1
|
Qian H, Xi Z, Liang M, An R, Gao B, Chen X, Liu Z, Shang Q, Chen G, Xiao L, Liu X. Polygalae Radix Attenuates Methamphetamine-Induced Behavioral Sensitization Through the TrkB/ERK Pathway in the Caudate Putamen of Mice. Neurochem Res 2025; 50:120. [PMID: 40095175 DOI: 10.1007/s11064-025-04368-0] [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: 10/10/2024] [Revised: 02/27/2025] [Accepted: 03/03/2025] [Indexed: 03/19/2025]
Abstract
Methamphetamine (METH) addiction is a chronic brain disorder characterized by intense drug cravings and high relapse rates. Traditional Chinese medicines (TCM) have shown efficacy in treating METH addiction via TrkB/ERK signaling. However, the role of Polygalae Radix (PR), a neuropharmacological active TCM, in METH addiction remains unclear. This study examined the effects of PR (25, 50, and 100 mg/kg) on locomotor activity in mice and its impact on METH-induced behavioral sensitization (BS) at different stages. Western blotting (WB) assessed TrkB and ERK expression across brain regions. PR (25 and 50 mg/kg) alone had no effect on locomotor activity in mice, whereas 100 mg/kg significantly reduced locomotor activity. PR (25 and 50 mg/kg) administered during the development phase inhibited METH-induced locomotor activity, but its administration during the expression phase had no impact. Continuous PR (25 and 50 mg/kg) administration throughout the entire process prevented METH-induced BS in mice. WB analysis revealed that PR alone elevated ERK in the prefrontal cortex, nucleus accumbens (NAc), caudate putamen (CPu), and TrkB in the CPu. During the development phase, PR inhibited METH-induced TrkB/ERK increases in the CPu, whereas, during the expression phase, ERK elevation in the CPu was mitigated. Continuous PR administration blocked METH-induced TrkB/ERK increases in the CPu and ERK levels in the NAc. These findings indicate that PR attenuates METH-induced BS and locomotor activity during the developmental phase through the TrkB/ERK pathway in the CPu, highlighting its therapeutic potential for METH addiction.
Collapse
Affiliation(s)
- Hongyan Qian
- Institute of Forensic Injury, Institute of Forensic Bioevidence, Western China Science and Technology Innovation Harbor, Xi'an Jiaotong University, Xi'an, Shaanxi, PR China
- Department of Forensic Medicine, School of Basic Medical Science, Wenzhou Medical University, Wenzhou, Zhejiang, PR China
| | - Zhijia Xi
- Institute of Forensic Injury, Institute of Forensic Bioevidence, Western China Science and Technology Innovation Harbor, Xi'an Jiaotong University, Xi'an, Shaanxi, PR China
| | - Min Liang
- Institute of Forensic Injury, Institute of Forensic Bioevidence, Western China Science and Technology Innovation Harbor, Xi'an Jiaotong University, Xi'an, Shaanxi, PR China
| | - Ran An
- Institute of Forensic Injury, Institute of Forensic Bioevidence, Western China Science and Technology Innovation Harbor, Xi'an Jiaotong University, Xi'an, Shaanxi, PR China
| | - Baoyao Gao
- Institute of Forensic Injury, Institute of Forensic Bioevidence, Western China Science and Technology Innovation Harbor, Xi'an Jiaotong University, Xi'an, Shaanxi, PR China
| | - Xingyao Chen
- Institute of Forensic Injury, Institute of Forensic Bioevidence, Western China Science and Technology Innovation Harbor, Xi'an Jiaotong University, Xi'an, Shaanxi, PR China
| | - Zijun Liu
- Institute of Forensic Injury, Institute of Forensic Bioevidence, Western China Science and Technology Innovation Harbor, Xi'an Jiaotong University, Xi'an, Shaanxi, PR China
| | - Qing Shang
- Institute of Forensic Injury, Institute of Forensic Bioevidence, Western China Science and Technology Innovation Harbor, Xi'an Jiaotong University, Xi'an, Shaanxi, PR China
- Department of Forensic Medicine, School of Basic Medical Science, Wenzhou Medical University, Wenzhou, Zhejiang, PR China
| | - Gang Chen
- Institute of Forensic Injury, Institute of Forensic Bioevidence, Western China Science and Technology Innovation Harbor, Xi'an Jiaotong University, Xi'an, Shaanxi, PR China
- Department of Forensic Medicine, School of Basic Medical Science, Wenzhou Medical University, Wenzhou, Zhejiang, PR China
| | - Lei Xiao
- National Narcotics Laboratory Shaanxi Regional Center, Xi'an, Shaanxi, PR China.
| | - Xinshe Liu
- Institute of Forensic Injury, Institute of Forensic Bioevidence, Western China Science and Technology Innovation Harbor, Xi'an Jiaotong University, Xi'an, Shaanxi, PR China.
- College of Medicine & Forensics, Xi'an Jiaotong University Health Science Center, Yanta Road W.76, Xi'an, Shaanxi, 710061, PR China.
| |
Collapse
|
2
|
Bernardo HT, Lodetti G, de Farias ACS, de Pieri Pickler K, Baldin SL, Dondossola ER, Rico EP. Naltrexone Alters Neurochemical and Behavioral Parameters in a Zebrafish Model of Repeated Alcohol Exposure. Neurochem Res 2025; 50:97. [PMID: 39920352 DOI: 10.1007/s11064-025-04349-3] [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: 10/01/2024] [Revised: 01/04/2025] [Accepted: 01/27/2025] [Indexed: 02/09/2025]
Abstract
Between the neurotransmission systems modulated by alcohol, the opioid system has been receiving attention in studies that seek to understand its relationship to the effects of addictive substances and different neuropsychiatric disorders. The use of naltrexone stands out in determining the mechanisms of the opioid system, as it acts as an opioid antagonist and consequently generates neurochemical responses. This study aimed to evaluate the pharmacological modulation of opioids on behavioral and neurobiological aspects in adult zebrafish submitted to the protocol of repeated exposure to ethanol and treated with naltrexone. Opioid modulation using naltrexone has been shown to modulate anxiety-like behavior, presenting anxiolytic properties in isolation, in addition to reversing the anxiogenic effect of ethanol through the Novel tank and Light/dark test. Naltrexone increased serotonin and dopamine levels, while ethanol antagonized these effects. In contrast, the interaction between ethanol and naltrexone raised noradrenaline levels. Naltrexone altered glutamate levels, however, ethanol reversed it. Ethanol acted on glutamate transporters increasing their activities, while naltrexone treatment reduced activities. No significant results were found in the pro-oxidant parameters, however, ethanol reduced SOD activity while naltrexone reversed. The same occurred in CAT activity. Also, naltrexone up-regulated the expression of genes related to the dopaminergic, glutamatergic, and opioid systems. The genes used as markers of the inflammatory process and glial activity were modulated by ethanol and together with naltrexone, respectively. Taken together, our findings reinforce the importance of opioid signaling on biochemical and molecular bases related to neuropsychiatric behaviors and diseases, such as anxiety and substance dependence.
