1
|
Ji S, Guo Y, Yan W, Wei F, Ding J, Hong W, Wu X, Ku T, Yue H, Sang N. PM 2.5 exposure contributes to anxiety and depression-like behaviors via phenyl-containing compounds interfering with dopamine receptor. Proc Natl Acad Sci U S A 2024; 121:e2319595121. [PMID: 38739786 PMCID: PMC11127009 DOI: 10.1073/pnas.2319595121] [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: 11/27/2023] [Accepted: 04/04/2024] [Indexed: 05/16/2024] Open
Abstract
As a global problem, fine particulate matter (PM2.5) really needs local fixes. Considering the increasing epidemiological relevance to anxiety and depression but inconsistent toxicological results, the most important question is to clarify whether and how PM2.5 causally contributes to these mental disorders and which components are the most dangerous for crucial mitigation in a particular place. In the present study, we chronically subjected male mice to a real-world PM2.5 exposure system throughout the winter heating period in a coal combustion area and revealed that PM2.5 caused anxiety and depression-like behaviors in adults such as restricted activity, diminished exploratory interest, enhanced repetitive stereotypy, and elevated acquired immobility, through behavioral tests including open field, elevated plus maze, marble-burying, and forced swimming tests. Importantly, we found that dopamine signaling was perturbed using mRNA transcriptional profile and bioinformatics analysis, with Drd1 as a potential target. Subsequently, we developed the Drd1 expression-directed multifraction isolating and nontarget identifying framework and identified a total of 209 compounds in PM2.5 organic extracts capable of reducing Drd1 expression. Furthermore, by applying hierarchical characteristic fragment analysis and molecular docking and dynamics simulation, we clarified that phenyl-containing compounds competitively bound to DRD1 and interfered with dopamine signaling, thereby contributing to mental disorders. Taken together, this work provides experimental evidence for researchers and clinicians to identify hazardous factors in PM2.5 and prevent adverse health outcomes and for local governments and municipalities to control source emissions for diminishing specific disease burdens.
Collapse
Affiliation(s)
- Shaoyang Ji
- Department of Environment Science, College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi030006, People’s Republic of China
| | - Yuqiong Guo
- Department of Environment Science, College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi030006, People’s Republic of China
| | - Wei Yan
- Department of Environment Science, College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi030006, People’s Republic of China
- Xuzhou Engineering Research Center of Medical Genetics and Transformation, Key Laboratory of Genetic Foundation and Clinical Application, Department of Genetics, Xuzhou Medical University, Xuzhou, Jiangsu221004, People’s Republic of China
| | - Fang Wei
- Department of Environment Engineering, College of Quality and Safety Engineering, China Jiliang University, Hangzhou, Zhejiang310018, People’s Republic of China
| | - Jinjian Ding
- Department of Environment Engineering, College of Quality and Safety Engineering, China Jiliang University, Hangzhou, Zhejiang310018, People’s Republic of China
- Institute of Environmental and Health Sciences, China Jiliang University, Hangzhou, Zhejiang310018, People’s Republic of China
| | - Wenjun Hong
- Department of Environment Engineering, College of Quality and Safety Engineering, China Jiliang University, Hangzhou, Zhejiang310018, People’s Republic of China
- Institute of Environmental and Health Sciences, China Jiliang University, Hangzhou, Zhejiang310018, People’s Republic of China
| | - Xiaoyun Wu
- Department of Environment Science, College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi030006, People’s Republic of China
| | - Tingting Ku
- Department of Environment Science, College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi030006, People’s Republic of China
| | - Huifeng Yue
- Department of Environment Science, College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi030006, People’s Republic of China
| | - Nan Sang
- Department of Environment Science, College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi030006, People’s Republic of China
| |
Collapse
|
2
|
Salomon JD, Qiu H, Feng D, Owens J, Khailova L, Osorio Lujan S, Iguidbashian J, Chhonker YS, Murry DJ, Riethoven JJ, Lindsey ML, Singh AB, Davidson JA. Piglet cardiopulmonary bypass induces intestinal dysbiosis and barrier dysfunction associated with systemic inflammation. Dis Model Mech 2023; 16:dmm049742. [PMID: 36426663 PMCID: PMC9844230 DOI: 10.1242/dmm.049742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Accepted: 11/14/2022] [Indexed: 11/26/2022] Open
Abstract
The intestinal microbiome is essential to human health and homeostasis, and is implicated in the pathophysiology of disease, including congenital heart disease and cardiac surgery. Improving the microbiome and reducing inflammatory metabolites may reduce systemic inflammation following cardiac surgery with cardiopulmonary bypass (CPB) to expedite recovery post-operatively. Limited research exists in this area and identifying animal models that can replicate changes in the human intestinal microbiome after CPB is necessary. We used a piglet model of CPB with two groups, CPB (n=5) and a control group with mechanical ventilation (n=7), to evaluate changes to the microbiome, intestinal barrier dysfunction and intestinal metabolites with inflammation after CPB. We identified significant changes to the microbiome, barrier dysfunction, intestinal short-chain fatty acids and eicosanoids, and elevated cytokines in the CPB/deep hypothermic circulatory arrest group compared to the control group at just 4 h after intervention. This piglet model of CPB replicates known human changes to intestinal flora and metabolite profiles, and can be used to evaluate gut interventions aimed at reducing downstream inflammation after cardiac surgery with CPB.
Collapse
Affiliation(s)
- Jeffrey D. Salomon
- Department of Pediatrics, University of Nebraska Medical Center, Omaha, NE 68102, USA
- Department of Cellular & Integrative Physiology, University of Nebraska Medical Center, Omaha, NE 68102, USA
| | - Haowen Qiu
- Center for Biotechnology, University of Nebraska Lincoln, Lincoln, NE 68588, USA
| | - Dan Feng
- Department of Hematology/Oncology, University of Nebraska Medical Center, Omaha, NE 68102, USA
| | - Jacob Owens
- Department of Pediatrics, University of Nebraska Medical Center, Omaha, NE 68102, USA
| | - Ludmila Khailova
- Department of Pediatrics, University of Colorado, Aurora, CO 80045, USA
| | | | - John Iguidbashian
- Department of Pediatrics, University of Colorado, Aurora, CO 80045, USA
| | - Yashpal S. Chhonker
- Department of Pharmacy Practice, University of Nebraska Medical Center College of Pharmacy, Omaha, NE 68102, USA
| | - Daryl J. Murry
- Department of Pharmacy Practice, University of Nebraska Medical Center College of Pharmacy, Omaha, NE 68102, USA
| | - Jean-Jack Riethoven
- Center for Biotechnology, University of Nebraska Lincoln, Lincoln, NE 68588, USA
| | - Merry L. Lindsey
- School of Graduate Studies and Research, Meharry Medical College, Nashville, TN 37208, USA
- Research Service, Nebraska-Western Iowa Health Care System, Omaha, NE 68105, USA
| | - Amar B. Singh
- Research Service, Nebraska-Western Iowa Health Care System, Omaha, NE 68105, USA
- Department of Biochemistry & Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68102, USA
| | - Jesse A. Davidson
- Department of Pediatrics, University of Colorado, Aurora, CO 80045, USA
| |
Collapse
|
3
|
Kim A, Gu SM, Lee H, Kim DE, Hong JT, Yun J, Cha HJ. Prenatal ketamine exposure impairs prepulse inhibition via arginine vasopressin receptor 1A-mediated GABAergic neuronal dysfunction in the striatum. Biomed Pharmacother 2023; 160:114318. [PMID: 36738499 DOI: 10.1016/j.biopha.2023.114318] [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: 06/18/2021] [Revised: 01/17/2023] [Accepted: 01/26/2023] [Indexed: 02/05/2023] Open
Abstract
Ketamine is a widely used anesthetic with N-methyl-D-aspartate (NMDA) receptor antagonism. Exposure to ketamine and NMDA receptor antagonists may induce psychosis. However, the mechanism underlying the effects of ketamine on the immature brain remains unclear. In this study, NMDA receptor antagonists, ketamine and methoxetamine, were administered to pregnant F344 rats (E17). These regimens induce psychosis-like behaviors in the offspring, such as hyperlocomotion induced by MK-801, a non-competitive NMDA receptor antagonist. We also observed that prepulse inhibition (PPI) was significantly reduced. Interestingly, ketamine administration increased the arginine vasopressin receptor 1A (Avpr1a) expression levels in the striatum of offspring with abnormal behaviors. Methoxetamine, another NMDA receptor antagonist, also showed similar results. In addition, we demonstrated a viral vector-induced Avpr1a overexpression in the striatum-inhibited PPI. In the striatum of offspring, ketamine or methoxetamine treatment increased glutamate decarboxylase 67 (GAD67) and δ-aminobutyric acid (GABA) levels. These results show that prenatal NMDA receptor antagonist treatment induces GABAergic neuronal dysfunction and abnormalities in sensorimotor gating via regulating Avpr1a expression in the striatum.
