1
|
Xie L, Wu Q, Li K, Khan MAS, Zhang A, Sinha B, Li S, Chang SL, Brody DL, Grinstaff MW, Zhou S, Alterovitz G, Liu P, Wang X. Tryptophan Metabolism in Alzheimer's Disease with the Involvement of Microglia and Astrocyte Crosstalk and Gut-Brain Axis. Aging Dis 2024; 15:2168-2190. [PMID: 38916729 PMCID: PMC11346405 DOI: 10.14336/ad.2024.0134] [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: 02/02/2024] [Accepted: 05/03/2024] [Indexed: 06/26/2024] Open
Abstract
Alzheimer's disease (AD) is an age-dependent neurodegenerative disease characterized by extracellular Amyloid Aβ peptide (Aβ) deposition and intracellular Tau protein aggregation. Glia, especially microglia and astrocytes are core participants during the progression of AD and these cells are the mediators of Aβ clearance and degradation. The microbiota-gut-brain axis (MGBA) is a complex interactive network between the gut and brain involved in neurodegeneration. MGBA affects the function of glia in the central nervous system (CNS), and microbial metabolites regulate the communication between astrocytes and microglia; however, whether such communication is part of AD pathophysiology remains unknown. One of the potential links in bilateral gut-brain communication is tryptophan (Trp) metabolism. The microbiota-originated Trp and its metabolites enter the CNS to control microglial activation, and the activated microglia subsequently affect astrocyte functions. The present review highlights the role of MGBA in AD pathology, especially the roles of Trp per se and its metabolism as a part of the gut microbiota and brain communications. We (i) discuss the roles of Trp derivatives in microglia-astrocyte crosstalk from a bioinformatics perspective, (ii) describe the role of glia polarization in the microglia-astrocyte crosstalk and AD pathology, and (iii) summarize the potential of Trp metabolism as a therapeutic target. Finally, we review the role of Trp in AD from the perspective of the gut-brain axis and microglia, as well as astrocyte crosstalk, to inspire the discovery of novel AD therapeutics.
Collapse
Affiliation(s)
- Lushuang Xie
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA.
- Acupuncture and Moxibustion College, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 610075, China.
| | - Qiaofeng Wu
- Acupuncture and Moxibustion College, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 610075, China.
| | - Kelin Li
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA.
- Department of Chemistry, Boston University, Boston, MA 02215, USA.
| | - Mohammed A. S. Khan
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA.
| | - Andrew Zhang
- Biomedical Cybernetics Laboratory, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA.
| | - Bharati Sinha
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA.
| | - Sihui Li
- Acupuncture and Moxibustion College, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 610075, China.
| | - Sulie L. Chang
- Department of Biological Sciences, Institute of NeuroImmune Pharmacology, Seton Hall University, South Orange, NJ 07079, USA.
| | - David L. Brody
- Department of Neurology, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA.
| | | | - Shuanhu Zhou
- Harvard Medical School, Harvard Stem Cell Institute, Boston, MA 02115, USA.
| | - Gil Alterovitz
- Biomedical Cybernetics Laboratory, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA.
| | - Pinghua Liu
- Department of Chemistry, Boston University, Boston, MA 02215, USA.
| | - Xin Wang
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
2
|
Romero-Ramírez L, Mey J. Emerging Roles of Bile Acids and TGR5 in the Central Nervous System: Molecular Functions and Therapeutic Implications. Int J Mol Sci 2024; 25:9279. [PMID: 39273226 PMCID: PMC11395147 DOI: 10.3390/ijms25179279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 08/15/2024] [Accepted: 08/17/2024] [Indexed: 09/15/2024] Open
Abstract
Bile acids (BAs) are cholesterol derivatives synthesized in the liver and released into the digestive tract to facilitate lipid uptake during the digestion process. Most of these BAs are reabsorbed and recycled back to the liver. Some of these BAs progress to other tissues through the bloodstream. The presence of BAs in the central nervous system (CNS) has been related to their capacity to cross the blood-brain barrier (BBB) from the systemic circulation. However, the expression of enzymes and receptors involved in their synthesis and signaling, respectively, support the hypothesis that there is an endogenous source of BAs with a specific function in the CNS. Over the last decades, BAs have been tested as treatments for many CNS pathologies, with beneficial effects. Although they were initially reported as neuroprotective substances, they are also known to reduce inflammatory processes. Most of these effects have been related to the activation of the Takeda G protein-coupled receptor 5 (TGR5). This review addresses the new challenges that face BA research for neuroscience, focusing on their molecular functions. We discuss their endogenous and exogenous sources in the CNS, their signaling through the TGR5 receptor, and their mechanisms of action as potential therapeutics for neuropathologies.