Collapse
Affiliation(s)
- Henrique Teza Bernardo
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Guilherme Lodetti
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Ana Caroline Salvador de Farias
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Karolyne de Pieri Pickler
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Samira Leila Baldin
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Eduardo Ronconi Dondossola
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Eduardo Pacheco Rico
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil.
| |
Collapse
|
3
|
Barkus A, Baltrūnienė V, Baušienė J, Baltrūnas T, Barkienė L, Kazlauskaitė P, Baušys A. The Gut-Brain Axis in Opioid Use Disorder: Exploring the Bidirectional Influence of Opioids and the Gut Microbiome-A Comprehensive Review. Life (Basel) 2024; 14:1227. [PMID: 39459527 PMCID: PMC11508959 DOI: 10.3390/life14101227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 09/15/2024] [Accepted: 09/20/2024] [Indexed: 10/28/2024] Open
Abstract
Opioid Use Disorder is a chronic condition characterized by compulsive opioid use despite negative consequences, resulting in severe health risks such as overdose and contraction of infectious diseases. High dropout rates in opioid agonist therapy highlight the need for more effective relapse prevention strategies. Animal and clinical studies indicate that opioids influence gut microbiota, which in turn plays a critical role in addiction development and alters behavioral responses to opioids. This study provides a comprehensive review of the literature on the effects of opioids on the gut microbiome and explores the potential of microbiome manipulation as a therapeutic target in opioid addiction.
Collapse
Affiliation(s)
- Artūras Barkus
- Department of Pathology and Forensic Medicine, Institute of Biomedical Sciences, Faculty of Medicine, Vilnius University, 03101 Vilnius, Lithuania
- Laboratory of Experimental Surgery and Oncology, Faculty of Medicine, Vilnius University, 03101 Vilnius, Lithuania
| | - Vaida Baltrūnienė
- Department of Pathology and Forensic Medicine, Institute of Biomedical Sciences, Faculty of Medicine, Vilnius University, 03101 Vilnius, Lithuania
- Laboratory of Experimental Surgery and Oncology, Faculty of Medicine, Vilnius University, 03101 Vilnius, Lithuania
| | - Justė Baušienė
- Department of Pathology and Forensic Medicine, Institute of Biomedical Sciences, Faculty of Medicine, Vilnius University, 03101 Vilnius, Lithuania
- Laboratory of Experimental Surgery and Oncology, Faculty of Medicine, Vilnius University, 03101 Vilnius, Lithuania
| | - Tomas Baltrūnas
- Department of Pathology and Forensic Medicine, Institute of Biomedical Sciences, Faculty of Medicine, Vilnius University, 03101 Vilnius, Lithuania
- Laboratory of Experimental Surgery and Oncology, Faculty of Medicine, Vilnius University, 03101 Vilnius, Lithuania
| | - Lina Barkienė
- Department of Pathology and Forensic Medicine, Institute of Biomedical Sciences, Faculty of Medicine, Vilnius University, 03101 Vilnius, Lithuania
- Laboratory of Experimental Surgery and Oncology, Faculty of Medicine, Vilnius University, 03101 Vilnius, Lithuania
| | - Paulina Kazlauskaitė
- Department of Pathology and Forensic Medicine, Institute of Biomedical Sciences, Faculty of Medicine, Vilnius University, 03101 Vilnius, Lithuania
- Laboratory of Experimental Surgery and Oncology, Faculty of Medicine, Vilnius University, 03101 Vilnius, Lithuania
| | - Augustinas Baušys
- Department of Pathology and Forensic Medicine, Institute of Biomedical Sciences, Faculty of Medicine, Vilnius University, 03101 Vilnius, Lithuania
- Laboratory of Experimental Surgery and Oncology, Faculty of Medicine, Vilnius University, 03101 Vilnius, Lithuania
| |
Collapse
|
4
|
Yu R, Kong DL, Liao C, Yu YJ, He ZW, Wang Y. Natural products as the therapeutic strategies for addiction. Biomed Pharmacother 2024; 175:116687. [PMID: 38701568 DOI: 10.1016/j.biopha.2024.116687] [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: 01/29/2024] [Revised: 04/25/2024] [Accepted: 04/29/2024] [Indexed: 05/05/2024] Open
Abstract
World Drug Report 2023 concluded that 296 million people abused drugs, 39.5 million became addiction and 494,000 died as a direct or indirect result of addiction. Addiction has become a growing problem that affects individuals, their families, societies, countries and even the world. However, treatment for addiction is only limited to some developed countries because of the high cost, difficult implementation, and time consuming. Therefore, there is an urgent need to develop a low-cost, effective drug for the development of addiction treatment in more countries, which is essential for the stability and sustainable development of the world. In this review, it provided an overview of the abuse of common addictive drugs, related disorders, and current therapeutic regimen worldwide, and summarized the mechanisms of drug addiction as reward circuits, neuroadaptation and plasticity, cognitive decision-making, genetics, and environment. According to their chemical structure, 43 natural products and 5 herbal combinations with potential to treat addiction were classified, and their sources, pharmacological effects and clinical trials were introduced. It was also found that mitragine, ibogine, L-tetrahydropalmatine and crocin had greater potential for anti-addiction.
Collapse
Affiliation(s)
- Rui Yu
- Department of Clinical Pharmacology, School of Pharmacy, China Medical University, Shenyang, Liaoning, China
| | - De-Lei Kong
- Department of Respiratory and Critical Care Medicine, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Cai Liao
- Department of Clinical Pharmacology, School of Pharmacy, China Medical University, Shenyang, Liaoning, China
| | - Ya-Jie Yu
- Department of Clinical Pharmacology, School of Pharmacy, China Medical University, Shenyang, Liaoning, China
| | - Zhen-Wei He
- Department of Neurology, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning, China.
| | - Yun Wang
- Department of Clinical Pharmacology, School of Pharmacy, China Medical University, Shenyang, Liaoning, China.
| |
Collapse
|
5
|
Adedara IA, Mohammed KA, Canzian J, Ajayi BO, Farombi EO, Emanuelli T, Rosemberg DB, Aschner M. Utility of zebrafish-based models in understanding molecular mechanisms of neurotoxicity mediated by the gut-brain axis. ADVANCES IN NEUROTOXICOLOGY 2024; 11:177-208. [PMID: 38741945 PMCID: PMC11090488 DOI: 10.1016/bs.ant.2024.02.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
The gut microbes perform several beneficial functions which impact the periphery and central nervous systems of the host. Gut microbiota dysbiosis is acknowledged as a major contributor to the development of several neuropsychiatric and neurological disorders including bipolar disorder, depression, anxiety, Parkinson's disease, Alzheimer's disease, attention deficit hyperactivity disorder, and autism spectrum disorder. Thus, elucidation of how the gut microbiota-brain axis plays a role in health and disease conditions is a potential novel approach to prevent and treat brain disorders. The zebrafish (Danio rerio) is an invaluable vertebrate model that possesses conserved brain and intestinal features with those of humans, thus making zebrafish a valued model to investigate the interplay between the gut microbiota and host health. This chapter describes current findings on the utility of zebrafish in understanding molecular mechanisms of neurotoxicity mediated via the gut microbiota-brain axis. Specifically, it highlights the utility of zebrafish as a model organism for understanding how anthropogenic chemicals, pharmaceuticals and bacteria exposure affect animals and human health via the gut-brain axis.
