevalent gastrointestinal disorder characterized by periods of remission and exacerbation. Among the risk factors to develop IBS, psychosocial stress is widely acknowledged. The water avoidance stress repeatedly applied (rWAS) is considered effective to study IBS etio-pathogenesis. Otilonium bromide (OB), a drug with multiple mechanisms of action, is largely used to treat IBS patients. Orally administered, it concentrates in the large bowel and significantly ameliorates the IBS symptomatology. Presently, we tested whether rWAS rats developed neuro-muscular abnormalities in the distal colon and whether OB treatment prevented them. The investigation was focussed on the nitrergic neurotransmission by combining functional and morphological methodologies. The results confirm rWAS as reliable animal model to investigate the cellular mechanisms responsible for IBS: exposure to one-hour psychosocial stress for 10 days depressed muscle contractility and increased iNOS expression in myenteric neurons. OB treatment counteracted these effects. We hypothesize that these effects are due to the corticotropin-releasing factor (CRF) release, the main mediator of the psychosocial stress, followed by a CRF1receptor activation. OB, that was shown to prevent CRF1r activation, reasonably interrupted the cascade events that bring to the mechanical and immunohistochemical changes affecting rWAS rat colon.
Collapse
Affiliation(s)
- Chiara Traini
- Research Unit of Histology and Embryology, Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Eglantina Idrizaj
- Section of Physiological Sciences, Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Rachele Garella
- Section of Physiological Sciences, Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | | | - Maria Caterina Baccari
- Section of Physiological Sciences, Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Maria Giuliana Vannucchi
- Research Unit of Histology and Embryology, Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| |
Collapse
|
37
|
Cerantola S, Caputi V, Contarini G, Mereu M, Bertazzo A, Bosi A, Banfi D, Mantini D, Giaroni C, Giron MC. Dopamine Transporter Genetic Reduction Induces Morpho-Functional Changes in the Enteric Nervous System. Biomedicines 2021; 9:biomedicines9050465. [PMID: 33923250 PMCID: PMC8146213 DOI: 10.3390/biomedicines9050465] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 04/19/2021] [Accepted: 04/20/2021] [Indexed: 12/23/2022] Open
Abstract
Antidopaminergic gastrointestinal prokinetics are indeed commonly used to treat gastrointestinal motility disorders, although the precise role of dopaminergic transmission in the gut is still unclear. Since dopamine transporter (DAT) is involved in several brain disorders by modulating extracellular dopamine in the central nervous system, this study evaluated the impact of DAT genetic reduction on the morpho-functional integrity of mouse small intestine enteric nervous system (ENS). In DAT heterozygous (DAT+/-) and wild-type (DAT+/+) mice (14 ± 2 weeks) alterations in small intestinal contractility were evaluated by isometrical assessment of neuromuscular responses to receptor and non-receptor-mediated stimuli. Changes in ENS integrity were studied by real-time PCR and confocal immunofluorescence microscopy in longitudinal muscle-myenteric plexus whole-mount preparations (). DAT genetic reduction resulted in a significant increase in dopamine-mediated effects, primarily via D1 receptor activation, as well as in reduced cholinergic response, sustained by tachykininergic and glutamatergic neurotransmission via NMDA receptors. These functional anomalies were associated to architectural changes in the neurochemical coding and S100β immunoreactivity in small intestine myenteric plexus. Our study provides evidence that genetic-driven DAT defective activity determines anomalies in ENS architecture and neurochemical coding together with ileal dysmotility, highlighting the involvement of dopaminergic system in gut disorders, often associated to neurological conditions.
Collapse
Affiliation(s)
- Silvia Cerantola
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 35131 Padova, Italy; (S.C.); (V.C.); (M.M.); (A.B.)
| | - Valentina Caputi
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 35131 Padova, Italy; (S.C.); (V.C.); (M.M.); (A.B.)
- Department of Poultry Science, University of Arkansas, Fayetteville, AR 72704, USA
| | - Gabriella Contarini
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95131 Catania, Italy;
| | - Maddalena Mereu
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 35131 Padova, Italy; (S.C.); (V.C.); (M.M.); (A.B.)
| | - Antonella Bertazzo
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 35131 Padova, Italy; (S.C.); (V.C.); (M.M.); (A.B.)
| | - Annalisa Bosi
- Department of Medicine and Surgery, University of Insubria, 21100 Varese, Italy; (A.B.); (D.B.); (C.G.)
| | - Davide Banfi
- Department of Medicine and Surgery, University of Insubria, 21100 Varese, Italy; (A.B.); (D.B.); (C.G.)
| | - Dante Mantini
- IRCCS San Camillo Hospital, 30126 Venice, Italy; or
- Motor Control and Neuroplasticity Research Group, KU Leuven, 3000 Leuven, Belgium
| | - Cristina Giaroni
- Department of Medicine and Surgery, University of Insubria, 21100 Varese, Italy; (A.B.); (D.B.); (C.G.)
| | - Maria Cecilia Giron
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 35131 Padova, Italy; (S.C.); (V.C.); (M.M.); (A.B.)