Collapse
Affiliation(s)
- Lorenzo Romero-Ramírez
- Laboratorio de Regeneración Neuronal, Hospital Nacional de Parapléjicos, Servicio de Salud de Castilla-La Mancha, 45071 Toledo, Spain
| | - Jörg Mey
- Laboratorio de Regeneración Neuronal, Hospital Nacional de Parapléjicos, Servicio de Salud de Castilla-La Mancha, 45071 Toledo, Spain
- EURON Graduate School of Neuroscience, Maastricht University, 6229 ER Maastricht, The Netherlands
| |
Collapse
|
3
|
Xuan L, Hu Z, Jiang Z, Zhang C, Sun X, Ming W, Liu H, Qiao R, Shen L, Liu S, Wang G, Wen L, Luan Z, Yin J. Pregnane X receptor (PXR) deficiency protects against spinal cord injury by activating NRF2/HO-1 pathway. CNS Neurosci Ther 2023; 29:3460-3478. [PMID: 37269088 PMCID: PMC10580351 DOI: 10.1111/cns.14279] [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: 10/28/2022] [Revised: 05/12/2023] [Accepted: 05/14/2023] [Indexed: 06/04/2023] Open
Abstract
INTRODUCTION As a devastating neurological disease, spinal cord injury (SCI) results in severe tissue loss and neurological dysfunction. Pregnane X receptor (PXR) is a ligand-activated nuclear receptor with a major regulatory role in xenobiotic and endobiotic metabolism and recently has been implicated in the central nervous system. In the present study, we aimed to investigate the role and mechanism of PXR in SCI. METHODS The clip-compressive SCI model was performed in male wild-type C57BL/6 (PXR+/+ ) and PXR-knockout (PXR-/- ) mice. The N2a H2 O2 -induced injury model mimicked the pathological process of SCI in vitro. Pregnenolone 16α-carbonitrile (PCN), a mouse-specific PXR agonist, was used to activate PXR in vivo and in vitro. The siRNA was applied to knock down the PXR expression in vitro. Transcriptome sequencing analysis was performed to discover the relevant mechanism, and the NRF2 inhibitor ML385 was used to validate the involvement of PXR in influencing the NRF2/HO-1 pathway in the SCI process. RESULTS The expression of PXR decreased after SCI and reached a minimum on the third day. In vivo, PXR knockout significantly improved the motor function of mice after SCI, meanwhile, inhibited apoptosis, inflammation, and oxidative stress induced by SCI. On the contrary, activation of PXR by PCN negatively influenced the recovery of SCI. Mechanistically, transcriptome sequencing analysis revealed that PXR activation downregulated the mRNA level of heme oxygenase-1 (HO-1) after SCI. We further verified that PXR deficiency activated the NRF2/HO-1 pathway and PXR activation inhibited this pathway in vitro. CONCLUSION PXR is involved in the recovery of motor function after SCI by regulating NRF2/HO-1 pathway.