Collapse
Affiliation(s)
- Aeseul Kim
- College of Pharmacy, Chungbuk National University, 194-31, Osongsaengmyeong 1-ro, Osong-eup, Heungdeok-gu, Cheongju-si, Chungcheongbuk-do 28160, the Republic of Korea
| | - Sun Mi Gu
- College of Pharmacy, Chungbuk National University, 194-31, Osongsaengmyeong 1-ro, Osong-eup, Heungdeok-gu, Cheongju-si, Chungcheongbuk-do 28160, the Republic of Korea
| | - Haemiru Lee
- Pharmacological Research Division, National Institute of Food and Drug Safety Evaluation (NIFDS), Ministry of Food and Drug Safety (MFDS), OHTAC 187, Osongsaengmyong 2-ro, Cheongju-si, Chungcheongbuk-do 28159, the Republic of Korea
| | - Dong Eun Kim
- Pharmacological Research Division, National Institute of Food and Drug Safety Evaluation (NIFDS), Ministry of Food and Drug Safety (MFDS), OHTAC 187, Osongsaengmyong 2-ro, Cheongju-si, Chungcheongbuk-do 28159, the Republic of Korea
| | - Jin Tae Hong
- College of Pharmacy, Chungbuk National University, 194-31, Osongsaengmyeong 1-ro, Osong-eup, Heungdeok-gu, Cheongju-si, Chungcheongbuk-do 28160, the Republic of Korea
| | - Jaesuk Yun
- College of Pharmacy, Chungbuk National University, 194-31, Osongsaengmyeong 1-ro, Osong-eup, Heungdeok-gu, Cheongju-si, Chungcheongbuk-do 28160, the Republic of Korea.
| | - Hye Jin Cha
- College of Veterinary Medicine, Gyeongsang National University, 501, Jinju-daero, Jinju-si, Gyeongsangnam-do 52828, the Republic of Korea.
| |
Collapse
|
4
|
Donovan M, Mackey CS, Lynch MDJ, Platt GN, Brown AN, Washburn BK, Trickey DJ, Curtis JT, Liu Y, Charles TC, Wang Z, Jones KM. Limosilactobacillus reuteri administration alters the gut-brain-behavior axis in a sex-dependent manner in socially monogamous prairie voles. Front Microbiol 2023; 14:1015666. [PMID: 36846764 PMCID: PMC9945313 DOI: 10.3389/fmicb.2023.1015666] [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: 08/10/2022] [Accepted: 01/05/2023] [Indexed: 02/11/2023] Open
Abstract
Research on the role of gut microbiota in behavior has grown dramatically. The probiotic L. reuteri can alter social and stress-related behaviors - yet, the underlying mechanisms remain largely unknown. Although traditional laboratory rodents provide a foundation for examining the role of L. reuteri on the gut-brain axis, they do not naturally display a wide variety of social behaviors. Using the highly-social, monogamous prairie vole (Microtus ochrogaster), we examined the effects of L. reuteri administration on behaviors, neurochemical marker expression, and gut-microbiome composition. Females, but not males, treated with live L. reuteri displayed lower levels of social affiliation compared to those treated with heat-killed L. reuteri. Overall, females displayed a lower level of anxiety-like behaviors than males. Live L. reuteri-treated females had lower expression of corticotrophin releasing factor (CRF) and CRF type-2-receptor in the nucleus accumbens, and lower vasopressin 1a-receptor in the paraventricular nucleus of the hypothalamus (PVN), but increased CRF in the PVN. There were both baseline sex differences and sex-by-treatment differences in gut microbiome composition. Live L. reuteri increased the abundance of several taxa, including Enterobacteriaceae, Lachnospiraceae NK4A136, and Treponema. Interestingly, heat-killed L. reuteri increased abundance of the beneficial taxa Bifidobacteriaceae and Blautia. There were significant correlations between changes in microbiota, brain neurochemical markers, and behaviors. Our data indicate that L. reuteri impacts gut microbiota, gut-brain axis and behaviors in a sex-specific manner in socially-monogamous prairie voles. This demonstrates the utility of the prairie vole model for further examining causal impacts of microbiome on brain and behavior.
Collapse
Affiliation(s)
- Meghan Donovan
- Department of Psychology and Program in Neuroscience, Florida State University, Tallahassee, FL, United States
- Rocky Mountain Mental Illness Research Education and Clinical Center, Rocky Mountain Regional VA Medical Center, Aurora, CO, United States
- Department of Physical Medicine and Rehabilitation, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Calvin S. Mackey
- Department of Biological Science, Florida State University, Tallahassee, FL, United States
| | - Michael D. J. Lynch
- Metagenom Bio Life Science Inc, Waterloo, ON, Canada
- Department of Biology, University of Waterloo, Waterloo, ON, Canada
| | - Grayson N. Platt
- Department of Psychology and Program in Neuroscience, Florida State University, Tallahassee, FL, United States
- Department of Biological Science, Florida State University, Tallahassee, FL, United States
| | - Amber N. Brown
- Department of Biological Science Core Facilities, Florida State University, Tallahassee, FL, United States
| | - Brian K. Washburn
- Department of Biological Science Core Facilities, Florida State University, Tallahassee, FL, United States
| | - Darryl J. Trickey
- Department of Biological Science, Florida State University, Tallahassee, FL, United States
| | - J. Thomas Curtis
- Department of Pharmacology and Physiology, Oklahoma State University Center for Health Sciences, Tulsa, OK, United States
| | - Yan Liu
- Department of Psychology and Program in Neuroscience, Florida State University, Tallahassee, FL, United States
| | - Trevor C. Charles
- Metagenom Bio Life Science Inc, Waterloo, ON, Canada
- Department of Biology, University of Waterloo, Waterloo, ON, Canada
| | - Zuoxin Wang
- Department of Psychology and Program in Neuroscience, Florida State University, Tallahassee, FL, United States
| | - Kathryn M. Jones
- Department of Biological Science, Florida State University, Tallahassee, FL, United States
| |
Collapse
|
5
|
Diviccaro S, Giatti S, Cioffi L, Falvo E, Herian M, Caruso D, Melcangi RC. Gut Inflammation Induced by Finasteride Withdrawal: Therapeutic Effect of Allopregnanolone in Adult Male Rats. Biomolecules 2022; 12:1567. [PMID: 36358917 PMCID: PMC9687671 DOI: 10.3390/biom12111567] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 10/21/2022] [Accepted: 10/22/2022] [Indexed: 07/29/2023] Open
Abstract
The treatment with finasteride (i.e., an inhibitor of 5α-reductase) may be associated with different side effects (i.e., depression, anxiety, cognitive impairment and sexual dysfunction) inducing the so-called post finasteride syndrome (PFS). Moreover, previous observations in PFS patients and an experimental model showed alterations in gut microbiota populations, suggesting an inflammatory environment. To confirm this hypothesis, we have explored the effect of chronic treatment with finasteride (i.e., for 20 days) and its withdrawal (i.e., for 1 month) on the levels of steroids, neurotransmitters, pro-inflammatory cytokines and gut permeability markers in the colon of adult male rat. The obtained data demonstrate that the levels of allopregnanolone (ALLO) decreased after finasteride treatment and after its withdrawal. Following the drug suspension, the decrease in ALLO levels correlates with an increase in IL-1β and TNF-α, serotonin and a decrease in dopamine. Importantly, ALLO treatment is able to counteract some of these alterations. The relation between ALLO and GABA-A receptors and/or pregnenolone (ALLO precursor) could be crucial in their mode of action. These observations provide an important background to explore further the protective effect of ALLO in the PFS experimental model and the possibility of its translation into clinical therapy.