- IRCCS San Camillo Hospital, 30126 Venice, Italy; or
- Correspondence: ; Tel.: +39-049-827-5091; Fax: +39-049-827-5093
| |
Collapse
|
38
|
|
39
|
Kyoung H, Lee JJ, Cho JH, Choe J, Kang J, Lee H, Liu Y, Kim Y, Kim HB, Song M. Dietary Glutamic Acid Modulates Immune Responses and Gut Health of Weaned Pigs. Animals (Basel) 2021; 11:ani11020504. [PMID: 33671988 PMCID: PMC7919271 DOI: 10.3390/ani11020504] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 02/09/2021] [Accepted: 02/10/2021] [Indexed: 12/14/2022] Open
Abstract
Simple Summary Weaning stress can lead to intestinal barrier dysfunction, immune system destruction, and intestinal microbiota disruption, thereby reducing the absorption of nutrients and causing intestinal diseases. Glutamic acid is a non-essential amino acid that is abundantly present in the body and plays an essential function in cellular metabolism and immune responses. In this study, the effects of dietary glutamic acid on the growth performance, nutrient digestibility, immune responses, and intestinal health of weaned pigs were evaluated. Based on the results, dietary glutamic acid increased growth performance, nutrient digestibility, intestinal morphology, and ileal gene expression of tight junction proteins of weaned pigs and modified immune responses and gut microbiota. This study provides information to understand the functional use of dietary glutamic acid as a feed additive for improving the growth performance and intestinal health of weaned pigs. Abstract Dietary glutamic acid (GLU) is used as a feed additive because of its functional characteristics that may affect the growth performance and health of pigs. This study was carried out to determine the effects of dietary GLU on growth performance, nutrient digestibility, immune responses, and intestinal health of weaned pigs. A total of ninety-six weaned pigs (8.07 ± 1.17 kg of body weight; 28 days of age) were assigned to two dietary treatments (8 pigs/pen; 6 replicates/treatment) in a randomized complete block design (block: body weight): (1) a typical weaner diet (CON) and (2) CON supplemented with 0.5% GLU. The experimental period was for 4 weeks. All data and sample collections were performed at the specific time points during the experimental period. Pigs fed GLU had higher average daily gain and average daily feed intake for the first two weeks and nutrient digestibility than pigs fed CON. In addition, dietary GLU increased villus height to crypt depth ratio, number of goblet cells, and ileal gene expression of claudin family and occludin compared with CON, but decreased serum TNF-α and IL-6 and ileal gene expression of TNF-α. Moreover, pigs fed GLU had increased relative composition of bacterial communities of genus Prevotella and Anaerovibrio and decreased genus Clostridium and Terrisporobacter compared with those fed CON. This study suggests that dietary GLU influences growth performance and health of weaned pigs by modulating nutrient digestibility, intestinal morphology, ileal gene expression of tight junction proteins and cytokines, immune responses, and microbial community in the gut.
Collapse
Affiliation(s)
- Hyunjin Kyoung
- Division of Animal and Dairy Science, Chungnam National University, Daejeon 34134, Korea; (H.K.); (J.J.L.); (J.K.)
| | - Jeong Jae Lee
- Division of Animal and Dairy Science, Chungnam National University, Daejeon 34134, Korea; (H.K.); (J.J.L.); (J.K.)
| | - Jin Ho Cho
- Division of Food and Animal Science, Chungbuk National University, Cheongju 28644, Korea;
| | - Jeehwan Choe
- Department of Beef Science, Korea National College of Agriculture and Fisheries, Jeonju 54874, Korea;
| | - Joowon Kang
- Division of Animal and Dairy Science, Chungnam National University, Daejeon 34134, Korea; (H.K.); (J.J.L.); (J.K.)
| | - Hanbae Lee
- Pathway Intermediates, Seoul 06253, Korea;
| | - Yanhong Liu
- Department of Animal Science, University of California, Davis, CA 95616, USA;
| | - Younghoon Kim
- Department of Agricultural Biotechnology and Research, Institute of Agriculture and Life Science, Seoul National University, Seoul 08826, Korea;
| | - Hyeun Bum Kim
- Department of Animal Resources Science, Dankook University, Cheonan 31116, Korea
- Correspondence: (H.B.K.); (M.S.)
| | - Minho Song
- Division of Animal and Dairy Science, Chungnam National University, Daejeon 34134, Korea; (H.K.); (J.J.L.); (J.K.)