Collapse
Affiliation(s)
- Li‐Na Xuan
- Department of Neurosurgerythe Second Affiliated Hospital of Dalian Medical UniversityDalianChina
- Epileptic Center of Liaoningthe Second Affiliated Hospital of Dalian Medical UniversityDalianChina
| | - Zhen‐Xin Hu
- Department of OrthopedicsThe First Affiliated Hospital of Dalian Medical UniversityDalianChina
| | - Zhen‐Fu Jiang
- Department of Neurosurgerythe Second Affiliated Hospital of Dalian Medical UniversityDalianChina
- Epileptic Center of Liaoningthe Second Affiliated Hospital of Dalian Medical UniversityDalianChina
| | - Cong Zhang
- Advanced Institute for Medical SciencesDalian Medical UniversityDalianChina
| | - Xiao‐Wan Sun
- Advanced Institute for Medical SciencesDalian Medical UniversityDalianChina
| | - Wen‐Hua Ming
- Advanced Institute for Medical SciencesDalian Medical UniversityDalianChina
| | - Hui‐Tao Liu
- Department of OrthopedicsTaizhou Hospital of Zhejiang ProvinceLinhaiChina
| | - Rong‐Fang Qiao
- Advanced Institute for Medical SciencesDalian Medical UniversityDalianChina
| | - Lin‐Jie Shen
- Department of GastroenterologyNingbo First HospitalNingboChina
| | - Shao‐Bo Liu
- Department of Neurosurgerythe Second Affiliated Hospital of Dalian Medical UniversityDalianChina
- Epileptic Center of Liaoningthe Second Affiliated Hospital of Dalian Medical UniversityDalianChina
| | - Guan‐Yu Wang
- Department of Neurosurgerythe Second Affiliated Hospital of Dalian Medical UniversityDalianChina
- Epileptic Center of Liaoningthe Second Affiliated Hospital of Dalian Medical UniversityDalianChina
| | - Lin Wen
- Advanced Institute for Medical SciencesDalian Medical UniversityDalianChina
| | - Zhi‐Lin Luan
- Advanced Institute for Medical SciencesDalian Medical UniversityDalianChina
- Dalian Key Laboratory for Nuclear Receptors in Major Metabolic DiseasesDalianChina
| | - Jian Yin
- Department of Neurosurgerythe Second Affiliated Hospital of Dalian Medical UniversityDalianChina
- Epileptic Center of Liaoningthe Second Affiliated Hospital of Dalian Medical UniversityDalianChina
| |
Collapse
|
4
|
Ren A, Wu T, Wang Y, Fan Q, Yang Z, Zhang S, Cao Y, Cui G. Integrating animal experiments, mass spectrometry and network-based approach to reveal the sleep-improving effects of Ziziphi Spinosae Semen and γ-aminobutyric acid mixture. Chin Med 2023; 18:99. [PMID: 37573423 PMCID: PMC10422734 DOI: 10.1186/s13020-023-00814-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 07/30/2023] [Indexed: 08/14/2023] Open
Abstract
BACKGROUND Ziziphi Spinosae Semen (ZSS) is a plant widely used as medicine and food in Asian countries due to its numerous health benefits. γ-aminobutyric acid (GABA), a non-proteinaceous amino acid, is one of the major inhibitory neurotransmitters with a relaxant function. In this study, a system pharmacology approach was employed to assess the effects of a mixture composed of ZSS and GABA (ZSSG) on sleep improvement. METHODS Mice were divided into five groups (n = 10) and received either no treatment, sodium pentobarbital, or sodium barbital with diazepam or ZSSG. The effects of ZSSG on sleep quality were evaluated in mice, and differential metabolites associated with sleep were identified among the control, ZSS, GABA, and ZSSG groups. Additionally, network-based ingredient-insomnia proximity analysis was applied to explore the major ingredients. RESULTS ZSSG significantly improved sleep quality by decreasing sleep latency and prolonging sleep duration in sodium pentobarbital-induced sleeping mouse model (P < 0.05). ZSSG significantly enhanced the brain content of GABA in mice. Furthermore, ZSSG also significantly decreased sleep latency-induced by sodium barbital in mice (P < 0.05). Metabolic analysis revealed significant differences in 10 metabolites between ZSSG group and the groups administering ZSS or GABA. Lastly, using the network-based ingredient screening model, we discovered potential four active ingredients and three pairwise ingredient combinations with synergistic effect on insomnia from ZSSG among 85 ingredients identified by UPLC-Q/TOF-MS. Also, we have constructed an online computation platform. CONCLUSION Our data demonstrated that ZSSG improved the sleeping quality of mice and helped to balance metabolic disorders-associated with sleep disorders. Moreover, based on the network-based prediction method, the four potential active ingredients in ZSSG could serve as quality markers-associated with insomnia. The network-based framework may open up a new avenue for the discovery of active ingredients of herbal medicine for treating complex chronic diseases or symptoms, such as insomnia.