Collapse
|
6
|
Functional Plasmon-Activated Water Increases Akkermansia muciniphila Abundance in Gut Microbiota to Ameliorate Inflammatory Bowel Disease. Int J Mol Sci 2022; 23:ijms231911422. [PMID: 36232724 PMCID: PMC9570201 DOI: 10.3390/ijms231911422] [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] [Received: 08/22/2022] [Revised: 09/22/2022] [Accepted: 09/22/2022] [Indexed: 11/16/2022] Open
Abstract
Inflammatory bowel disease (IBD) is associated with dysbiosis and intestinal barrier dysfunction, as indicated by epithelial hyperpermeability and high levels of mucosal-associated bacteria. Changes in gut microbiota may be correlated with IBD pathogenesis. Additionally, microbe-based treatments could mitigate clinical IBD symptoms. Plasmon-activated water (PAW) is known to have an anti-inflammatory potential. In this work, we studied the association between the anti-inflammatory ability of PAW and intestinal microbes, thereby improving IBD treatment. We examined the PAW-induced changes in the colonic immune activity and microbiota of mice by immunohistochemistry and next generation sequencing, determined whether drinking PAW can mitigate IBD induced by 2,4,6-trinitrobenzene sulfonic acid (TNBS) and dysbiosis through mice animal models. The effects of specific probiotic species on mice with TNBS-induced IBD were also investigated. Experimental results indicated that PAW could change the local inflammation in the intestinal microenvironment. Moreover, the abundance of Akkermansia spp. was degraded in the TNBS-treated mice but elevated in the PAW-drinking mice. Daily rectal injection of Akkermansia muciniphila, a potential probiotic species in Akkermansia spp., also improved the health of the mice. Correspondingly, both PAW consumption and increasing the intestinal abundance of Akkermansia muciniphila can mitigate IBD in mice. These findings indicate that increasing the abundance of Akkermansia muciniphila in the gut through PAW consumption or other methods may mitigate IBD in mice with clinically significant IBD.
Collapse
|
7
|
Abstract
To date, much of the focus of gut-brain axis research has been on gut microbiota regulation of anxiety and stress-related behaviors. Much less attention has been directed to potential connections between gut microbiota and compulsive behavior. Here, we discuss a potential link between gut barrier dysfunction and compulsive behavior that is mediated through "type 2" rather than "type 1" inflammation. We examine connections between compulsive behavior and type 2 inflammation in Tourette syndrome, obsessive-compulsive disorder, autism, addiction, and post-traumatic stress disorder. Next, we discuss potential connections between gut barrier dysfunction, type 2 inflammation, and compulsive behavior. We posit a potential mechanism whereby gut barrier dysfunction-associated type 2 inflammation may drive compulsive behavior through histamine regulation of dopamine neurotransmission. Finally, we discuss the possibility of exploiting the greater accessibility of the gut relative to the brain in identifying targets to treat compulsive behavior disorders.
Collapse
|
8
|
Liu J, Huang S, Zhang X, Li G, Batsuren E, Lu W, Xu X, He C, Song Y, Zhang Z. Gut microbiota reflect the crowding stress of space shortage, physical and non-physical contact in Brandt's voles (Lasiopodomys brandtii). Microbiol Res 2021; 255:126928. [PMID: 34883384 DOI: 10.1016/j.micres.2021.126928] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 11/07/2021] [Accepted: 11/22/2021] [Indexed: 12/19/2022]
Abstract
Density-dependence plays a critical role in behavior and population regulation of small mammals, which is likely mediated by hormones and gut microbiota. High density-induced crowding effects often cause a combination of various social stresses including space shortage, physical contact and non-physical contact, but their distinct effects on gut microbiota in animals have not been investigated. In this study, we examined the crowding effects of space shortage and physical or non-physical contact stress on serum corticosterone and gut microbiota of Brandt's voles in both laboratory and field conditions. Our results demonstrated that the space shortage stress showed a more predominant impact on serum corticosterone and gut microbiota of voles than physical or non-physical contact stress; the crowding effects of non-physical contact stress became stronger in high density conditions, while physical contact stress was stronger in a larger group without density effects. High density or group size treatments under both laboratory and semi-natural enclosure conditions significantly increased the relative abundance of key differential taxa, including Bacteroidetes, TM7, S24_7, Streptococcus, and Lactobacillus; while high density or group size treatments decreased the relative abundance of Firmicutes, Staphylococcaceae, Bacteroides, Faecalibacterium, and Adlercreutzia. Our study suggests that high density-induced space shortage and physical contact or non-physical contact stress may play a significant role in behavior and population regulation through altering gut microbiota in small mammals. Our results may also have significant implications in rodent control or health management for livestock.
Collapse
Affiliation(s)
- Jing Liu
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China; CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Shuli Huang
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China; CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xin Zhang
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China; CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Guoliang Li
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China; CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Erdenetuya Batsuren
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China; CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing, 100049, China; Institute of Plant Protection, Ulaanbaatar, 17024, Mongolia
| | - Wei Lu
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China; CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xiaoming Xu
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China; CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Chen He
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China; CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yiran Song
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China; CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Zhibin Zhang
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China; CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
9
|
Diviccaro S, Caputi V, Cioffi L, Giatti S, Lyte JM, Caruso D, O’Mahony SM, Melcangi RC. Exploring the Impact of the Microbiome on Neuroactive Steroid Levels in Germ-Free Animals. Int J Mol Sci 2021; 22:ijms222212551. [PMID: 34830433 PMCID: PMC8622241 DOI: 10.3390/ijms222212551] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 11/16/2021] [Accepted: 11/17/2021] [Indexed: 02/07/2023] Open
Abstract
Steroid hormones are essential biomolecules for human physiology as they modulate the endocrine system, nervous function and behaviour. Recent studies have shown that the gut microbiota is directly involved in the production and metabolism of steroid hormones in the periphery. However, the influence of the gut microbiota on levels of steroids acting and present in the brain (i.e., neuroactive steroids) is not fully understood. Therefore, using liquid chromatography–tandem mass spectrometry, we assessed the levels of several neuroactive steroids in various brain areas and the plasma of germ-free (GF) male mice and conventionally colonized controls. The data obtained indicate an increase in allopregnanolone levels associated with a decrease in those of 5α-androstane-3α, 17β-diol (3α-diol) in the plasma of GF mice. Moreover, an increase of dihydroprogesterone and isoallopregnanolone in the hippocampus, cerebellum, and cerebral cortex was also reported. Changes in dihydrotestosterone and 3α-diol levels were also observed in the hippocampus of GF mice. In addition, an increase in dehydroepiandrosterone was associated with a decrease in testosterone levels in the hypothalamus of GF mice. Our findings suggest that the absence of microbes affects the neuroactive steroids in the periphery and the brain, supporting the evidence of a microbiota-mediated modulation of neuroendocrine pathways involved in preserving host brain functioning.
Collapse
Affiliation(s)
- Silvia Diviccaro
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Via Balzaretti 9, 20133 Milano, Italy; (S.D.); (L.C.); (S.G.); (D.C.)
| | - Valentina Caputi
- APC Microbiome Ireland, University College Cork, T12 YT20 Cork, Ireland; (V.C.); (J.M.L.); (S.M.O.)
| | - Lucia Cioffi
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Via Balzaretti 9, 20133 Milano, Italy; (S.D.); (L.C.); (S.G.); (D.C.)
| | - Silvia Giatti
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Via Balzaretti 9, 20133 Milano, Italy; (S.D.); (L.C.); (S.G.); (D.C.)
| | - Joshua M. Lyte
- APC Microbiome Ireland, University College Cork, T12 YT20 Cork, Ireland; (V.C.); (J.M.L.); (S.M.O.)
- Poultry Production and Product Safety Research Unit, United States Department of Agriculture, Agricultural Research Service, Fayetteville, AR 72701, USA
| | - Donatella Caruso
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Via Balzaretti 9, 20133 Milano, Italy; (S.D.); (L.C.); (S.G.); (D.C.)
| | - Siobhain M. O’Mahony
- APC Microbiome Ireland, University College Cork, T12 YT20 Cork, Ireland; (V.C.); (J.M.L.); (S.M.O.)
- Department of Anatomy and Neuroscience, University College Cork, T12 ND89 Cork, Ireland
| | - Roberto Cosimo Melcangi
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Via Balzaretti 9, 20133 Milano, Italy; (S.D.); (L.C.); (S.G.); (D.C.)