- Correspondence: (H.B.K.); (M.S.)
| |
Collapse
|
40
|
Huang Y, Wang C, Tian X, Mao Y, Hou B, Sun Y, Gu X, Ma Z. Pioglitazone Attenuates Experimental Colitis-Associated Hyperalgesia through Improving the Intestinal Barrier Dysfunction. Inflammation 2021; 43:568-578. [PMID: 31989391 PMCID: PMC7170986 DOI: 10.1007/s10753-019-01138-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Impaired intestinal mucosal integrity during colitis involves the peroxisome proliferator-activated receptor-γ (PPARγ), an important anti-inflammatory factor in intestinal mucosa homoeostasis, which is a potential target in colitis. Recurrent chronic pain is a vital pathogenetic feature of colitis. Nevertheless, potential functions of PPARγ in the colitis-associated hyperalgesia remain unclear. This study aimed to investigate biological roles of pioglitazone in relieving colitis-associated pain hypersensitivity by a PPARγ tight junction protein-dependent mechanism during the course of dextran sodium sulfate (DSS)-induced intestinal inflammation. The DSS-induced colitis model was generated in C57BL/6 mice. Changes in colitis induced the injury of intestinal mucosal barrier and hyperalgesia after a 6-day treatment of pioglitazone (25 mg/kg, IP injection) were assessed through immunofluorescent, hematoxylin and eosin (H&E) staining, western blot analysis, and determination of paw withdrawal mechanical threshold. A significant reduction of paw withdrawal mechanical threshold occurred after DSS treatment. Follow-up data showed that systematic administration of PPARγ agonist pioglitazone ameliorated the DSS-induced colitis and the development of colitis-associated hyperalgesia by repairing the intestinal mucosal barrier. The tight junction proteins ZO-1 and Claudin-5 were upregulated by PPARγ signaling, which in turn promoted the improvement of intestinal barrier function. Moreover, pioglitazone inhibited phosphorylation of ERK and NF-κB in the colon and decreased the levels of inflammatory cytokines in both colon spine tissues. Furthermore, systemically pioglitazone treatment inhibited the activation of microglia and astrocytes, as well as DSS-induced phosphorylation of NR2B subunit in spinal cord, which was correspondingly consistent with the pain behavior. Pioglitazone ameliorates DSS-induced colitis and attenuates colitis-associated mechanical hyperalgesia, with improving integrity of the intestinal mucosal barrier by directly upregulating tight junction proteins. The PPARγ-tight junction protein signaling might be a potential therapeutic target for the treatment of colitis-associated chronic pain.
Collapse
Affiliation(s)
- Yulin Huang
- Department of Anaesthesiology, Affiliated Drum Tower Hospital of Medical School of Nanjing University, Nanjing, 210008, Jiangsu province, China
| | - Chenchen Wang
- Department of Anaesthesiology, Affiliated Drum Tower Hospital of Medical School of Nanjing University, Nanjing, 210008, Jiangsu province, China
| | - Xinyu Tian
- Department of Anaesthesiology, Affiliated Drum Tower Hospital of Medical School of Nanjing University, Nanjing, 210008, Jiangsu province, China
| | - Yanting Mao
- Department of Anaesthesiology, Affiliated Drum Tower Hospital of Medical School of Nanjing University, Nanjing, 210008, Jiangsu province, China
| | - Bailin Hou
- Department of Anaesthesiology, Affiliated Drum Tower Hospital of Medical School of Nanjing University, Nanjing, 210008, Jiangsu province, China
| | - Yu'e Sun
- Department of Anaesthesiology, Affiliated Drum Tower Hospital of Medical School of Nanjing University, Nanjing, 210008, Jiangsu province, China
| | - Xiaoping Gu
- Department of Anaesthesiology, Affiliated Drum Tower Hospital of Medical School of Nanjing University, Nanjing, 210008, Jiangsu province, China.
| | - Zhengliang Ma
- Department of Anaesthesiology, Affiliated Drum Tower Hospital of Medical School of Nanjing University, Nanjing, 210008, Jiangsu province, China.