Collapse
Affiliation(s)
- Airong Ren
- Department of Bioengineering, Zhuhai Campus of Zunyi Medical University, Zhuhai, 519000, Guangdong, China
| | - Tingbiao Wu
- Department of Bioengineering, Zhuhai Campus of Zunyi Medical University, Zhuhai, 519000, Guangdong, China
| | - Yarong Wang
- Department of Bioengineering, Zhuhai Campus of Zunyi Medical University, Zhuhai, 519000, Guangdong, China
| | - Qing Fan
- Basic Medical Science Department, Zhuhai Campus of Zunyi Medical University, Zhuhai, 519000, Guangdong, China
| | - Zhenhao Yang
- Department of Bioengineering, Zhuhai Campus of Zunyi Medical University, Zhuhai, 519000, Guangdong, China
| | - Shixun Zhang
- Department of Bioengineering, Zhuhai Campus of Zunyi Medical University, Zhuhai, 519000, Guangdong, China
| | - Yongjun Cao
- Department of Bioengineering, Zhuhai Campus of Zunyi Medical University, Zhuhai, 519000, Guangdong, China
| | - Guozhen Cui
- Department of Bioengineering, Zhuhai Campus of Zunyi Medical University, Zhuhai, 519000, Guangdong, China.
| |
Collapse
|
5
|
Lukić I, Ivković S, Mitić M, Adžić M. Tryptophan metabolites in depression: Modulation by gut microbiota. Front Behav Neurosci 2022; 16:987697. [PMID: 36172468 PMCID: PMC9510596 DOI: 10.3389/fnbeh.2022.987697] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 08/19/2022] [Indexed: 11/13/2022] Open
Abstract
Clinical depression is a multifactorial disorder and one of the leading causes of disability worldwide. The alterations in tryptophan metabolism such as changes in the levels of serotonin, kynurenine, and kynurenine acid have been implicated in the etiology of depression for more than 50 years. In recent years, accumulated evidence has revealed that gut microbial communities, besides being essential players in various aspects of host physiology and brain functioning are also implicated in the etiology of depression, particularly through modulation of tryptophan metabolism. Therefore, the aim of this review is to summarize the evidence of the role of gut bacteria in disturbed tryptophan metabolism in depression. We summed up the effects of microbiota on serotonin, kynurenine, and indole pathway of tryptophan conversion relevant for understanding the pathogenesis of depressive behavior. Moreover, we reviewed data regarding the therapeutic effects of probiotics, particularly through the regulation of tryptophan metabolites. Taken together, these findings can open new possibilities for further improvement of treatments for depression based on the microbiota-mediated modulation of the tryptophan pathway.
Collapse
|
6
|
Ashour NH, El-Tanbouly DM, El Sayed NS, Khattab MM. Roflumilast ameliorates cognitive deficits in a mouse model of amyloidogenesis and tauopathy: Involvement of nitric oxide status, Aβ extrusion transporter ABCB1, and reversal by PKA inhibitor H89. Prog Neuropsychopharmacol Biol Psychiatry 2021; 111:110366. [PMID: 34051306 DOI: 10.1016/j.pnpbp.2021.110366] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 05/01/2021] [Accepted: 05/23/2021] [Indexed: 01/13/2023]
Abstract
The biological cascade of second messenger-cyclic adenosine monophosphate (cAMP) -as a molecular mechanism implicated in memory and learning regulation has captured the attention of neuroscientists worldwide. cAMP triggers its foremost effector, protein kinase A (PKA), resulting in the activation of innumerable downstream targets. Roflumilast (ROF), a phosphodiesterase 4 inhibitor, has demonstrated a greater efficiency in enhancing cAMP signaling in various neurological disorders. This study was conducted to identify various downstream targets of PKA as mechanistic tools through which ROF could hinder the progressive cognitive impairment following central streptozotocin (STZ) administration in mice. Animals were injected with STZ (3 mg/kg/i.c.v) once. Five hours later, mice received ROF (0.4 mg/kg) with or without the PKA inhibitor, H89, for 21 days. ROF highly preserved the structure of hippocampal neurons. It improved the ability of mice to develop short-term memories and retrieve spatial memories in Y-maze and Morris water maze tests, respectively. ROF enhanced the gene expression of ABCB1 transporters and pregnane X receptors (PXR), and hampered Aβ accumulation in hippocampus. Simultaneously, it interfered with the processes of tau phosphorylation and nitration. This effect was associated with an upsurge in hippocampal arginase activity as well as a decline in glycogen synthase kinase-3β activity, nitric oxide synthase (NOS) activity, and inducible NOS expression. Contrariwise, ROF's beneficial effects were utterly abolished by co-administration of H89. In conclusion, boosting PKA, by ROF, modulated PXR/ABCB1 expression and arginase/NOS activities to restrict the main post-translational modifications of tau, Aβ deposition and, accordingly, cognitive deterioration of sporadic Alzheimer's disease.