- Correspondence: ; Tel.: +39-02-50318238; Fax: +39-02-50318202
| |
Collapse
|
10
|
Al-Kuraishy HM, Al-Gareeb AI, Qusti S, Alshammari EM, Atanu FO, Batiha GES. Arginine vasopressin and pathophysiology of COVID-19: An innovative perspective. Biomed Pharmacother 2021; 143:112193. [PMID: 34543987 PMCID: PMC8440235 DOI: 10.1016/j.biopha.2021.112193] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 09/06/2021] [Accepted: 09/10/2021] [Indexed: 12/25/2022] Open
Abstract
In Covid-19, systemic disturbances may progress due to development of cytokine storm and dysregulation of and plasma osmolarility due to high release of pro-inflammatory cytokines and neuro-hormonal disorders. Arginine vasopressin (AVP) which is involve in the regulation of body osmotic system, body water content, blood pressure and plasma volume, that are highly disturbed in Covid-19 and linked with poor clinical outcomes. Therefore, this present study aimed to find the potential association between AVP serum level and inflammatory disorders in Covid-19. It has been observed by different recent studies that physiological response due to fever, pain, hypovolemia, dehydration, and psychological stress is characterized by activation release of AVP to counter-balance high blood viscosity in Covid-19 patients. In addition, activated immune cells mainly T and B lymphocytes and released pro-inflammatory cytokines stimulate discharge of stored AVP from immune cells, which in a vicious cycle trigger release of pro-inflammatory cytokines. Vasopressin receptor antagonists have antiviral and anti-inflammatory effects that may inhibit AVP-induced hyponatremia and release of pro-inflammatory cytokines in Covid-19. In conclusion, release of AVP from hypothalamus is augmented in Covid-19 due to stress, high pro-inflammatory cytokines, high circulating AngII and inhibition of GABAergic neurons. In turn, high AVP level leads to induction of hyponatremia, inflammatory disorders, and development of complications in Covid-19 by activation of NF-κB and NLRP3 inflammasome with release of pro-inflammatory cytokines. Therefore, AVP antagonists might be novel potential therapeutic modality in treating Covid-19 through mitigation of AVP-mediated inflammatory disorders and hyponatremia.
Collapse
Affiliation(s)
- Hayder M Al-Kuraishy
- Department of Clinical Pharmacology and Medicine, College of Medicine, ALmustansiriyia University, Baghdad, Iraq.
| | - Ali I Al-Gareeb
- Department of Clinical Pharmacology and Medicine, College of Medicine, ALmustansiriyia University, Baghdad, Iraq.
| | - Safaa Qusti
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia.
| | - Eida M Alshammari
- Department of Chemistry, College of Sciences, University of Ha'il, Ha'il, Saudi Arabia.
| | - Francis O Atanu
- Department of Biochemistry, Faculty of Natural Sciences, Kogi State University, P.M.B. 1008 Anyigba, Nigeria.
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour 22511, AlBeheira, Egypt.
| |
Collapse
|
11
|
Borgo F, Macandog AD, Diviccaro S, Falvo E, Giatti S, Cavaletti G, Melcangi RC. Alterations of gut microbiota composition in post-finasteride patients: a pilot study. J Endocrinol Invest 2021; 44:1263-1273. [PMID: 32951160 PMCID: PMC8124058 DOI: 10.1007/s40618-020-01424-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Accepted: 09/08/2020] [Indexed: 12/11/2022]
Abstract
PURPOSE Post-finasteride syndrome (PFS) has been reported in a subset of patients treated with finasteride (an inhibitor of the enzyme 5alpha-reductase) for androgenetic alopecia. These patients showed, despite the suspension of the treatment, a variety of persistent symptoms, like sexual dysfunction and cognitive and psychological disorders, including depression. A growing body of literature highlights the relevance of the gut microbiota-brain axis in human health and disease. For instance, alterations in gut microbiota composition have been reported in patients with major depressive disorder. Therefore, we have here analyzed the gut microbiota composition in PFS patients in comparison with a healthy cohort. METHODS Fecal microbiota of 23 PFS patients was analyzed by 16S rRNA gene sequencing and compared with that reported in ten healthy male subjects. RESULTS Sexual dysfunction, psychological and cognitive complaints, muscular problems, and physical alterations symptoms were reported in more than half of the PFS patients at the moment of sample collection. The quality sequence check revealed a low library depth for two fecal samples. Therefore, the gut microbiota analyses were conducted on 21 patients. The α-diversity was significantly lower in PFS group, showing a reduction of richness and diversity of gut microbiota structure. Moreover, when visualizing β-diversity, a clustering effect was found in the gut microbiota of a subset of PFS subjects, which was also characterized by a reduction in Faecalibacterium spp. and Ruminococcaceae UCG-005, while Alloprevotella and Odoribacter spp were increased compared to healthy control. CONCLUSION Gut microbiota population is altered in PFS patients, suggesting that it might represent a diagnostic marker and a possible therapeutic target for this syndrome.
Collapse
Affiliation(s)
- F Borgo
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - A D Macandog
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, Milan, Italy
- Department of Oncology and Hemato-Oncology, Università Degli Studi di Milano, Milan, Italy
| | - S Diviccaro
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università Degli Studi di Milano, Via Balzaretti 9, 20133, Milan, Italy
| | - E Falvo
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università Degli Studi di Milano, Via Balzaretti 9, 20133, Milan, Italy
| | - S Giatti
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università Degli Studi di Milano, Via Balzaretti 9, 20133, Milan, Italy
| | - G Cavaletti
- Experimental Neurology Unit, Università di Milano-Bicocca, Monza, Italy
- Milan Center for Neuroscience, Università di Milano-Bicocca, Monza, Italy
| | - R C Melcangi
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università Degli Studi di Milano, Via Balzaretti 9, 20133, Milan, Italy.
| |
Collapse
|
12
|
Manosso LM, Lin J, Carlessi AS, Recco KCC, Quevedo J, Gonçalves CL, Réus GZ. Sex-related patterns of the gut-microbiota-brain axis in the neuropsychiatric conditions. Brain Res Bull 2021; 171:196-208. [PMID: 33838211 DOI: 10.1016/j.brainresbull.2021.04.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 03/30/2021] [Accepted: 04/02/2021] [Indexed: 12/19/2022]
Abstract
Sex differences are often observed in psychiatric patients, especially major depressive disorders (MDD), schizophrenia, and developmental disorders, including autism spectrum disorders (ASDs). The prevalence rates between males and females seem variate according to the clinical condition. Although the findings are still incipient, it is suggested that these differences can involve neuroanatomical, neurochemical, and physiological sex differences. In this context, the microbiota-gut-brain axis hypothesis arises to explain some aspects of the complex pathophysiology of neuropsychiatric disorders. The microbiota composition is host-specific and can change conforming to age, sex, diet, medication, exercise, and others. The communication between the brain and the gut is bidirectional and may impact the entire system homeostasis. Many pathways appear to be involved, including neuroanatomic communication, neuroendocrine pathways, immune system, bacteria-derived metabolites, hormones, neurotransmitters, and neurotrophic factors. Although the clinical and preclinical studies are sparse and not very consistent, they suggest that sex differences in the gut microbiota may play an essential role in some neuropsychiatric conditions. Thus, this narrative review has as a mainly aim to show the points sex-related patterns associated to the gut-microbiota-brain axis in the MDD, ASDs, and schizophrenia.