| |
Collapse
|
41
|
Banfi D, Moro E, Bosi A, Bistoletti M, Cerantola S, Crema F, Maggi F, Giron MC, Giaroni C, Baj A. Impact of Microbial Metabolites on Microbiota-Gut-Brain Axis in Inflammatory Bowel Disease. Int J Mol Sci 2021; 22:1623. [PMID: 33562721 PMCID: PMC7915037 DOI: 10.3390/ijms22041623] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 01/29/2021] [Accepted: 02/02/2021] [Indexed: 02/07/2023] Open
Abstract
The complex bidirectional communication system existing between the gastrointestinal tract and the brain initially termed the "gut-brain axis" and renamed the "microbiota-gut-brain axis", considering the pivotal role of gut microbiota in sustaining local and systemic homeostasis, has a fundamental role in the pathogenesis of Inflammatory Bowel Disease (IBD). The integration of signals deriving from the host neuronal, immune, and endocrine systems with signals deriving from the microbiota may influence the development of the local inflammatory injury and impacts also more distal brain regions, underlying the psychophysiological vulnerability of IBD patients. Mood disorders and increased response to stress are frequently associated with IBD and may affect the disease recurrence and severity, thus requiring an appropriate therapeutic approach in addition to conventional anti-inflammatory treatments. This review highlights the more recent evidence suggesting that alterations of the microbiota-gut-brain bidirectional communication axis may concur to IBD pathogenesis and sustain the development of both local and CNS symptoms. The participation of the main microbial-derived metabolites, also defined as "postbiotics", such as bile acids, short-chain fatty acids, and tryptophan metabolites in the development of IBD-associated gut and brain dysfunction will be discussed. The last section covers a critical evaluation of the main clinical evidence pointing to the microbiome-based therapeutic approaches for the treatment of IBD-related gastrointestinal and neuropsychiatric symptoms.
Collapse
Affiliation(s)
- Davide Banfi
- Department of Medicine and Surgery, University of Insubria, via H Dunant 5, 21100 Varese, Italy; (D.B.); (A.B.); (M.B.); (F.M.); (A.B.)
| | - Elisabetta Moro
- Department of Internal Medicine and Therapeutics, Section of Pharmacology, University of Pavia, via Ferrata 9, 27100 Pavia, Italy; (E.M.); (F.C.)
| | - Annalisa Bosi
- Department of Medicine and Surgery, University of Insubria, via H Dunant 5, 21100 Varese, Italy; (D.B.); (A.B.); (M.B.); (F.M.); (A.B.)
| | - Michela Bistoletti
- Department of Medicine and Surgery, University of Insubria, via H Dunant 5, 21100 Varese, Italy; (D.B.); (A.B.); (M.B.); (F.M.); (A.B.)
| | - Silvia Cerantola
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Largo Meneghetti 2, 35131 Padova, Italy; (S.C.); (M.C.G.)
| | - Francesca Crema
- Department of Internal Medicine and Therapeutics, Section of Pharmacology, University of Pavia, via Ferrata 9, 27100 Pavia, Italy; (E.M.); (F.C.)
| | - Fabrizio Maggi
- Department of Medicine and Surgery, University of Insubria, via H Dunant 5, 21100 Varese, Italy; (D.B.); (A.B.); (M.B.); (F.M.); (A.B.)
| | - Maria Cecilia Giron
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Largo Meneghetti 2, 35131 Padova, Italy; (S.C.); (M.C.G.)
| | - Cristina Giaroni
- Department of Medicine and Surgery, University of Insubria, via H Dunant 5, 21100 Varese, Italy; (D.B.); (A.B.); (M.B.); (F.M.); (A.B.)
- Centre of Neuroscience, University of Insubria, 21100 Varese, Italy
| | - Andreina Baj
- Department of Medicine and Surgery, University of Insubria, via H Dunant 5, 21100 Varese, Italy; (D.B.); (A.B.); (M.B.); (F.M.); (A.B.)
| |
Collapse
|
42
|
Mishima Y, Ishihara S. Molecular Mechanisms of Microbiota-Mediated Pathology in Irritable Bowel Syndrome. Int J Mol Sci 2020; 21:ijms21228664. [PMID: 33212919 PMCID: PMC7698457 DOI: 10.3390/ijms21228664] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Revised: 11/12/2020] [Accepted: 11/13/2020] [Indexed: 02/07/2023] Open
Abstract
Irritable bowel syndrome (IBS) is one of the most prevalent functional gastrointestinal disorders, and accumulating evidence gained in both preclinical and clinical studies indicate the involvement of enteric microbiota in its pathogenesis. Gut resident microbiota appear to influence brain activity through the enteric nervous system, while their composition and function are affected by the central nervous system. Based on these results, the term “brain–gut–microbiome axis” has been proposed and enteric microbiota have become a potential therapeutic target in IBS cases. However, details regarding the microbe-related pathophysiology of IBS remain elusive. This review summarizes the existing knowledge of molecular mechanisms in the pathogenesis of IBS as well as recent progress related to microbiome-derived neurotransmitters, compounds, metabolites, neuroendocrine factors, and enzymes.