Collapse
Affiliation(s)
- Nada H Ashour
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Dalia M El-Tanbouly
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Nesrine S El Sayed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt.
| | - Mahmoud M Khattab
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| |
Collapse
|
7
|
Forner-Piquer I, Klement W, Gangarossa G, Zub E, de Bock F, Blaquiere M, Maurice T, Audinat E, Faucherre A, Lasserre F, Ellero-Simatos S, Gamet-Payrastre L, Jopling C, Marchi N. Varying modalities of perinatal exposure to a pesticide cocktail elicit neurological adaptations in mice and zebrafish. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2021; 278:116755. [PMID: 33725534 DOI: 10.1016/j.envpol.2021.116755] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 02/08/2021] [Accepted: 02/12/2021] [Indexed: 06/12/2023]
Abstract
Epidemiological indications connect maternal and developmental presence or exposure to pesticides with an increased risk for a spectrum of neurological trajectories. To provide pre-clinical data in support of this hypothesis, we used two distinct experimental models. First, female and male mice were fed immediately prior to mating, and the resulting pregnant dams were continously fed during gestation and lactation periods using chow pellets containing a cocktail of six pesticides at tolerable daily intake levels. Male and female offspring were then tracked for behavioral and in vivo electrophysiological adaptations. Second, a zebrafish model allowed us to screen toxicity and motor-behavior outcomes specifically associated with the developmental exposure to a low-to-high concentration range of the cocktail and of each individual pesticide. Here, we report anxiety-like behavior in aging male mice maternally exposed to the cocktail, as compared to age and gender matched sham animals. In parallel, in vivo electrocorticography revealed a decrease in gamma (40-80 Hz) and an increase of theta (6-9 Hz) waves, delineating a long-term, age-dependent, neuronal slowing. Neurological changes were not accompanied by brain structural malformations. Next, by using zebrafish larvae, we showed an increase of all motor-behavioral parameters resulting from the developmental exposure to 10 μg/L of pesticide cocktail, an outcome that was not associated with midbrain structural or neurovascular modifications as assessed by in vivo 2-photon microscopy. When screening each pesticide, chlorpyrifos elicited modifications of swimming parameters at 0.1 μg/L, while other components provoked changes from 0.5 μg/L. Ziram was the single most toxic component inducing developmental malformations and mortality at 10 μg/L. Although we have employed non-equivalent modalities and timing of exposure in two dissimilar experimental models, these outcomes indicate that presence of a pesticide cocktail during perinatal periods represents an element promoting behavioral and neurophysiological modifications. The study limitations and the possible pertinence of our findings to ecotoxicology and public health are critically discussed.