Collapse
Affiliation(s)
- Luana M Manosso
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Jaime Lin
- Experimental Neurology Laboratory, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina, Criciúma, SC, Brazil
| | - Anelise S Carlessi
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Kelen C C Recco
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - João Quevedo
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil; Translational Psychiatry Program, Faillace Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA; Center of Excellence on Mood Disorders, Faillace Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA; Neuroscience Graduate Program, The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
| | - Cinara L Gonçalves
- Experimental Neurology Laboratory, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina, Criciúma, SC, Brazil
| | - Gislaine Z Réus
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil.
| |
Collapse
|
13
|
Kenkel W. Birth signalling hormones and the developmental consequences of caesarean delivery. J Neuroendocrinol 2021; 33:e12912. [PMID: 33145818 PMCID: PMC10590550 DOI: 10.1111/jne.12912] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 10/05/2020] [Accepted: 10/06/2020] [Indexed: 12/12/2022]
Abstract
Rates of delivery by caesarean section (CS) are increasing around the globe and, although several epidemiological associations have already been observed between CS and health outcomes in later life, more are sure to be discovered as this practice continues to gain popularity. The components of vaginal delivery that protect offspring from the negative consequences of CS delivery in later life are currently unknown, although much attention to date has focused on differences in microbial colonisation. Here, we present the case that differing hormonal experiences at birth may also contribute to the neurodevelopmental consequences of CS delivery. Levels of each of the 'birth signalling hormones' (oxytocin, arginine vasopressin, epinephrine, norepinephrine and the glucocorticoids) are lower following CS compared to vaginal delivery, and there is substantial evidence for each that manipulations in early life results in long-term neurodevelopmental consequences. We draw from the research traditions of neuroendocrinology and developmental psychobiology to suggest that the perinatal period is a sensitive period, during which hormones achieve organisational effects. Furthermore, there is much to be learned from research on developmental programming by early-life stress that may inform research on CS, as a result of shared neuroendocrine mechanisms at work. We compare and contrast the effects of early-life stress with those of CS delivery and propose new avenues of research based on the links between the two bodies of literature. The research conducted to date suggests that the differences in hormone signalling seen in CS neonates may produce long-term neurodevelopmental consequences.
Collapse
Affiliation(s)
- William Kenkel
- Department of Psychological and Brain Sciences, University of Delaware, Newark, DE, USA
| |
Collapse
|
14
|
Willis NB, Muñoz CX, Mysonhimer AR, Edwards CG, Wolf PG, Hillman CH, Burd NA, Holscher HD, Khan NA. Hydration Biomarkers Are Related to the Differential Abundance of Fecal Microbiota and Plasma Lipopolysaccharide-Binding Protein in Adults. ANNALS OF NUTRITION AND METABOLISM 2021; 77 Suppl 4:37-45. [PMID: 35226903 DOI: 10.1159/000520478] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 10/27/2021] [Indexed: 01/04/2023]
Abstract
INTRODUCTION Prevalence of chronic hypohydration remains elevated among adults in the USA; however, the health effects of hypohydration in regards to human gut health have not been explored. METHODS This study examined the relationship between total water intake, hydration biomarkers (first-morning urine specific gravity [FMUsg], first-morning urine volume [FMUvol], and plasma copeptin), fecal microbiota, and plasma lipopolysaccharide-binding protein (LBP) in adults (25-45 years, 64% female). Fecal microbiota composition was assessed using 16S rRNA gene sequencing (V4 region). Immunoassays quantified plasma copeptin and LBP in fasted venous blood samples. Dietary variables were measured using 7-day food records. Linear discriminant analysis effect size (LEfSe) analyzed differentially abundant microbiota based on median cutoffs for hydration markers. Multiple linear regressions examined the relationship between LBP and copeptin. RESULTS LEfSe identified 6 common taxa at the genus or species level that were differentially abundant in FMUsg, total water (g/day), or plasma copeptin (µg/mL) groups when split by their median values. Uncultured species in the Bacteroides, Desulfovibrio, Roseburia, Peptococcus, and Akkermansia genera were more abundant in groups that might indicate poorer hydration status. Multivariate linear analyses revealed a positive relationship between plasma copeptin and LBP when controlling confounding variables (F(6,52) = 4.45, p = 0.002, R2 = 0.34). CONCLUSIONS Taxa common between markers are associated with the intestinal mucus layer, which suggests a potential link between hydration status and intestinal mucus homeostasis. The relationship between LBP and copeptin indicates that copeptin may be sensitive to metabolic endotoxemia and potentially gut barrier function.
Collapse
Affiliation(s)
- Nathaniel B Willis
- Division of Nutritional Science, University of Illinois at Urbana-Champaign, Champaign, Illinois, USA,
| | - Colleen X Muñoz
- Department of Health Sciences, University of Hartford, West Hartford, Connecticut, USA
| | - Annemarie R Mysonhimer
- Department of Food Science and Human Nutrition, University of Illinois at Urbana-Champaign, Champaign, Illinois, USA
| | - Caitlyn G Edwards
- Department of Nutritional Sciences, Pennsylvania State University, State College, Pennsylvania, USA
| | - Patricia G Wolf
- Institute for Health Research and Policy, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Charles H Hillman
- Department of Psychology, Northeastern University, Boston, Massachusetts, USA.,Department of Physical Therapy, Movement, & Rehabilitation Sciences, Northeastern University, Boston, Massachusetts, USA
| | - Nicholas A Burd
- Division of Nutritional Science, University of Illinois at Urbana-Champaign, Champaign, Illinois, USA.,Department of Kinesiology and Community Health, University of Illinois at Urbana-Champaign, Champaign, Illinois, USA
| | - Hannah D Holscher
- Division of Nutritional Science, University of Illinois at Urbana-Champaign, Champaign, Illinois, USA.,Department of Food Science and Human Nutrition, University of Illinois at Urbana-Champaign, Champaign, Illinois, USA.,Department of Kinesiology and Community Health, University of Illinois at Urbana-Champaign, Champaign, Illinois, USA
| | - Naiman A Khan
- Division of Nutritional Science, University of Illinois at Urbana-Champaign, Champaign, Illinois, USA.,Department of Kinesiology and Community Health, University of Illinois at Urbana-Champaign, Champaign, Illinois, USA.,Neuroscience Program, University of Illinois at Urbana-Champaign, Champaign, Illinois, USA
| |
Collapse
|
15
|
Sarkar A, Harty S, Johnson KVA, Moeller AH, Carmody RN, Lehto SM, Erdman SE, Dunbar RIM, Burnet PWJ. The role of the microbiome in the neurobiology of social behaviour. Biol Rev Camb Philos Soc 2020; 95:1131-1166. [PMID: 32383208 PMCID: PMC10040264 DOI: 10.1111/brv.12603] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 04/01/2020] [Accepted: 04/02/2020] [Indexed: 12/13/2022]
Abstract
Microbes colonise all multicellular life, and the gut microbiome has been shown to influence a range of host physiological and behavioural phenotypes. One of the most intriguing and least understood of these influences lies in the domain of the microbiome's interactions with host social behaviour, with new evidence revealing that the gut microbiome makes important contributions to animal sociality. However, little is known about the biological processes through which the microbiome might influence host social behaviour. Here, we synthesise evidence of the gut microbiome's interactions with various aspects of host sociality, including sociability, social cognition, social stress, and autism. We discuss evidence of microbial associations with the most likely physiological mediators of animal social interaction. These include the structure and function of regions of the 'social' brain (the amygdala, the prefrontal cortex, and the hippocampus) and the regulation of 'social' signalling molecules (glucocorticoids including corticosterone and cortisol, sex hormones including testosterone, oestrogens, and progestogens, neuropeptide hormones such as oxytocin and arginine vasopressin, and monoamine neurotransmitters such as serotonin and dopamine). We also discuss microbiome-associated host genetic and epigenetic processes relevant to social behaviour. We then review research on microbial interactions with olfaction in insects and mammals, which contribute to social signalling and communication. Following these discussions, we examine evidence of microbial associations with emotion and social behaviour in humans, focussing on psychobiotic studies, microbe-depression correlations, early human development, autism, and issues of statistical power, replication, and causality. We analyse how the putative physiological mediators of the microbiome-sociality connection may be investigated, and discuss issues relating to the interpretation of results. We also suggest that other candidate molecules should be studied, insofar as they exert effects on social behaviour and are known to interact with the microbiome. Finally, we consider different models of the sequence of microbial effects on host physiological development, and how these may contribute to host social behaviour.