Collapse
|
43
|
Shaffer M, Borton MA, McGivern BB, Zayed AA, La Rosa SL, Solden LM, Liu P, Narrowe AB, Rodríguez-Ramos J, Bolduc B, Gazitúa MC, Daly RA, Smith GJ, Vik DR, Pope PB, Sullivan MB, Roux S, Wrighton KC. DRAM for distilling microbial metabolism to automate the curation of microbiome function. Nucleic Acids Res 2020; 48:8883-8900. [PMID: 32766782 PMCID: PMC7498326 DOI: 10.1093/nar/gkaa621] [Citation(s) in RCA: 419] [Impact Index Per Article: 83.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 06/29/2020] [Accepted: 07/21/2020] [Indexed: 12/20/2022] Open
Abstract
Microbial and viral communities transform the chemistry of Earth's ecosystems, yet the specific reactions catalyzed by these biological engines are hard to decode due to the absence of a scalable, metabolically resolved, annotation software. Here, we present DRAM (Distilled and Refined Annotation of Metabolism), a framework to translate the deluge of microbiome-based genomic information into a catalog of microbial traits. To demonstrate the applicability of DRAM across metabolically diverse genomes, we evaluated DRAM performance on a defined, in silico soil community and previously published human gut metagenomes. We show that DRAM accurately assigned microbial contributions to geochemical cycles and automated the partitioning of gut microbial carbohydrate metabolism at substrate levels. DRAM-v, the viral mode of DRAM, established rules to identify virally-encoded auxiliary metabolic genes (AMGs), resulting in the metabolic categorization of thousands of putative AMGs from soils and guts. Together DRAM and DRAM-v provide critical metabolic profiling capabilities that decipher mechanisms underpinning microbiome function.
Collapse
Affiliation(s)
- Michael Shaffer
- Department of Soil and Crop Sciences, Colorado State University, Fort Collins, CO 80523, USA
| | - Mikayla A Borton
- Department of Soil and Crop Sciences, Colorado State University, Fort Collins, CO 80523, USA
| | - Bridget B McGivern
- Department of Soil and Crop Sciences, Colorado State University, Fort Collins, CO 80523, USA
| | - Ahmed A Zayed
- Department of Microbiology, The Ohio State University, Columbus, OH 43210, USA
| | | | - Lindsey M Solden
- Department of Microbiology, The Ohio State University, Columbus, OH 43210, USA
| | - Pengfei Liu
- Department of Soil and Crop Sciences, Colorado State University, Fort Collins, CO 80523, USA
| | - Adrienne B Narrowe
- Department of Soil and Crop Sciences, Colorado State University, Fort Collins, CO 80523, USA
| | - Josué Rodríguez-Ramos
- Department of Soil and Crop Sciences, Colorado State University, Fort Collins, CO 80523, USA
| | - Benjamin Bolduc
- Department of Microbiology, The Ohio State University, Columbus, OH 43210, USA
| | - M Consuelo Gazitúa
- Department of Microbiology, The Ohio State University, Columbus, OH 43210, USA
| | - Rebecca A Daly
- Department of Soil and Crop Sciences, Colorado State University, Fort Collins, CO 80523, USA
| | - Garrett J Smith
- Department of Microbiology, Radboud University, Nijmegen 6525, Netherlands
| | - Dean R Vik
- Department of Microbiology, The Ohio State University, Columbus, OH 43210, USA
| | - Phil B Pope
- Faculty of Biosciences, Norwegian University of Life Sciences, Aas 1432, Norway
| | - Matthew B Sullivan
- Department of Microbiology, The Ohio State University, Columbus, OH 43210, USA
| | - Simon Roux
- Joint Genome Institute, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | - Kelly C Wrighton
- Department of Soil and Crop Sciences, Colorado State University, Fort Collins, CO 80523, USA
| |
Collapse
|
44
|
Suganya K, Koo BS. Gut-Brain Axis: Role of Gut Microbiota on Neurological Disorders and How Probiotics/Prebiotics Beneficially Modulate Microbial and Immune Pathways to Improve Brain Functions. Int J Mol Sci 2020; 21:E7551. [PMID: 33066156 PMCID: PMC7589356 DOI: 10.3390/ijms21207551] [Citation(s) in RCA: 148] [Impact Index Per Article: 29.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 10/06/2020] [Accepted: 10/09/2020] [Indexed: 02/07/2023] Open
Abstract
The gut microbiome acts as an integral part of the gastrointestinal tract (GIT) that has the largest and vulnerable surface with desirable features to observe foods, nutrients, and environmental factors, as well as to differentiate commensals, invading pathogens, and others. It is well-known that the gut has a strong connection with the central nervous system (CNS) in the context of health and disease. A healthy gut with diverse microbes is vital for normal brain functions and emotional behaviors. In addition, the CNS controls most aspects of the GI physiology. The molecular interaction between the gut/microbiome and CNS is complex and bidirectional, ensuring the maintenance of gut homeostasis and proper digestion. Besides this, several mechanisms have been proposed, including endocrine, neuronal, toll-like receptor, and metabolites-dependent pathways. Changes in the bidirectional relationship between the GIT and CNS are linked with the pathogenesis of gastrointestinal and neurological disorders; therefore, the microbiota/gut-and-brain axis is an emerging and widely accepted concept. In this review, we summarize the recent findings supporting the role of the gut microbiota and immune system on the maintenance of brain functions and the development of neurological disorders. In addition, we highlight the recent advances in improving of neurological diseases by probiotics/prebiotics/synbiotics and fecal microbiota transplantation via the concept of the gut-brain axis.