Collapse
Affiliation(s)
- Isabel Forner-Piquer
- Institute of Functional Genomics, University of Montpellier, CNRS, INSERM, Montpellier, France
| | - Wendy Klement
- Institute of Functional Genomics, University of Montpellier, CNRS, INSERM, Montpellier, France
| | | | - Emma Zub
- Institute of Functional Genomics, University of Montpellier, CNRS, INSERM, Montpellier, France
| | - Frederic de Bock
- Institute of Functional Genomics, University of Montpellier, CNRS, INSERM, Montpellier, France
| | - Marine Blaquiere
- Institute of Functional Genomics, University of Montpellier, CNRS, INSERM, Montpellier, France
| | - Tangui Maurice
- MMDN, University of Montpellier, EPHE, INSERM, UMR_S1198, Montpellier, France
| | - Etienne Audinat
- Institute of Functional Genomics, University of Montpellier, CNRS, INSERM, Montpellier, France
| | - Adèle Faucherre
- Institute of Functional Genomics, University of Montpellier, CNRS, INSERM, Montpellier, France
| | - Frederic Lasserre
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, UPS, 31300, Toulouse, France
| | - Sandrine Ellero-Simatos
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, UPS, 31300, Toulouse, France
| | - Laurence Gamet-Payrastre
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, UPS, 31300, Toulouse, France
| | - Chris Jopling
- Institute of Functional Genomics, University of Montpellier, CNRS, INSERM, Montpellier, France
| | - Nicola Marchi
- Institute of Functional Genomics, University of Montpellier, CNRS, INSERM, Montpellier, France.
| |
Collapse
|
8
|
Klement W, Oliviero F, Gangarossa G, Zub E, De Bock F, Forner-Piquer I, Blaquiere M, Lasserre F, Pascussi JM, Maurice T, Audinat E, Ellero-Simatos S, Gamet-Payrastre L, Mselli-Lakhal L, Marchi N. Life-long Dietary Pesticide Cocktail Induces Astrogliosis Along with Behavioral Adaptations and Activates p450 Metabolic Pathways. Neuroscience 2020; 446:225-237. [PMID: 32736067 DOI: 10.1016/j.neuroscience.2020.07.039] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Revised: 07/20/2020] [Accepted: 07/21/2020] [Indexed: 02/06/2023]
Abstract
Exposure to environmental contaminants is a public health concern. However, pre-clinical studies that examine the impact of pesticides at low-dose and the long-term consequences are uncommon. Here, C57BL6/j male and female mice were daily fed from weaning and up to 12 months, corresponding to early-childhood into middle-age in humans, using chow pellets containing a cocktail of pesticides at tolerable daily intake levels. We found that 12 months of dietary exposure to pesticides was associated with a moderate perenchymal or perivascular astrogliosis in specific hippocampal sub-regions. The expression of platelet-derived growth factor receptor beta was modified at the perivascular level. Examination of Iba1+ microglial cells did not reveal sizeable changes. Concomitantly to astrogliosis, spontaneous spatial memory and sociability were modified in males at 12 months of dietary exposure to pesticides. Telemetry electrocorticograhic explorations ruled out the presence of epileptiform activity or theta-gamma wave modifications in these conditions. Long-term pesticides impacted the periphery where the hepatic P450 metabolic cytochromes Cyp4a14 and Cyp4a10 were significantly upregulated in male and female mice during the 12 months of exposure. The expression of β-oxidation genes, such as Acox1, Cpt1a and Eci, was also significantly increased in male and female mice in response to pesticides. Collectively, our results indicate that a life-long exposure to a pesticide cocktail elicits sex-dependent, spatio-temporally restricted brain modifications and significant activation of P450 pathways in the periphery. These brain-peripheral adjustments are discussed as time or age-dependent vulnerability elements.