Collapse
Affiliation(s)
- Amar Sarkar
- Trinity College, Trinity Street, University of Cambridge, Cambridge, CB2 1TQ, U.K.,Leverhulme Centre for Human Evolutionary Studies, Department of Archaeology, Fitzwilliam Street, University of Cambridge, Cambridge, CB2 1QH, U.K
| | - Siobhán Harty
- Institute of Neuroscience, Trinity College Dublin, Dublin 2, Dublin, Ireland.,School of Psychology, Trinity College Dublin, Dublin 2, Dublin, Ireland
| | - Katerina V-A Johnson
- Department of Experimental Psychology, Radcliffe Observatory Quarter, University of Oxford, Oxford, OX2 6GG, U.K.,Pembroke College, University of Oxford, Oxford, OX1 1DW, U.K.,Department of Psychiatry, Warneford Hospital, University of Oxford, Oxford, OX3 7JX, U.K
| | - Andrew H Moeller
- Department of Ecology and Evolutionary Biology, Corson Hall, Tower Road, Cornell University, Ithaca, NY, 14853, U.S.A
| | - Rachel N Carmody
- Department of Human Evolutionary Biology, Harvard University, Peabody Museum, 11 Divinity Avenue, Cambridge, Massachusetts, 02138, USA
| | - Soili M Lehto
- Psychiatry, University of Helsinki and Helsinki University Hospital, PL 590, FI-00029, Helsinki, Finland.,Department of Psychology and Logopedics, Faculty of Medicine, University of Helsinki, P.O. Box 6, FI-00014, Helsinki, Finland.,Institute of Clinical Medicine/Psychiatry, University of Eastern Finland, P.O. Box 1627, FI-70211, Kuopio, Finland
| | - Susan E Erdman
- Division of Comparative Medicine, Massachusetts Institute of Technology, Building 16-825, 77 Massachusetts Avenue, Cambridge, MA, 02139, U.S.A
| | - Robin I M Dunbar
- Department of Experimental Psychology, Radcliffe Observatory Quarter, University of Oxford, Oxford, OX2 6GG, U.K
| | - Philip W J Burnet
- Department of Psychiatry, Warneford Hospital, University of Oxford, Oxford, OX3 7JX, U.K
| |
Collapse
|
16
|
Microbial transmission in animal social networks and the social microbiome. Nat Ecol Evol 2020; 4:1020-1035. [DOI: 10.1038/s41559-020-1220-8] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Accepted: 05/11/2020] [Indexed: 12/15/2022]
|
17
|
Jaggar M, Rea K, Spichak S, Dinan TG, Cryan JF. You've got male: Sex and the microbiota-gut-brain axis across the lifespan. Front Neuroendocrinol 2020; 56:100815. [PMID: 31805290 DOI: 10.1016/j.yfrne.2019.100815] [Citation(s) in RCA: 121] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 10/16/2019] [Accepted: 11/11/2019] [Indexed: 02/07/2023]
Abstract
Sex is a critical factor in the diagnosis and development of a number of mental health disorders including autism, schizophrenia, depression, anxiety, Parkinson's disease, multiple sclerosis, anorexia nervosa and others; likely due to differences in sex steroid hormones and genetics. Recent evidence suggests that sex can also influence the complexity and diversity of microbes that we harbour in our gut; and reciprocally that our gut microbes can directly and indirectly influence sex steroid hormones and central gene activation. There is a growing emphasis on the role of gastrointestinal microbiota in the maintenance of mental health and their role in the pathogenesis of disease. In this review, we introduce mechanisms by which gastrointestinal microbiota are thought to mediate positive health benefits along the gut-brain axis, we report how they may be modulated by sex, the role they play in sex steroid hormone regulation, and their sex-specific effects in various disorders relating to mental health.
Collapse
Affiliation(s)
- Minal Jaggar
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Kieran Rea
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Simon Spichak
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| | - Timothy G Dinan
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland
| | - John F Cryan
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland.
| |
Collapse
|
18
|
Diviccaro S, Melcangi RC, Giatti S. Post-finasteride syndrome: An emerging clinical problem. Neurobiol Stress 2019; 12:100209. [PMID: 32435662 PMCID: PMC7231981 DOI: 10.1016/j.ynstr.2019.100209] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Revised: 12/11/2019] [Accepted: 12/23/2019] [Indexed: 12/14/2022] Open
Abstract
The presence of side effects during pharmacological treatment is unfortunately a quite common problem. In this review, we focused our attention on adverse events related to 5 alpha-reductase (5α-R) inhibitors (i.e., finasteride and dutasteride), approved for the treatment of benign prostatic hyperplasia and androgenetic alopecia (AGA). Although these drugs are generally well tolerated, many reports described adverse effects in men during treatment, such as sexual dysfunction and mood alteration. In addition, it has been also reported that persistent side effects may occur in some AGA patients. This condition, termed post-finasteride syndrome (PFS) is characterized by sexual side effects (i.e., low libido, erectile dysfunction, decreased arousal and difficulty in achieving orgasm), depression, anxiety and cognitive complaints that are still present despite drug withdrawal. Indeed, some national agencies (e.g., Swedish Medical Products Agency, the Medicines and Healthcare Products Regulatory Agency of UK and the U.S. Food and Drug Administration) required to include multiple persistent side effects within the finasteride labels. As here reported, these observations are mainly based on self-reporting of the symptomatology by the patients and few clinical studies have been performed so far. In addition, molecular mechanisms and/or genetic determinants behind such adverse effects have been poorly explored both in patients and animal models. Therefore, results here discussed indicate that PFS is an emerging clinical problem that needs to be further elucidated.
Collapse
Affiliation(s)
- Silvia Diviccaro
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università Degli Studi di Milano, Milano, Italy
| | - Roberto Cosimo Melcangi
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università Degli Studi di Milano, Milano, Italy
| | - Silvia Giatti
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università Degli Studi di Milano, Milano, Italy
| |
Collapse
|
19
|
Kanbay M, Yilmaz S, Dincer N, Ortiz A, Sag AA, Covic A, Sánchez-Lozada LG, Lanaspa MA, Cherney DZI, Johnson RJ, Afsar B. Antidiuretic Hormone and Serum Osmolarity Physiology and Related Outcomes: What Is Old, What Is New, and What Is Unknown? J Clin Endocrinol Metab 2019; 104:5406-5420. [PMID: 31365096 DOI: 10.1210/jc.2019-01049] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Accepted: 07/25/2019] [Indexed: 12/16/2022]
Abstract
CONTEXT Although the physiology of sodium, water, and arginine vasopressin (AVP), also known as antidiuretic hormone, has long been known, accumulating data suggest that this system operates as a more complex network than previously thought. EVIDENCE ACQUISITION English-language basic science and clinical studies of AVP and osmolarity on the development of kidney and cardiovascular disease and overall outcomes. EVIDENCE SYNTHESIS Apart from osmoreceptors and hypovolemia, AVP secretion is modified by novel factors such as tongue acid-sensing taste receptor cells and brain median preoptic nucleus neurons. Moreover, pharyngeal, esophageal, and/or gastric sensors and gut microbiota modulate AVP secretion. Evidence is accumulating that increased osmolarity, AVP, copeptin, and dehydration are all associated with worse outcomes in chronic disease states such as chronic kidney disease (CKD), diabetes, and heart failure. On the basis of these pathophysiological relationships, an AVP receptor 2 blocker is now licensed for CKD related to polycystic kidney disease. CONCLUSION From a therapeutic perspective, fluid intake may be associated with increased AVP secretion if it is driven by loss of urine concentration capacity or with suppressed AVP if it is driven by voluntary fluid intake. In the current review, we summarize the literature on the relationship between elevated osmolarity, AVP, copeptin, and dehydration with renal and cardiovascular outcomes and underlying classical and novel pathophysiologic pathways. We also review recent unexpected and contrasting findings regarding AVP physiology in an attempt to explain and understand some of these relationships.