Collapse
Affiliation(s)
- Isaac A. Adedara
- Department of Food Science and Technology, Center of Rural Sciences, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Khadija A. Mohammed
- Laboratory of Experimental Neuropsychobiology, Department of Biochemistry and Molecular Biology, Federal University of Santa Maria, Santa Maria, RS, Brazil
- Graduate Program in Biological Sciences: Toxicological Biochemistry, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Julia Canzian
- Laboratory of Experimental Neuropsychobiology, Department of Biochemistry and Molecular Biology, Federal University of Santa Maria, Santa Maria, RS, Brazil
- Graduate Program in Biological Sciences: Toxicological Biochemistry, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Babajide O. Ajayi
- Graduate Program in Biological Sciences: Toxicological Biochemistry, Federal University of Santa Maria, Santa Maria, RS, Brazil
- Department of Chemical Sciences, Faculty of Natural Sciences, Ajayi Crowther University, Oyo, Nigeria
| | - Ebenezer O. Farombi
- Drug Metabolism and Toxicology Research Laboratories, Department of Biochemistry, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Tatiana Emanuelli
- Department of Food Science and Technology, Center of Rural Sciences, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Denis B. Rosemberg
- Laboratory of Experimental Neuropsychobiology, Department of Biochemistry and Molecular Biology, Federal University of Santa Maria, Santa Maria, RS, Brazil
- Graduate Program in Biological Sciences: Toxicological Biochemistry, Federal University of Santa Maria, Santa Maria, RS, Brazil
- The International Zebrafish Neuroscience Research Consortium (ZNRC), Slidell, LA, United States
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, United States
| |
Collapse
|
6
|
Pamanji R, Kumareshan TN, Priya S L, Sivan G, Selvin J. Exploring the impact of antibiotics, microplastics, nanoparticles, and pesticides on zebrafish gut microbiomes: Insights into composition, interactions, and health implications. CHEMOSPHERE 2024; 349:140867. [PMID: 38048833 DOI: 10.1016/j.chemosphere.2023.140867] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 11/28/2023] [Accepted: 11/29/2023] [Indexed: 12/06/2023]
Abstract
This review addresses the impact of various chemical entities like pesticides, antibiotics, nanoparticles and microplastic on gut microbiota of zebrafish. Gut microbiota plays a vital role in metabolic regulation in every organism. As majority of metabolic pathways coordinated by microbiota, small alterations associated with mild to serious outcomes. Because of their unstoppable usage in day-to-day life, the present-day research on gut microbiota is mostly comprising aforementioned chemicals. It is better to understand how gut microbiome is dysbiosed by various environmental factors, to keep our microbiota safe. We tried to delineate the natural flora of zebrafish gut microbiome and the metabolic and other pathways associated and what are the common flora that was dysbiosed during the treatment. Based on the existing literature, we reviewed pesticides like Imazalil, Difenoconazole, Chlorpyrifos, Metamifop, Carbendazim, Imidacloprid, Phoxim, Niclosamide, Dieldrin, and antibiotics like Oxytetracycline, Enrofloxacin, Florfenicol, Sulfamethoxazole, Tetracycline, Streptomycin, Doxycycline, and in the category of nanoparticles, Titanium dioxide nanoparticles (nTiO2), Abalone viscera hydrolysates decorated silver nanoparticles (AVH-AgNPs), Lead-halide perovskite nanoparticles (LHP NPs), Copper nanoparticles (Cu-NPs), silver nanoparticles (Ag-NPs) and microplastic types like polyethylene and polystyrene microplastic. Other studies with miscellaneous chemical entities on zebrafish gut microbiome include Ferulic acid, Polychlorinated biphenyls, Cadmium, Disinfection by-products, Triclosan, microcystin-LR, Fluoride, and Amitriptyline.
Collapse
Affiliation(s)
- Rajesh Pamanji
- Department of Microbiology, Pondicherry University, Puducherry, 605014, India.
| | - T N Kumareshan
- Department of Microbiology, Pondicherry University, Puducherry, 605014, India
| | - Lakshmi Priya S
- Department of Microbiology, Pondicherry University, Puducherry, 605014, India
| | - Gisha Sivan
- Division Medical Research, SRM Institute of Science and Technology, Chennai, 603203, India
| | - Joseph Selvin
- Department of Microbiology, Pondicherry University, Puducherry, 605014, India
| |
Collapse
|
7
|
Liu W, Liu L, Deng Z, Liu R, Ma T, Xin Y, Xie Y, Zhou Y, Tang Y. Associations between impulsivity and fecal microbiota in individuals abstaining from methamphetamine. CNS Neurosci Ther 2024; 30:e14580. [PMID: 38421126 PMCID: PMC10851322 DOI: 10.1111/cns.14580] [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: 08/10/2023] [Revised: 12/13/2023] [Accepted: 12/15/2023] [Indexed: 03/02/2024] Open
Abstract
INTRODUCTION Methamphetamine (MA) abuse is a major public problem, and impulsivity is both a prominent risk factor and a consequence of addiction. Hence, clarifying the biological mechanism of impulsivity may facilitate the understanding of addiction to MA. The microbiota-gut-brain axis was suggested to underlie a biological mechanism of impulsivity induced by MA. METHODS We therefore recruited 62 MA addicts and 50 healthy controls (HCs) to investigate the alterations in impulsivity and fecal microbiota and the associations between them in the MA group. Thereafter, 25 MA abusers who abstained from MA for less than 3 months were followed up for 2 months to investigate the relationship between impulsivity and microbiota as abstinence became longer. 16S rRNA sequencing was conducted for microbiota identification. RESULTS Elevated impulsivity and dysbiosis characterized by an increase in opportunistic pathogens and a decrease in probiotics were identified in MA abusers, and both the increased impulsivity and disrupted microbiota tended to recover after longer abstinence from MA. Impulsivity was related to microbiota, and the effect of MA abuse on impulsivity was mediated by microbiota. CONCLUSION Our findings potentially highlighted the importance of abstention and implicated the significant role of the microbiota-gut-brain axis in the interrelationship between microbiota and behaviors, as well as the potential of microbiota as a target for intervention of impulsivity.