Collapse
Affiliation(s)
- Kanmani Suganya
- Department of Oriental Medicine, Dongguk University, Gyeongju 38066, Korea;
- Department of Oriental Neuropsychiatry, Graduate School of Oriental Medicine, Dongguk University, Ilsan Hospital, 814 Siksa-dong, Goyang-si, Gyeonggi-do 10326, Korea
| | - Byung-Soo Koo
- Department of Oriental Medicine, Dongguk University, Gyeongju 38066, Korea;
- Department of Oriental Neuropsychiatry, Graduate School of Oriental Medicine, Dongguk University, Ilsan Hospital, 814 Siksa-dong, Goyang-si, Gyeonggi-do 10326, Korea
| |
Collapse
|
45
|
Up-regulation of HTR1A reverses stress-induced visceral hypersensitivity through modulating interactions among the anterior cingulate cortex, insular cortex and hippocampus. Pteridines 2020. [DOI: 10.1515/pteridines-2020-0016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Abstract
Background: This study aimed to explore the effect of 5-HT1A receptors (HTR1A) on activation of the anterior cingulate cortex and simultaneous regulation of neural activity in the insular cortex and hippocampus.
Methods: The IBS rat model was established via chronic water avoidance stress (WAS). Visceral sensitivity was measured by electromyogram, and anxiety-like behaviours were evaluated by the open field test. HTR1A-specific lentivirus expressing green fluorescent protein was used to overexpress or down-regulate HTR1A expression. Protein expression levels were detected by western blot.
Results: Up-regulation of HTR1A in ACC could inhibit ACC sensitization and reverse the visceral hypersensitivity and anxiety-like behaviours induced by chronic psychological stress. In contrast, down-regulation of HTR1A in ACC might promote these behaviors in IBS rats. Additionally, up-regulation of HTR1A in ACC could inhibit IC and hippocampus sensitization, while down-regulation might have the opposite effect.
Conclusions: In IBS rats, HTR1A could modulate ACC activation and interactions among the ACC, IC and hippocampus. These effects might in turn contribute to the development of visceral hypersensitivity and anxiety-like behaviours induced by chronic psychological stress.
Collapse
|
46
|
Bistoletti M, Bosi A, Banfi D, Giaroni C, Baj A. The microbiota-gut-brain axis: Focus on the fundamental communication pathways. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2020; 176:43-110. [PMID: 33814115 DOI: 10.1016/bs.pmbts.2020.08.012] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Michela Bistoletti
- Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Annalisa Bosi
- Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Davide Banfi
- Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Cristina Giaroni
- Department of Medicine and Surgery, University of Insubria, Varese, Italy.
| | - Andreina Baj
- Department of Medicine and Surgery, University of Insubria, Varese, Italy
| |
Collapse
|
47
|
Neurotoxicity in Gulf War Illness and the potential role of glutamate. Neurotoxicology 2020; 80:60-70. [DOI: 10.1016/j.neuro.2020.06.008] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 06/16/2020] [Accepted: 06/19/2020] [Indexed: 02/06/2023]
|
48
|
Pajares M, I. Rojo A, Manda G, Boscá L, Cuadrado A. Inflammation in Parkinson's Disease: Mechanisms and Therapeutic Implications. Cells 2020; 9:cells9071687. [PMID: 32674367 PMCID: PMC7408280 DOI: 10.3390/cells9071687] [Citation(s) in RCA: 407] [Impact Index Per Article: 81.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 07/11/2020] [Accepted: 07/13/2020] [Indexed: 02/07/2023] Open
Abstract
Parkinson's disease (PD) is a common neurodegenerative disorder primarily characterized by the death of dopaminergic neurons that project from the substantia nigra pars compacta. Although the molecular bases for PD development are still little defined, extensive evidence from human samples and animal models support the involvement of inflammation in onset or progression. However, the exact trigger for this response remains unclear. Here, we provide a systematic review of the cellular mediators, i.e., microglia, astroglia and endothelial cells. We also discuss the genetic and transcriptional control of inflammation in PD and the immunomodulatory role of dopamine and reactive oxygen species. Finally, we summarize the preclinical and clinical approaches targeting neuroinflammation in PD.