Collapse
Affiliation(s)
- Mehmet Kanbay
- Division of Nephrology, Department of Medicine, Koc University School of Medicine, Istanbul, Turkey
| | - Sezen Yilmaz
- Department of Medicine, Koc University School of Medicine, Istanbul, Turkey
| | - Neris Dincer
- Department of Medicine, Koc University School of Medicine, Istanbul, Turkey
| | - Alberto Ortiz
- Dialysis Unit, School of Medicine, IIS-Fundacion Jimenez Diaz, Universidad Autónoma de Madrid, Madrid, Spain
| | - Alan A Sag
- Division of Vascular and Interventional Radiology, Department of Radiology, Duke University Medical Center, Durham, North Carolina
| | - Adrian Covic
- Nephrology Department, Dialysis and Renal Transplant Center, "Dr. C. I. Parhon" University Hospital, "Grigore T. Popa" University of Medicine and Pharmacy, Iasi, Romania
| | - Laura G Sánchez-Lozada
- Laboratory of Renal Physiopathology, Department of Nephrology, INC Ignacio Chávez, Mexico City, Mexico
| | - Miguel A Lanaspa
- Division of Renal Diseases and Hypertension, School of Medicine, University of Colorado Denver, Aurora, Colorado
| | - David Z I Cherney
- Department of Medicine, Division of Nephrology, Toronto General Hospital, University of Toronto, Toronto, Ontario, Canada
| | - Richard J Johnson
- Division of Renal Diseases and Hypertension, School of Medicine, University of Colorado Denver, Aurora, Colorado
| | - Baris Afsar
- Division of Nephrology, Department of Medicine, Suleyman Demirel University School of Medicine, Isparta, Turkey
| |
Collapse
|
20
|
Abstract
PURPOSE OF REVIEW The neurohypophysial endocrine system is identified here as a potential target for therapeutic interventions toward improving obesity-related metabolic dysfunction, given its coinciding pleiotropic effects on psychological, neurological and metabolic systems that are disrupted in obesity. RECENT FINDINGS Copeptin, the C-terminal portion of the precursor of arginine-vasopressin, is positively associated with body mass index and risk of type 2 diabetes. Plasma oxytocin is decreased in obesity and several other conditions of abnormal glucose homeostasis. Recent data also show non-classical tissues, such as myocytes, hepatocytes and β-cells, exhibit responses to oxytocin and vasopressin receptor binding that may contribute to alterations in metabolic function. The modulation of anorexigenic and orexigenic pathways appears to be the dominant mechanism underlying the effects of oxytocin and vasopressin on body weight regulation; however, there are apparent limitations associated with their use in direct pharmacological applications. A clearer picture of their wider physiological effects is needed before either system can be considered for therapeutic use.
Collapse
Affiliation(s)
| | - Faidon Magkos
- Department of Nutrition, Exercise and Sports-Section of Obesity Research, Faculty of Science, University of Copenhagen, Rolighedsvej 26, 1958 Frederiksberg C; Building 2-85, Room H134, Copenhagen, Denmark.
| |
Collapse
|
21
|
Norheim F, Hasin-Brumshtein Y, Vergnes L, Chella Krishnan K, Pan C, Seldin MM, Hui ST, Mehrabian M, Zhou Z, Gupta S, Parks BW, Walch A, Reue K, Hofmann SM, Arnold AP, Lusis AJ. Gene-by-Sex Interactions in Mitochondrial Functions and Cardio-Metabolic Traits. Cell Metab 2019; 29:932-949.e4. [PMID: 30639359 PMCID: PMC6447452 DOI: 10.1016/j.cmet.2018.12.013] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Revised: 08/29/2018] [Accepted: 12/17/2018] [Indexed: 12/20/2022]
Abstract
We studied sex differences in over 50 cardio-metabolic traits in a panel of 100 diverse inbred strains of mice. The results clearly showed that the effects of sex on both clinical phenotypes and gene expression depend on the genetic background. In support of this, genetic loci associated with the traits frequently showed sex specificity. For example, Lyplal1, a gene implicated in human obesity, was shown to underlie a sex-specific locus for diet-induced obesity. Global gene expression analyses of tissues across the panel implicated adipose tissue "beiging" and mitochondrial functions in the sex differences. Isolated mitochondria showed gene-by-sex interactions in oxidative functions, such that some strains (C57BL/6J) showed similar function between sexes, whereas others (DBA/2J and A/J) showed increased function in females. Reduced adipose mitochondrial function in males as compared to females was associated with increased susceptibility to obesity and insulin resistance. Gonadectomy studies indicated that gonadal hormones acting in a tissue-specific manner were responsible in part for the sex differences.
Collapse
Affiliation(s)
- Frode Norheim
- Department of Medicine/Division of Cardiology and Department of Human Genetics, University of California, Los Angeles, Los Angeles, CA, USA; Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Yehudit Hasin-Brumshtein
- Department of Medicine/Division of Cardiology and Department of Human Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Laurent Vergnes
- Department of Human Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Karthickeyan Chella Krishnan
- Department of Medicine/Division of Cardiology and Department of Human Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Calvin Pan
- Department of Medicine/Division of Cardiology and Department of Human Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Marcus M Seldin
- Department of Medicine/Division of Cardiology and Department of Human Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Simon T Hui
- Department of Medicine/Division of Cardiology and Department of Human Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Margarete Mehrabian
- Department of Medicine/Division of Cardiology and Department of Human Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Zhiqiang Zhou
- Department of Medicine/Division of Cardiology and Department of Human Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Sonul Gupta
- Department of Medicine/Division of Cardiology and Department of Human Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Brian W Parks
- Department of Nutritional Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Axel Walch
- Research Unit Analytical Pathology, Helmholtz Zentrum München, Neuherberg, Germany
| | - Karen Reue
- Department of Human Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Susanna M Hofmann
- Institute of Diabetes and Regeneration Research, Helmholtz Zentrum Muenchen, German Research Center for Environmental Health (GmbH), Neuherberg, Germany; German Center for Diabetes Research (DZD), Neuherberg, München 80336, Germany; Medizinische Klinik und Poliklinik IV, Klinikum der Ludwig Maximilian Universität (LMU), Munich, Germany
| | - Arthur P Arnold
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Aldons J Lusis
- Department of Medicine/Division of Cardiology and Department of Human Genetics, University of California, Los Angeles, Los Angeles, CA, USA; Department of Human Genetics, University of California, Los Angeles, Los Angeles, CA, USA; Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
22
|
Yang T, Aquino V, Lobaton GO, Li H, Colon‐Perez L, Goel R, Qi Y, Zubcevic J, Febo M, Richards EM, Pepine CJ, Raizada MK. Sustained Captopril-Induced Reduction in Blood Pressure Is Associated With Alterations in Gut-Brain Axis in the Spontaneously Hypertensive Rat. J Am Heart Assoc 2019; 8:e010721. [PMID: 30755073 PMCID: PMC6405665 DOI: 10.1161/jaha.118.010721] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Accepted: 01/09/2019] [Indexed: 12/21/2022]
Abstract
Background We have demonstrated that the antihypertensive effect of the angiotensin-converting enzyme inhibitor, captopril ( CAP ), is associated with beneficial effects on gut pathology. Coupled with the evidence that CAP exerts prolonged reduction in blood pressure ( BP ) after discontinuation of treatment, we investigate whether persistent beneficial actions of CAP are linked to alterations of gut microbiota and improvement of hypertension-induced gut pathology. Methods and Results Spontaneously hypertensive rats ( SHR ) and Wistar Kyoto rats were treated with CAP (250 mg/kg/day) for 4 weeks followed by withdrawal for 16 weeks. Gut microbiota, gut pathology, BP, and brain neuronal activity were assessed. CAP resulted in a ≈60 mm Hg decrease in systolic BP after 3 weeks of treatment in SHR , and the decrease remained significant at least 5 weeks after CAP withdrawal. In contrast, CAP caused modest decrease in systolic BP in Wistar Kyoto. 16S rRNA gene-sequencing-based gut microbial analyses in SHR showed sustained alteration of gut microbiota and increase in Allobaculum after CAP withdrawal. Phylogenetic investigation of communities by reconstruction of unobserved states analysis revealed significant increase in bacterial sporulation upon CAP treatment in SHR . These were associated with persistent improvement in gut pathology and permeability. Furthermore, manganese-enhanced magnetic resonance imaging showed significantly decreased neuronal activity in the posterior pituitary of SHR 4 weeks after withdrawal. Conclusions Decreased BP , altered gut microbiota, improved gut pathology and permeability, and dampened posterior pituitary neuronal activity were maintained after CAP withdrawal in the SHR . They suggest that CAP influences the brain-gut axis to maintain the sustained antihypertensive effect of CAP after withdrawal.