Collapse
Affiliation(s)
- Wen Liu
- Department of PsychiatryThe First Hospital of China Medical UniversityShenyangLiaoningPR China
| | - Linzi Liu
- Department of PsychiatryThe First Hospital of China Medical UniversityShenyangLiaoningPR China
| | - Zijing Deng
- Department of PsychiatryThe First Hospital of China Medical UniversityShenyangLiaoningPR China
| | - Ruina Liu
- Department of PsychiatryThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShanxiPR China
| | - Tao Ma
- Department of PsychiatryThe First Hospital of China Medical UniversityShenyangLiaoningPR China
| | - Yide Xin
- Department of PsychiatryThe First Hospital of China Medical UniversityShenyangLiaoningPR China
| | - Yu Xie
- Department of PsychiatryThe First Hospital of China Medical UniversityShenyangLiaoningPR China
| | - Yifang Zhou
- Department of PsychiatryThe First Hospital of China Medical UniversityShenyangLiaoningPR China
| | - Yanqing Tang
- Department of PsychiatryShengjing Hospital of China Medical UniversityShenyangLiaoningPR China
| |
Collapse
|
8
|
Chen J, Guo P, Han M, Chen K, Qin J, Yang F. Cognitive protection of sinomenine in type 2 diabetes mellitus through regulating the EGF/Nrf2/HO-1 signaling, the microbiota-gut-brain axis, and hippocampal neuron ferroptosis. Phytother Res 2023; 37:3323-3341. [PMID: 37036428 DOI: 10.1002/ptr.7807] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 02/26/2023] [Accepted: 03/05/2023] [Indexed: 04/11/2023]
Abstract
Recent years have witnessed a growing research interest in traditional Chinese medicine as a neuroprotective nutrient in the management of diabetic cognitive dysfunction. However, the underlying molecular mechanisms of sinomenine in mediating ferroptosis of hippocampal neurons have been poorly understood. This study sought to decipher the potential effect and molecular mechanism of sinomenine in the cognitive dysfunction following type 2 diabetes mellitus (T2DM). Multi-omics analysis was conducted to identify the microbiota-gut-brain axis in T2DM patient samples obtained from the publicly available database. In HT-22 cells, erastin was utilized to create a ferroptosis model, and streptozotocin was injected intraperitoneally to create a rat model of DM. It was noted that intestinal flora imbalance occurred in patients with T2DM-associated cognitive dysfunction. Sinomenine could reduce Erastin-induced hippocampus neuronal ferroptosis by increasing EGF expression. EGF protected hippocampal neurons against ferroptosis by activating the Nrf2/HO-1 signaling pathway. Furthermore, in vivo results confirmed that sinomenine blocked ferroptosis of hippocampal neurons and alleviated cognitive dysfunction in T2DM rats. Collectively, these results suggest that sinomenine confers neuroprotective effects by curtailing hippocampal neuron ferroptosis via the EGF/Nrf2/HO-1 signaling and microbiota-gut-brain axis. It may be a candidate for the treatment of diabetic cognitive dysfunction.
Collapse
Affiliation(s)
- Ji Chen
- Department of Endocrinology, The First People's Hospital of Huaihua, Huaihua, P.R. China
| | - Peng Guo
- Department of Anesthesiology, The First People's Hospital of Huaihua, Huaihua, P.R. China
| | - Mingming Han
- Department of Anesthesiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China|, Hefei, P.R. China
| | - Kemin Chen
- Department of Anesthesiology, The First Affiliated Hospital, University of South China, Hengyang, P.R. China
| | - Jie Qin
- Department of Anesthesiology, The First Affiliated Hospital, University of South China, Hengyang, P.R. China
| | - Fengrui Yang
- Department of Anesthesiology, The First People's Hospital of Huaihua, Huaihua, P.R. China
- Department of Anesthesiology, The First Affiliated Hospital, University of South China, Hengyang, P.R. China
| |
Collapse
|
9
|
Jiang ZM, Zeng SL, Huang TQ, Lin Y, Wang FF, Gao XJ, Li J, Li P, Liu EH. Sinomenine ameliorates rheumatoid arthritis by modulating tryptophan metabolism and activating aryl hydrocarbon receptor via gut microbiota regulation. Sci Bull (Beijing) 2023:S2095-9273(23)00410-3. [PMID: 37422372 DOI: 10.1016/j.scib.2023.06.027] [Citation(s) in RCA: 57] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Revised: 04/19/2023] [Accepted: 05/19/2023] [Indexed: 07/10/2023]
Abstract
Gut microbiota dysbiosis is associated with the development of rheumatoid arthritis (RA). Sinomenine (SIN) is an effective immunosuppressive and anti-inflammatory drug used for treating RA, but how SIN regulates gut microbiota to alleviate RA remains underexplored. To identify the critical gut microbial species and microbial metabolites associated with the RA-protective effects of SIN, the microbiota-dependent anti-RA effects of SIN were assessed by 16S rRNA gene sequencing, antibiotic treatment, and fecal microbiota transplantation. Metabolomics analysis, transcriptional analysis, and targeted bacteria/metabolites gavage were conducted to explore how SIN regulates gut microbiota to reduce the severity of RA. SIN could restore intestinal microbial balance by mainly modulating the abundance of Lactobacillus, and significantly relieve collagen-induced arthritis (CIA) symptoms in a gut microbiota-dependent manner. SIN significantly elevated microbial tryptophan metabolites indole-3-acrylic acid (IA), indole-3-propionic acid (IPA), and indole-3-acetic acid (IAA). Tryptophan metabolites supplementation could activate aryl hydrocarbon receptor (AhR) and regulate Th17/Treg balance in CIA rats. Intriguingly, SIN relieved the arthritis symptoms involving the enrichment of two beneficial anti-CIA Lactobacillus species, L. paracasei and L. casei by mono-colonization. The promising therapeutic function of SIN was mostly attributed to the activation of AhR by explicitly targeting the Lactobacillus and microbial tryptophan metabolites. The intestinal bacterium L. paracasei and L. casei may be used to reduce the severity of CIA.
Collapse
Affiliation(s)
- Zheng-Meng Jiang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210003, China
| | - Su-Ling Zeng
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210003, China
| | - Tian-Qing Huang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210003, China
| | - Yang Lin
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210003, China
| | - Fang-Fang Wang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210003, China
| | - Xing-Jiao Gao
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210003, China
| | - Jing Li
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210003, China.
| | - Ping Li
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210003, China.
| | - E-Hu Liu
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210003, China.
| |
Collapse
|
10
|
Sree Kumar H, Wisner AS, Refsnider JM, Martyniuk CJ, Zubcevic J. Small fish, big discoveries: zebrafish shed light on microbial biomarkers for neuro-immune-cardiovascular health. Front Physiol 2023; 14:1186645. [PMID: 37324381 PMCID: PMC10267477 DOI: 10.3389/fphys.2023.1186645] [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: 03/15/2023] [Accepted: 05/22/2023] [Indexed: 06/17/2023] Open
Abstract
Zebrafish (Danio rerio) have emerged as a powerful model to study the gut microbiome in the context of human conditions, including hypertension, cardiovascular disease, neurological disorders, and immune dysfunction. Here, we highlight zebrafish as a tool to bridge the gap in knowledge in linking the gut microbiome and physiological homeostasis of cardiovascular, neural, and immune systems, both independently and as an integrated axis. Drawing on zebrafish studies to date, we discuss challenges in microbiota transplant techniques and gnotobiotic husbandry practices. We present advantages and current limitations in zebrafish microbiome research and discuss the use of zebrafish in identification of microbial enterotypes in health and disease. We also highlight the versatility of zebrafish studies to further explore the function of human conditions relevant to gut dysbiosis and reveal novel therapeutic targets.