Collapse
Affiliation(s)
- Marta Pajares
- Instituto de Investigaciones Biomédicas “Alberto Sols” UAM-CSIC, 28029 Madrid, Spain; (M.P.); (A.I.R.); (L.B.)
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), ISCIII, 28031 Madrid, Spain
| | - Ana I. Rojo
- Instituto de Investigaciones Biomédicas “Alberto Sols” UAM-CSIC, 28029 Madrid, Spain; (M.P.); (A.I.R.); (L.B.)
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), ISCIII, 28031 Madrid, Spain
- Instituto de Investigación Sanitaria La Paz (IdiPaz), 28029 Madrid, Spain
- Department of Biochemistry, Faculty of Medicine, Autonomous University of Madrid, 28029 Madrid, Spain
| | - Gina Manda
- Department Cellular and Molecular Medicine, Victor Babes National Institute of Pathology, 050096 Bucharest, Romania;
| | - Lisardo Boscá
- Instituto de Investigaciones Biomédicas “Alberto Sols” UAM-CSIC, 28029 Madrid, Spain; (M.P.); (A.I.R.); (L.B.)
- Instituto de Investigación Sanitaria La Paz (IdiPaz), 28029 Madrid, Spain
- Centro de Investigación Biomédica en Red Sobre Enfermedades Cardiovasculares (CIBERcv), ISCIII, 28029 Madrid, Spain
| | - Antonio Cuadrado
- Instituto de Investigaciones Biomédicas “Alberto Sols” UAM-CSIC, 28029 Madrid, Spain; (M.P.); (A.I.R.); (L.B.)
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), ISCIII, 28031 Madrid, Spain
- Instituto de Investigación Sanitaria La Paz (IdiPaz), 28029 Madrid, Spain
- Department of Biochemistry, Faculty of Medicine, Autonomous University of Madrid, 28029 Madrid, Spain
- Department Cellular and Molecular Medicine, Victor Babes National Institute of Pathology, 050096 Bucharest, Romania;
- Correspondence: ; Tel.: +34-915854383; Fax: +34-915854401
| |
Collapse
|
49
|
Bistoletti M, Bosi A, Caon I, Chiaravalli AM, Moretto P, Genoni A, Moro E, Karousou E, Viola M, Crema F, Baj A, Passi A, Vigetti D, Giaroni C. Involvement of hyaluronan in the adaptive changes of the rat small intestine neuromuscular function after ischemia/reperfusion injury. Sci Rep 2020; 10:11521. [PMID: 32661417 PMCID: PMC7359366 DOI: 10.1038/s41598-020-67876-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 04/30/2020] [Indexed: 12/12/2022] Open
Abstract
Intestinal ischemia/reperfusion (I/R) injury has severe consequences on myenteric neurons, which can be irreversibly compromised resulting in slowing of transit and hindered food digestion. Myenteric neurons synthesize hyaluronan (HA) to form a well-structured perineuronal net, which undergoes derangement when myenteric ganglia homeostasis is perturbed, i.e. during inflammation. In this study we evaluated HA involvement in rat small intestine myenteric plexus after in vivo I/R injury induced by clamping a branch of the superior mesenteric artery for 60 min, followed by 24 h of reperfusion. In some experiments, 4-methylumbelliferone (4-MU, 25 mg/kg), a HA synthesis inhibitor, was intraperitoneally administered to normal (CTR), sham-operated (SH) and I/R animals for 24 h. In longitudinal muscle myenteric plexus (LMMP) whole-mount preparations, HA binding protein staining as well as HA levels were significantly higher in the I/R group, and were reduced after 4-MU treatment. HA synthase 1 and 2 (HAS1 and HAS2) labelled myenteric neurons and mRNA levels in LMMPs increased in the I/R group with respect to CTR, and were reduced by 4-MU. The efficiency of the gastrointestinal transit was significantly reduced in I/R and 4-MU-treated I/R groups with respect to CTR and SH groups. In the 4-MU-treated I/R group gastric emptying was reduced with respect to the CTR, SH and I/R groups. Carbachol (CCh) and electrical field (EFS, 0.1–40 Hz) stimulated contractions and EFS-induced (10 Hz) NANC relaxations were reduced in the I/R group with respect to both CTR and SH groups. After I/R, 4-MU treatment increased EFS contractions towards control values, but did not affect CCh-induced contractions. NANC on-relaxations after I/R were not influenced by 4-MU treatment. Main alterations in the neurochemical coding of both excitatory (tachykinergic) and inhibitory pathways (iNOS, VIPergic) were also observed after I/R, and were influenced by 4-MU administration. Overall, our data suggest that, after an intestinal I/R damage, changes of HA homeostasis in specific myenteric neuron populations may influence the efficiency of the gastrointestinal transit. We cannot exclude that modulation of HA synthesis in these conditions may ameliorate derangement of the enteric motor function preventing, at least in part, the development of dysmotility.