Collapse
Affiliation(s)
- Tao Yang
- Department of Physiology and Functional GenomicsCollege of MedicineUniversity of FloridaGainesvilleFL
| | - Victor Aquino
- Department of Physiology and Functional GenomicsCollege of MedicineUniversity of FloridaGainesvilleFL
| | - Gilberto O. Lobaton
- Department of Physiology and Functional GenomicsCollege of MedicineUniversity of FloridaGainesvilleFL
| | - Hongbao Li
- Department of Physiology and Functional GenomicsCollege of MedicineUniversity of FloridaGainesvilleFL
- Department of Physiology and PathophysiologySchool of Basic Medical SciencesXi'an Jiaotong UniversityXi'anChina
| | - Luis Colon‐Perez
- Department of PsychiatryCollege of MedicineUniversity of FloridaGainesvilleFL
| | - Ruby Goel
- Department of Physiology and Functional GenomicsCollege of MedicineUniversity of FloridaGainesvilleFL
| | - Yanfei Qi
- Division of Cardiovascular MedicineUniversity of FloridaGainesvilleFL
| | - Jasenka Zubcevic
- Department of Physiological SciencesCollege of Veterinary MedicineUniversity of FloridaGainesvilleFL
| | - Marcelo Febo
- Department of PsychiatryCollege of MedicineUniversity of FloridaGainesvilleFL
| | - Elaine M. Richards
- Department of Physiology and Functional GenomicsCollege of MedicineUniversity of FloridaGainesvilleFL
| | - Carl J. Pepine
- Division of Cardiovascular MedicineUniversity of FloridaGainesvilleFL
| | - Mohan K. Raizada
- Department of Physiology and Functional GenomicsCollege of MedicineUniversity of FloridaGainesvilleFL
| |
Collapse
|
23
|
Zhou W, Xu H, Zhan L, Lu X, Zhang L. Dynamic Development of Fecal Microbiome During the Progression of Diabetes Mellitus in Zucker Diabetic Fatty Rats. Front Microbiol 2019; 10:232. [PMID: 30837966 PMCID: PMC6382700 DOI: 10.3389/fmicb.2019.00232] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Accepted: 01/28/2019] [Indexed: 12/15/2022] Open
Abstract
Background: Although substantial efforts have been made to link the gut microbiota to type 2 diabetes, dynamic changes in the fecal microbiome under the pathological conditions of diabetes have not been investigated. Methods: Four male Zucker diabetic fatty (ZDF) rats received Purina 5008 chow [protein = 23.6%, Nitrogen-Free Extract (by difference) = 50.3%, fiber (crude) = 3.3%, ash = 6.1%, fat (ether extract) = 6.7%, and fat (acid hydrolysis) = 8.1%] for 8 weeks. A total of 32 stool samples were collected from weeks 8 to 15 in four rats. To decipher the microbial populations in these samples, we used a 16S rRNA gene sequencing approach. Results: Microbiome analysis showed that the changes in the fecal microbiome were associated with age and disease progression. In all the stages from 8 to 15 weeks, phyla Firmicutes, Bacteroidetes, Actinobacteria, and Proteobacteria primarily dominated the fecal microbiome of the rats. Although Lactobacillus and Turicibacter were the predominant genera in 8- to 10-week-old rats, Bifidobacterium, Lactobacillus, Ruminococcus, and Allobaculum were the most abundant genera in 15-week-old rats. Of interest, compared to the earlier weeks, relatively greater diversity (at the genus level) was observed at 10 weeks of age. Although the microbiome of 12-week-old rats had the highest diversity, the diversity in 13–15-week-old rats was reduced. Spearman’s correlation analysis showed that F/B was negatively correlated with age. Random blood glucose was negatively correlated with Lactobacillus and Turicibacter but positively correlated with Ruminococcus and Allobaculum and Simpson’s diversity index. Conclusion: We demonstrated the time-dependent alterations of the abundance and diversity of the fecal microbiome during the progression of diabetes in ZDF rats. At the genus level, dynamic changes were observed. We believe that this work will enhance our understanding of fecal microbiome development in ZDF rats and help to further analyze the role of the microbiome in metabolic diseases. Furthermore, our work may also provide an effective strategy for the clinical treatment of diabetes through microbial intervention.
Collapse
Affiliation(s)
- Wen Zhou
- Modern Research Laboratory of Spleen Visceral Manifestations Theory, Basic Medical College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Huiying Xu
- Modern Research Laboratory of Spleen Visceral Manifestations Theory, Basic Medical College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Libin Zhan
- Modern Research Laboratory of Spleen Visceral Manifestations Theory, Basic Medical College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Xiaoguang Lu
- Department of Emergency Medicine, Zhongshan Hospital, Dalian University, Dalian, China
| | - Lijing Zhang
- Modern Research Laboratory of Spleen Visceral Manifestations Theory, Basic Medical College, Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
24
|
Defining Dysbiosis in Disorders of Movement and Motivation. J Neurosci 2018; 38:9414-9422. [PMID: 30381433 PMCID: PMC6209841 DOI: 10.1523/jneurosci.1672-18.2018] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Revised: 09/28/2018] [Accepted: 09/28/2018] [Indexed: 02/06/2023] Open
Abstract
The gut microbiota has emerged as a critical player in shaping and modulating brain function and has been shown to influence numerous behaviors, including anxiety and depression-like behaviors, sociability, and cognition. However, the effects of the gut microbiota on specific disorders associated with thalamo-cortico-basal ganglia circuits, ranging from compulsive behavior and addiction to altered sensation and motor output, are only recently being explored. Wholesale depletion and alteration of gut microbial communities in rodent models of disorders, such as Parkinson's disease, autism, and addiction, robustly affect movement and motivated behavior. A new frontier therefore lies in identifying specific microbial alterations that affect these behaviors and understanding the underlying mechanisms of action. Comparing alterations in gut microbiota across multiple basal-ganglia associated disease states allows for identification of common mechanistic pathways that may interact with distinct environmental and genetic risk factors to produce disease-specific outcomes.
Collapse
|
25
|
Abstract
PURPOSE OF REVIEW Sex differences are pervasive in metabolic and cardiovascular traits, yet they have often been ignored in human and animal model research. Sex differences can arise from reversible hormonal effects, from irreversible organizational (developmental) processes, and from gene expression differences from the X and Y chromosomes. We briefly review our current understanding of the impact of these factors in metabolic traits and disorders, with an emphasis on the recent literature. RECENT FINDINGS Novel sex differences continue to be identified for metabolic and cardiovascular traits. For example, it is now clear that gut microbiota tend to differ between men and women, with potentially large implications for disease susceptibility. Also, tissue-specific gene regulation differs between men and women, contributing to differential metabolism. These new insights will open up personalized therapeutic avenues for cardiometabolic diseases. SUMMARY Sex differences in body fat distribution, glucose homeostasis, insulin signaling, ectopic fat accumulation, and lipid metabolism during normal growth and in response to hormonal or nutritional imbalance are mediated partly through sex hormones and the sex chromosome complement. Most of these differences are mediated in a tissue-specific manner. Important future goals are to better understand the interactions between genetic variation and sex differences, and to bring an understanding of sex differences into clinical practice.
Collapse
Affiliation(s)
| | | | - Aldons J. Lusis
- Department of Medicine/Division of Cardiology
- Department of Micro-biology, Immunology and Molecular Genetics
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, California, USA
| |
Collapse
|
26
|
Tetel MJ, de Vries GJ, Melcangi RC, Panzica G, O'Mahony SM. Steroids, stress and the gut microbiome-brain axis. J Neuroendocrinol 2018; 30:10.1111/jne.12548. [PMID: 29024170 PMCID: PMC6314837 DOI: 10.1111/jne.12548] [Citation(s) in RCA: 95] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Revised: 10/07/2017] [Accepted: 10/09/2017] [Indexed: 12/14/2022]
Abstract
It is becoming well established that the gut microbiome has a profound impact on human health and disease. In this review, we explore how steroids can influence the gut microbiota and, in turn, how the gut microbiota can influence hormone levels. Within the context of the gut microbiome-brain axis, we discuss how perturbations in the gut microbiota can alter the stress axis and behaviour. In addition, human studies on the possible role of gut microbiota in depression and anxiety are examined. Finally, we present some of the challenges and important questions that need to be addressed by future research in this exciting new area at the intersection of steroids, stress, gut-brain axis and human health.
Collapse
Affiliation(s)
- M J Tetel
- Neuroscience Program, Wellesley College, Wellesley, MA, USA
| | - G J de Vries
- Neuroscience Institute, Georgia State University, Atlanta, GA, USA
| | - R C Melcangi
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| | - G Panzica
- Dipartimento di Neuroscienze "Rita Levi Montalcini", Neuroscience Institute Cavalieri Ottolenghi (NICO), Università degli Studi di Torino, Orbassano, Italy
| | - S M O'Mahony
- Department of Anatomy and Neuroscience, APC Microbiome Institute, University College Cork, Cork, Ireland
| |
Collapse
|