Collapse
Affiliation(s)
- Hemaa Sree Kumar
- Department of Physiology and Pharmacology, University of Toledo, Toledo, OH, United States
- Department of Neuroscience and Neurological Disorders, University of Toledo, Toledo, OH, United States
| | - Alexander S. Wisner
- Department of Medicinal and Biological Chemistry, University of Toledo, Toledo, OH, United States
- Center for Drug Design and Development, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, OH, United States
| | - Jeanine M. Refsnider
- Department of Environmental Sciences, University of Toledo, Toledo, OH, United States
| | - Christopher J. Martyniuk
- Department of Physiological Sciences, College of Veterinary Medicine, University of Florida, Gainesville, OH, United States
| | - Jasenka Zubcevic
- Department of Physiology and Pharmacology, University of Toledo, Toledo, OH, United States
| |
Collapse
|
11
|
He L, Yang BZ, Ma YJ, Wen L, Liu F, Zhang XJ, Liu TQ. Differences in clinical features and gut microbiota between individuals with methamphetamine casual use and methamphetamine use disorder. Front Cell Infect Microbiol 2023; 13:1103919. [PMID: 36909722 PMCID: PMC9996337 DOI: 10.3389/fcimb.2023.1103919] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 01/30/2023] [Indexed: 02/25/2023] Open
Abstract
Background The transition from methamphetamine (MA) casual use (MCU) to compulsive use is enigmatic as some MA users can remain in casual use, but some cannot. There is a knowledge gap if gut microbiota (GM) play a role in differing MCU from MA use disorder (MUD). We aimed to investigate the clinical features and GM differences between individuals with MCU and MUD. Method We recruited two groups of MA users -MCU and MUD - and matched them according to age and body mass index (n=21 in each group). Participants were accessed using the Semi-Structured Assessment for Drug Dependence and Alcoholism, and their fecal samples were undergone 16S ribosomal DNA sequencing. We compared the hosts' clinical features and GM diversity, composition, and structure (represented by enterotypes) between the two groups. We have identified differential microbes between the two groups and performed network analyses connecting GM and the clinical traits. Result Compared with the casual users, individuals with MUD had higher incidences of MA-induced neuropsychiatric symptoms (e.g., paranoia, depression) and withdrawal symptoms (e.g., fatigue, drowsiness, and increased appetite), as well as stronger cravings for and intentions to use MA, and increased MA tolerance. The GM diversity showed no significant differences between the two groups, but four genera (Halomonas, Clostridium, Devosia, and Dorea) were enriched in the individuals with MUD (p<0.05). Three distinct enterotypes were identified in all MA users, and Ruminococcus-driven enterotype 2 was dominant in individuals with MUD compared to the MCU (61.90% vs. 28.60%, p=0.03). Network analysis shows that Devosia is the hub genus (hub index = 0.75), which is not only related to the counts of the MUD diagnostic criteria (ρ=0.40; p=0.01) but also to the clinical features of MA users such as reduced social activities (ρ=0.54; p<0.01). Devosia is also associated with the increased intention to use MA (ρ=0.48; p<0.01), increased MA tolerance (ρ=0.38; p=0.01), craving for MA (ρ=0.37; p=0.01), and MA-induced withdrawal symptoms (p<0.05). Conclusion Our findings suggest that Ruminococcus-driven enterotype 2 and the genera Devosia might be two influential factors that differentiate MA casual use from MUD, but further studies are warranted.
Collapse
Affiliation(s)
- Li He
- Department of Psychiatry, and National Clinical Research Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Bao-Zhu Yang
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, United States
| | - Yue-Jiao Ma
- Department of Psychiatry, and National Clinical Research Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Li Wen
- Department of Internal Medicine, Section of Endocrinology & Core Laboratory of Yale Center for Clinical Investigation, Yale University School of Medicine, New Haven, CT, United States
| | - Feng Liu
- Compulsory Detoxification Center of Changsha Public Security Bureau, Changsha, Hunan, China
| | - Xiao-Jie Zhang
- Department of Psychiatry, and National Clinical Research Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Tie-Qiao Liu
- Department of Psychiatry, and National Clinical Research Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| |
Collapse
|
12
|
Understanding CNS Effects of Antimicrobial Drugs Using Zebrafish Models. Vet Sci 2023; 10:vetsci10020096. [PMID: 36851400 PMCID: PMC9964482 DOI: 10.3390/vetsci10020096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 01/18/2023] [Accepted: 01/20/2023] [Indexed: 01/31/2023] Open
Abstract
Antimicrobial drugs represent a diverse group of widely utilized antibiotic, antifungal, antiparasitic and antiviral agents. Their growing use and clinical importance necessitate our improved understanding of physiological effects of antimicrobial drugs, including their potential effects on the central nervous system (CNS), at molecular, cellular, and behavioral levels. In addition, antimicrobial drugs can alter the composition of gut microbiota, and hence affect the gut-microbiota-brain axis, further modulating brain and behavioral processes. Complementing rodent studies, the zebrafish (Danio rerio) emerges as a powerful model system for screening various antimicrobial drugs, including probing their putative CNS effects. Here, we critically discuss recent evidence on the effects of antimicrobial drugs on brain and behavior in zebrafish, and outline future related lines of research using this aquatic model organism.
Collapse
|
13
|
Hong H, Lu X, Lu Q, Huang C, Cui Z. Potential therapeutic effects and pharmacological evidence of sinomenine in central nervous system disorders. Front Pharmacol 2022; 13:1015035. [PMID: 36188580 PMCID: PMC9523510 DOI: 10.3389/fphar.2022.1015035] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 09/05/2022] [Indexed: 11/30/2022] Open
Abstract
Sinomenine is a natural compound extracted from the medicinal plant Sinomenium acutum. Its supplementation has been shown to present benefits in a variety of animal models of central nervous system (CNS) disorders, such as cerebral ischemia, intracerebral hemorrhage, traumatic brain injury (TBI), Alzheimer’s disease (AD), Parkinson’s disease (PD), epilepsy, depression, multiple sclerosis, morphine tolerance, and glioma. Therefore, sinomenine is now considered a potential agent for the prevention and/or treatment of CNS disorders. Mechanistic studies have shown that inhibition of oxidative stress, microglia- or astrocyte-mediated neuroinflammation, and neuronal apoptosis are common mechanisms for the neuroprotective effects of sinomenine. Other mechanisms, including activation of nuclear factor E2-related factor 2 (Nrf2), induction of autophagy in response to inhibition of protein kinase B (Akt)-mammalian target of rapamycin (mTOR), and activation of cyclic adenosine monophosphate-response element-binding protein (CREB) and brain-derived neurotrophic factor (BDNF), may also mediate the anti-glioma and neuroprotective effects of sinomenine. Sinomenine treatment has also been shown to enhance dopamine receptor D2 (DRD2)-mediated nuclear translocation of αB-crystallin (CRYAB) in astrocytes, thereby suppressing neuroinflammation via inhibition of Signal Transducer and Activator of Transcription 3 (STAT3). In addition, sinomenine supplementation can suppress N-methyl-D-aspartate (NMDA) receptor-mediated Ca2+ influx and induce γ-aminobutyric acid type A (GABAA) receptor-mediated Cl− influx, each of which contributes to the improvement of morphine dependence and sleep disturbance. In this review, we outline the pharmacological effects and possible mechanisms of sinomenine in CNS disorders to advance the development of sinomenine as a new drug for the treatment of CNS disorders.