Collapse
Affiliation(s)
- Michela Bistoletti
- Department of Medicine and Surgery, University of Insubria, via H. Dunant 5, Varese, Italy
| | - Annalisa Bosi
- Department of Medicine and Surgery, University of Insubria, via H. Dunant 5, Varese, Italy
| | - Ilaria Caon
- Department of Medicine and Surgery, University of Insubria, via H. Dunant 5, Varese, Italy
| | - Anna Maria Chiaravalli
- Department of Pathology, ASST-Sette Laghi, Ospedale di Circolo Viale L. Borri 57, 21100, Varese, Italy
| | - Paola Moretto
- Department of Medicine and Surgery, University of Insubria, via H. Dunant 5, Varese, Italy
| | - Angelo Genoni
- Department of Medicine and Surgery, University of Insubria, via H. Dunant 5, Varese, Italy
| | - Elisabetta Moro
- Department of Internal Medicine and Therapeutics, Section of Pharmacology, University of Pavia, Pavia, Italy
| | - Evgenia Karousou
- Department of Medicine and Surgery, University of Insubria, via H. Dunant 5, Varese, Italy
| | - Manuela Viola
- Department of Medicine and Surgery, University of Insubria, via H. Dunant 5, Varese, Italy
| | - Francesca Crema
- Department of Internal Medicine and Therapeutics, Section of Pharmacology, University of Pavia, Pavia, Italy
| | - Andreina Baj
- Department of Medicine and Surgery, University of Insubria, via H. Dunant 5, Varese, Italy
| | - Alberto Passi
- Department of Medicine and Surgery, University of Insubria, via H. Dunant 5, Varese, Italy
| | - Davide Vigetti
- Department of Medicine and Surgery, University of Insubria, via H. Dunant 5, Varese, Italy.
| | - Cristina Giaroni
- Department of Medicine and Surgery, University of Insubria, via H. Dunant 5, Varese, Italy.
| |
Collapse
|
50
|
Bosi A, Banfi D, Bistoletti M, Giaroni C, Baj A. Tryptophan Metabolites Along the Microbiota-Gut-Brain Axis: An Interkingdom Communication System Influencing the Gut in Health and Disease. Int J Tryptophan Res 2020; 13:1178646920928984. [PMID: 32577079 PMCID: PMC7290275 DOI: 10.1177/1178646920928984] [Citation(s) in RCA: 105] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Accepted: 05/02/2020] [Indexed: 12/12/2022] Open
Abstract
The ‘microbiota-gut-brain axis’ plays a fundamental role in maintaining host homeostasis, and different immune, hormonal, and neuronal signals participate to this interkingdom communication system between eukaryota and prokaryota. The essential aminoacid tryptophan, as a precursor of several molecules acting at the interface between the host and the microbiota, is fundamental in the modulation of this bidirectional communication axis. In the gut, tryptophan undergoes 3 major metabolic pathways, the 5-HT, kynurenine, and AhR ligand pathways, which may be directly or indirectly controlled by the saprophytic flora. The importance of tryptophan metabolites in the modulation of the gastrointestinal tract is suggested by several preclinical and clinical studies; however, a thorough revision of the available literature has not been accomplished yet. Thus, this review attempts to cover the major aspects on the role of tryptophan metabolites in host-microbiota cross-talk underlaying regulation of gut functions in health conditions and during disease states, with particular attention to 2 major gastrointestinal diseases, such as irritable bowel syndrome (IBS) and inflammatory bowel disease (IBD), both characterized by psychiatric disorders. Research in this area opens the possibility to target tryptophan metabolism to ameliorate the knowledge on the pathogenesis of both diseases, as well as to discover new therapeutic strategies based either on conventional pharmacological approaches or on the use of pre- and probiotics to manipulate the microbial flora.
Collapse
Affiliation(s)
- Annalisa Bosi
- Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Davide Banfi
- Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Michela Bistoletti
- Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Cristina Giaroni
- Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Andreina Baj
- Department of Medicine and Surgery, University of Insubria, Varese, Italy
| |
Collapse
|