Collapse
Affiliation(s)
- Hongxiang Hong
- Department of Spine Surgery, The Second Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - Xu Lu
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, Jiangsu, China
| | - Qun Lu
- Department of Pharmacy, Nantong Third Hospital Affiliated to Nantong University, Nantong, Jiangsu, China
| | - Chao Huang
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, Jiangsu, China
| | - Zhiming Cui
- Department of Spine Surgery, The Second Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
- *Correspondence: Zhiming Cui,
| |
Collapse
|
14
|
Chen Z, Lin Y, Zhou Q, Xiao S, Li C, Lin R, Li J, Chen Y, Luo C, Mo Z. Ginsenoside Rg1 mitigates morphine dependence via regulation of gut microbiota, tryptophan metabolism, and serotonergic system function. Biomed Pharmacother 2022; 150:112935. [PMID: 35447543 DOI: 10.1016/j.biopha.2022.112935] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 04/04/2022] [Accepted: 04/06/2022] [Indexed: 11/02/2022] Open
Abstract
BACKGROUND Morphine dependence, a devastating neuropsychiatric condition, may be closely associated with gut microbiota dysbiosis. Ginsenoside Rg1 (Rg1), an active ingredient extracted from the roots of Panax ginseng C.A. Meyer, has potential health-promoting effects on the nervous system. However, its role in substance use disorders remains unclear. Here, we explored the potential modulatory roles of Rg1 in protection against morphine dependence. METHODS Conditioned place preference (CPP) was used for establishing a murine model of morphine dependence. 16S rRNA gene sequencing and metabolomics were performed for microbial and metabolite analysis. Molecular analysis was tested for evaluating the host serum and brain responses. RESULTS Rg1 prevented morphine-induced CPP in mice. The 16S rRNA gene-based microbiota analysis demonstrated that Rg1 ameliorated morphine-induced gut microbiota dysbiosis, specifically for Bacteroidetes. Moreover, Rg1 also inhibited gut microbiota-derived tryptophan metabolism and reduced the serotonin, 5-hydroxytryptamine receptor 1B (5-HTR1B), and 5-hydroxytryptamine receptor 2 A (5-HTR2A) levels. However, the Rg1-induced amelioration of CPP was not observed in mice when their gut microbiome was depleted by non-absorbable antibiotics. Subsequently, gavage with Bacteroides vulgatus increased the abundance of Bacteroidetes. B. vulgatus supplementation synergistically enhanced Rg1-alleviated morphine-induced CPP in mice with microbiome knockdown. Co-treatment with B. vulgatus and Rg1 produced suppressive effects against morphine dependency by inhibiting tryptophan metabolism and reducing the serotonin and 5-HTR1B/5-HTR2A levels. CONCLUSIONS The gut microbiota-tryptophan metabolism-serotonin plays an important role in gut-brain signaling in morphine disorders, which may represent a novel approach for drug dependence treatment via manipulation of the gut microbial composition and tryptophan metabolite.
Collapse
Affiliation(s)
- Zhijie Chen
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Yingbo Lin
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China; Department of Pharmacy, Jiangmen Central Hospital, Jiangmen, China
| | - Qichun Zhou
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Shilin Xiao
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Chan Li
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Rukun Lin
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Jing Li
- Central Laboratory, Southern Medical University, Guangzhou, China
| | - Yifei Chen
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China; College of Pharmacy, Guilin Medical University, Guilin, China
| | - Chaohua Luo
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China.
| | - Zhixian Mo
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China; Department of Chinese Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
15
|
Abstract
This paper is the forty-third consecutive installment of the annual anthological review of research concerning the endogenous opioid system, summarizing articles published during 2020 that studied the behavioral effects of molecular, pharmacological and genetic manipulation of opioid peptides and receptors as well as effects of opioid/opiate agonists and antagonists. The review is subdivided into the following specific topics: molecular-biochemical effects and neurochemical localization studies of endogenous opioids and their receptors (1), the roles of these opioid peptides and receptors in pain and analgesia in animals (2) and humans (3), opioid-sensitive and opioid-insensitive effects of nonopioid analgesics (4), opioid peptide and receptor involvement in tolerance and dependence (5), stress and social status (6), learning and memory (7), eating and drinking (8), drug abuse and alcohol (9), sexual activity and hormones, pregnancy, development and endocrinology (10), mental illness and mood (11), seizures and neurologic disorders (12), electrical-related activity and neurophysiology (13), general activity and locomotion (14), gastrointestinal, renal and hepatic functions (15), cardiovascular responses (16), respiration and thermoregulation (17), and immunological responses (18).
Collapse
Affiliation(s)
- Richard J Bodnar
- Department of Psychology and Neuropsychology Doctoral Sub-Program, Queens College, City University of New York, Flushing, NY, 11367, United States.
| |
Collapse
|
16
|
Li JS, Su SL, Xu Z, Zhao LH, Fan RY, Guo JM, Qian DW, Duan JA. Potential roles of gut microbiota and microbial metabolites in chronic inflammatory pain and the mechanisms of therapy drugs. Ther Adv Chronic Dis 2022; 13:20406223221091177. [PMID: 35924009 PMCID: PMC9340317 DOI: 10.1177/20406223221091177] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 03/15/2022] [Indexed: 01/21/2023] Open
Abstract
Observational findings achieved that gut microbes mediate human metabolic health
and disease risk. The types of intestinal microorganisms depend on the intake of
food and drugs and are also related to their metabolic level and genetic
factors. Recent studies have shown that chronic inflammatory pain is closely
related to intestinal microbial homeostasis. Compared with the normal intestinal
flora, the composition of intestinal flora in patients with chronic inflammatory
pain had significant changes in Actinomycetes,
Firmicutes, Bacteroidetes, etc. At the
same time, short-chain fatty acids and amino acids, the metabolites of
intestinal microorganisms, can regulate neural signal molecules and signaling
pathways, thus affecting the development trend of chronic inflammatory pain.
Glucocorticoids and non-steroidal anti-inflammatory drugs in the treatment of
chronic inflammatory pain, the main mechanism is to affect the secretion of
inflammatory factors and the abundance of intestinal bacteria. This article
reviews the relationship between intestinal microorganisms and their metabolites
on chronic inflammatory pain and the possible mechanism.
Collapse
Affiliation(s)
- Jia-Shang Li
- Jiangsu Collaborative Innovation Center of
Chinese Medicinal Resources Industrialization, National and Local
Collaborative Engineering Center of Chinese Medicinal Resources
Industrialization and Formulae Innovative Medicine, and Jiangsu Key
Laboratory for High Technology Research of TCM Formulae, Nanjing University
of Chinese Medicine, Nanjing, P.R. China
| | | | - Zhuo Xu
- Jiangsu Collaborative Innovation Center of
Chinese Medicinal Resources Industrialization, National and Local
Collaborative Engineering Center of Chinese Medicinal Resources
Industrialization and Formulae Innovative Medicine, and Jiangsu Key
Laboratory for High Technology Research of TCM Formulae, Nanjing University
of Chinese Medicine, Nanjing, P.R. China
| | - Li-Hui Zhao
- Jiangsu Collaborative Innovation Center of
Chinese Medicinal Resources Industrialization, National and Local
Collaborative Engineering Center of Chinese Medicinal Resources
Industrialization and Formulae Innovative Medicine, and Jiangsu Key
Laboratory for High Technology Research of TCM Formulae, Nanjing University
of Chinese Medicine, Nanjing, P.R. China
| | - Ruo-Ying Fan
- Jiangsu Collaborative Innovation Center of
Chinese Medicinal Resources Industrialization, National and Local
Collaborative Engineering Center of Chinese Medicinal Resources
Industrialization and Formulae Innovative Medicine, and Jiangsu Key
Laboratory for High Technology Research of TCM Formulae, Nanjing University
of Chinese Medicine, Nanjing, P.R. China
| | - Jian-Ming Guo
- Jiangsu Collaborative Innovation Center of
Chinese Medicinal Resources Industrialization, National and Local
Collaborative Engineering Center of Chinese Medicinal Resources
Industrialization and Formulae Innovative Medicine, and Jiangsu Key
Laboratory for High Technology Research of TCM Formulae, Nanjing University
of Chinese Medicine, Nanjing, P.R. China
| | - Da-Wei Qian
- Jiangsu Collaborative Innovation Center of
Chinese Medicinal Resources Industrialization, National and Local
Collaborative Engineering Center of Chinese Medicinal Resources
Industrialization and Formulae Innovative Medicine, and Jiangsu Key
Laboratory for High Technology Research of TCM Formulae, Nanjing University
of Chinese Medicine, Nanjing, P.R. China
| | - Jin-Ao Duan
- Jiangsu Collaborative Innovation Center of
Chinese Medicinal Resources Industrialization, National and Local
Collaborative Engineering Center of Chinese Medicinal Resources
Industrialization and Formulae Innovative Medicine, and Jiangsu Key
Laboratory for High Technology Research of TCM Formulae, Nanjing University
of Chinese Medicine, 138 Xianlin Road, Nanjing 210023, P.R. China
| |
Collapse
|
17
|
Xie J, Li M, Ye W, Shan J, Zhao X, Duan Y, Liu Y, Unger BH, Cheng Y, Zhang W, Wu N, Xia XQ. Sinomenine Hydrochloride Ameliorates Fish Foodborne Enteritis via α7nAchR-Mediated Anti-Inflammatory Effect Whilst Altering Microbiota Composition. Front Immunol 2021; 12:766845. [PMID: 34887862 PMCID: PMC8650311 DOI: 10.3389/fimmu.2021.766845] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 11/02/2021] [Indexed: 02/06/2023] Open
Abstract
Foodborne intestinal inflammation is a major health and welfare issue in aquaculture. To prevent enteritis, various additives have been incorporated into the fish diet. Considering anti-inflammatory immune regulation, an effective natural compound could potentially treat or prevent intestinal inflammation. Our previous study has revealed galantamine’s effect on soybean induced enteritis (SBMIE) and has highlighted the possible role of the cholinergic anti-inflammatory pathway in the fish gut. To further activate the intestinal cholinergic related anti-inflammatory function, α7nAchR signaling was considered. In this study, sinomenine, a typical agonist of α7nAChR in mammals, was tested to treat fish foodborne enteritis via its potential anti-inflammation effect using the zebrafish foodborne enteritis model. After sinomenine’s dietary inclusion, results suggested that there was an alleviation of intestinal inflammation at a pathological level. This outcome was demonstrated through the improved morphology of intestinal villi. At a molecular level, SN suppressed inflammatory cytokines’ expression (especially for tnf-α) and upregulated anti-inflammation-related functions (indicated by expression of il-10, il-22, and foxp3a). To systematically understand sinomenine’s intestinal effect on SBMIE, transcriptomic analysis was done on the SBMIE adult fish model. DEGs (sinomenine vs soybean meal groups) were enriched in GO terms related to the negative regulation of lymphocyte/leukocyte activation and alpha-beta T cell proliferation, as well as the regulation of lymphocyte migration. The KEGG pathways for glycolysis and insulin signaling indicated metabolic adjustments of α7nAchR mediated anti-inflammatory effect. To demonstrate the immune cells’ response, in the SBMIE larva model, inflammatory gatherings of neutrophils, macrophages, and lymphocytes caused by soybean meal could be relieved significantly with the inclusion of sinomenine. This was consistent within the sinomenine group as CD4+ or Foxp3+ lymphocytes were found with a higher proportion at the base of mucosal folds, which may suggest the Treg population. Echoing, the sinomenine group’s 16s sequencing result, there were fewer enteritis-related TM7, Sphingomonas and Shigella, but more Cetobacterium, which were related to glucose metabolism. Our findings indicate that sinomenine hydrochloride could be important in the prevention of fish foodborne enteritis at both immune and microbiota levels.
Collapse
Affiliation(s)
- Jiayuan Xie
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China.,College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Ming Li
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China.,College of Fisheries and Life Science, Dalian Ocean University, Dalian, China
| | - Weidong Ye
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China.,College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Junwei Shan
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China.,College of Fisheries and Life Science, Dalian Ocean University, Dalian, China
| | - Xuyang Zhao
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China.,College of Fisheries and Life Science, Dalian Ocean University, Dalian, China
| | - You Duan
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China.,College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Yuhang Liu
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China.,College of Fisheries and Life Science, Dalian Ocean University, Dalian, China
| | | | - Yingyin Cheng
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
| | - Wanting Zhang
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
| | - Nan Wu
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China.,College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Xiao-Qin Xia
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China.,College of Fisheries and Life Science, Dalian Ocean University, Dalian, China
| |
Collapse
|
18
|
Wei B, Zhang J, Wen R, Chen T, Xia N, Liu Y, Wang Z. Genetically Modified Sugarcane Intercropping Soybean Impact on Rhizosphere Bacterial Communities and Co-occurrence Patterns. Front Microbiol 2021; 12:742341. [PMID: 34970232 PMCID: PMC8713472 DOI: 10.3389/fmicb.2021.742341] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Accepted: 11/09/2021] [Indexed: 12/01/2022] Open
Abstract
Strategies involving genes in the dehydration-responsive element binding (DREB) family, which participates in drought stress regulation, and intercropping with legumes are becoming prominent options in promoting sustainable sugarcane cultivation. An increasing number of studies focusing on root interactions in intercropping systems, particularly involving transgenic crops, are being conducted to better understand and thus, harness beneficial soil microbes to enhance plant growth. We designed experiments to investigate the characteristics of two intercropping patterns, soybean with wild-type (WT) sugarcane and soybean with genetically modified (GM) Ea-DREB2B-overexpressing sugarcane, to assess the response of the rhizosphere microbiota to the different cropping patterns. Bacterial diversity in the rhizosphere microbial community differed between the two intercropping pattens. In addition, the biomass of GM sugarcane that intercropped with soybean was significantly improved compared with WT sugarcane, and the aboveground biomass and root biomass of GM soybean intercropping sugarcane increased by 49.15 and 46.03% compared with monoculture. Furthermore, a beneficial rhizosphere environment for the growth of Actinobacteria was established in the systems intercropped with GM sugarcane. Improving the production mode of crops by genetic modification is a key strategy to improving crop yields and provides new opportunities to further investigate the effects of intercropping on plant roots and soil microbiota. Thus, this study provides a basis for selecting suitable sugarcane-soybean intercropping patterns and a theoretical foundation for a sustainable sugarcane production.
Collapse
Affiliation(s)
- Beilei Wei
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Life Sciences, School of Public Health, Xiamen University, Xiamen, China
- College of Agronomy, Guangxi University, Nanning, China
- Guangxi Key Laboratory of Sugarcane Biology, Nanning, China
| | - Jinlian Zhang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Life Sciences, School of Public Health, Xiamen University, Xiamen, China
- Microbiology Research Institute, Guangxi Academy of Agricultural Sciences, Nanning, China
| | - Rushuang Wen
- College of Agronomy, Guangxi University, Nanning, China
| | - Tingsu Chen
- Microbiology Research Institute, Guangxi Academy of Agricultural Sciences, Nanning, China
| | - Ningshao Xia
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Life Sciences, School of Public Health, Xiamen University, Xiamen, China
| | - Yue Liu
- College of Agronomy, Guangxi University, Nanning, China
- Guangxi Key Laboratory of Sugarcane Biology, Nanning, China
| | - Ziting Wang
- College of Agronomy, Guangxi University, Nanning, China
- Guangxi Key Laboratory of Sugarcane Biology, Nanning, China
| |
Collapse
|
19
|
Cornuault JK, Byatt G, Paquet ME, De Koninck P, Moineau S. Zebrafish: a big fish in the study of the gut microbiota. Curr Opin Biotechnol 2021; 73:308-313. [PMID: 34653834 DOI: 10.1016/j.copbio.2021.09.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 09/04/2021] [Accepted: 09/11/2021] [Indexed: 11/03/2022]
Abstract
The importance of the gut microbiota in host health is now well established, but the underlying mechanisms remain poorly understood. Among the animal models used to investigate microbiota-host interactions, the zebrafish (Danio renio) is gaining attention. Several factors contribute to the recent interest in this model, including its low cost, the ability to assess large cohorts, the possibility to obtain germ-free larvae from non-axenic parents, and the availability of optical methodologies to probe the transparent larvae and adults from various genetic lines. We review recent findings on the zebrafish gut microbiota and its modulation by exogenous microbes, nutrition, and environmental factors. We also highlight the potential of this model for assessing the impact of the gut microbiota on brain development.
Collapse
Affiliation(s)
- Jeffrey K Cornuault
- Département de biochimie, de microbiologie et de bio-informatique, Faculté des sciences et de génie, Université Laval, Québec, QC, G1V 0A6, Canada; Groupe de recherche en écologie buccale, Faculté de médecine dentaire, Université Laval, Québec, QC, G1V 0A6, Canada
| | - Gabriel Byatt
- Département de biochimie, de microbiologie et de bio-informatique, Faculté des sciences et de génie, Université Laval, Québec, QC, G1V 0A6, Canada; Groupe de recherche en écologie buccale, Faculté de médecine dentaire, Université Laval, Québec, QC, G1V 0A6, Canada; CERVO Brain Research Centre, Québec, QC, G1J 2G3, Canada
| | - Marie-Eve Paquet
- Département de biochimie, de microbiologie et de bio-informatique, Faculté des sciences et de génie, Université Laval, Québec, QC, G1V 0A6, Canada; CERVO Brain Research Centre, Québec, QC, G1J 2G3, Canada
| | - Paul De Koninck
- Département de biochimie, de microbiologie et de bio-informatique, Faculté des sciences et de génie, Université Laval, Québec, QC, G1V 0A6, Canada; CERVO Brain Research Centre, Québec, QC, G1J 2G3, Canada
| | - Sylvain Moineau
- Département de biochimie, de microbiologie et de bio-informatique, Faculté des sciences et de génie, Université Laval, Québec, QC, G1V 0A6, Canada; Groupe de recherche en écologie buccale, Faculté de médecine dentaire, Université Laval, Québec, QC, G1V 0A6, Canada; Félix d'Hérelle Reference Center for Bacterial Viruses, Université Laval, Québec, QC, G1V 0A6, Canada.
| |
Collapse
|
20
|
Lucerne KE, Osman A, Meckel KR, Kiraly DD. Contributions of neuroimmune and gut-brain signaling to vulnerability of developing substance use disorders. Neuropharmacology 2021; 192:108598. [PMID: 33965398 PMCID: PMC8220934 DOI: 10.1016/j.neuropharm.2021.108598] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 03/19/2021] [Accepted: 05/03/2021] [Indexed: 02/06/2023]
Abstract
Epidemiology and clinical research indicate that only a subset of people who are exposed to drugs of abuse will go on to develop a substance use disorder. Numerous factors impact individual susceptibility to developing a substance use disorder, including intrinsic biological factors, environmental factors, and interpersonal/social factors. Given the extensive morbidity and mortality that is wrought as a consequence of substance use disorders, a substantial body of research has focused on understanding the risk factors that mediate the shift from initial drug use to pathological drug use. Understanding these risk factors provides a clear path for the development of risk mitigation strategies to help reduce the burden of substance use disorders in the population. Here we will review the rapidly growing body of literature that examines the importance of interactions between the peripheral immune system, the gut microbiome, and the central nervous system (CNS) in mediating the transition to pathological drug use. While these systems had long been viewed as distinct, there is growing evidence that there is bidirectional communication between both the immune system and the gut microbiome that drive changes in neural and behavioral plasticity relevant to substance use disorders. Further, both of these systems are highly sensitive to environmental perturbations and are implicated in numerous neuropsychiatric conditions. While the field of study examining these interactions in substance use disorders is in its relative infancy, clarifying the relationship between gut-immune-brain signaling and substance use disorders has potential to improve our understanding of individual propensity to developing addiction and yield important insight into potential treatment options.
Collapse
Affiliation(s)
- Kelsey E Lucerne
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Aya Osman
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Katherine R Meckel
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Drew D Kiraly
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
21
|
Yadav J, El Hassani M, Sodhi J, Lauschke VM, Hartman JH, Russell LE. Recent developments in in vitro and in vivo models for improved translation of preclinical pharmacokinetics and pharmacodynamics data. Drug Metab Rev 2021; 53:207-233. [PMID: 33989099 DOI: 10.1080/03602532.2021.1922435] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Improved pharmacokinetics/pharmacodynamics (PK/PD) prediction in the early stages of drug development is essential to inform lead optimization strategies and reduce attrition rates. Recently, there have been significant advancements in the development of new in vitro and in vivo strategies to better characterize pharmacokinetic properties and efficacy of drug leads. Herein, we review advances in experimental and mathematical models for clearance predictions, advancements in developing novel tools to capture slowly metabolized drugs, in vivo model developments to capture human etiology for supporting drug development, limitations and gaps in these efforts, and a perspective on the future in the field.
Collapse
Affiliation(s)
- Jaydeep Yadav
- Department of Pharmacokinetics, Pharmacodynamics, and Drug Metabolism, Merck & Co., Inc., Boston, MA, USA
| | | | - Jasleen Sodhi
- Department of Bioengineering and Therapeutic Sciences, Schools of Pharmacy and Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Volker M Lauschke
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Jessica H Hartman
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC, USA
| | | |
Collapse
|