Collapse
Affiliation(s)
- Wendy Klement
- Institute of Functional Genomics (UMR 5203 CNRS - U 1191 INSERM, University of Montpellier), Montpellier, France
| | - Fabiana Oliviero
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, UPS, 31300 Toulouse, France
| | | | - Emma Zub
- Institute of Functional Genomics (UMR 5203 CNRS - U 1191 INSERM, University of Montpellier), Montpellier, France
| | - Frederic De Bock
- Institute of Functional Genomics (UMR 5203 CNRS - U 1191 INSERM, University of Montpellier), Montpellier, France
| | - Isabel Forner-Piquer
- Institute of Functional Genomics (UMR 5203 CNRS - U 1191 INSERM, University of Montpellier), Montpellier, France
| | - Marine Blaquiere
- Institute of Functional Genomics (UMR 5203 CNRS - U 1191 INSERM, University of Montpellier), Montpellier, France
| | - Frederic Lasserre
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, UPS, 31300 Toulouse, France
| | - Jean-Marc Pascussi
- Institute of Functional Genomics (UMR 5203 CNRS - U 1191 INSERM, University of Montpellier), Montpellier, France
| | - Tangui Maurice
- MMDN, University of Montpellier, EPHE, INSERM, UMR_S1198, Montpellier, France
| | - Etienne Audinat
- Institute of Functional Genomics (UMR 5203 CNRS - U 1191 INSERM, University of Montpellier), Montpellier, France
| | - Sandrine Ellero-Simatos
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, UPS, 31300 Toulouse, France
| | - Laurence Gamet-Payrastre
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, UPS, 31300 Toulouse, France
| | - Laila Mselli-Lakhal
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, UPS, 31300 Toulouse, France
| | - Nicola Marchi
- Institute of Functional Genomics (UMR 5203 CNRS - U 1191 INSERM, University of Montpellier), Montpellier, France.
| |
Collapse
|
9
|
Marinelli R, Torquato P, Bartolini D, Mas-Bargues C, Bellezza G, Gioiello A, Borras C, De Luca A, Fallarino F, Sebastiani B, Mani S, Sidoni A, Viña J, Leri M, Bucciantini M, Nardiello P, Casamenti F, Galli F. Garcinoic acid prevents β-amyloid (Aβ) deposition in the mouse brain. J Biol Chem 2020; 295:11866-11876. [PMID: 32616652 PMCID: PMC7450134 DOI: 10.1074/jbc.ra120.013303] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 06/24/2020] [Indexed: 12/21/2022] Open
Abstract
Garcinoic acid (GA or δ-T3-13'COOH), is a natural vitamin E metabolite that has preliminarily been identified as a modulator of nuclear receptors involved in β-amyloid (Aβ) metabolism and progression of Alzheimer's disease (AD). In this study, we investigated GA's effects on Aβ oligomer formation and deposition. Specifically, we compared them with those of other vitamin E analogs and the soy isoflavone genistein, a natural agonist of peroxisome proliferator–activated receptor γ (PPARγ) that has therapeutic potential for managing AD. GA significantly reduced Aβ aggregation and accumulation in mouse cortical astrocytes. Similarly to genistein, GA up-regulated PPARγ expression and apolipoprotein E (ApoE) efflux in these cells with an efficacy that was comparable with that of its metabolic precursor δ-tocotrienol and higher than those of α-tocopherol metabolites. Unlike for genistein and the other vitamin E compounds, the GA-induced restoration of ApoE efflux was not affected by pharmacological inhibition of PPARγ activity, and specific activation of pregnane X receptor (PXR) was observed together with ApoE and multidrug resistance protein 1 (MDR1) membrane transporter up-regulation in both the mouse astrocytes and brain tissue. These effects of GA were associated with reduced Aβ deposition in the brain of TgCRND8 mice, a transgenic AD model. In conclusion, GA holds potential for preventing Aβ oligomerization and deposition in the brain. The mechanistic aspects of GA's properties appear to be distinct from those of other vitamin E metabolites and of genistein.
Collapse
Affiliation(s)
- Rita Marinelli
- Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy
| | - Pierangelo Torquato
- Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy
| | - Desirée Bartolini
- Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy
| | - Cristina Mas-Bargues
- Freshage Research Group, Dept. of Physiology, Faculty of Medicine, University of Valencia, CIBERFES, INCLIVA, Valencia, Spain
| | - Guido Bellezza
- Department of Experimental Medicine, Section of Anatomic Pathology and Histology, Medical School, University of Perugia, Perugia, Italy
| | - Antimo Gioiello
- Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy
| | - Consuelo Borras
- Freshage Research Group, Dept. of Physiology, Faculty of Medicine, University of Valencia, CIBERFES, INCLIVA, Valencia, Spain
| | - Antonella De Luca
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | | | | | - Sridhar Mani
- Departments of Medicine, Genetics and Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York USA
| | - Angelo Sidoni
- Department of Experimental Medicine, Section of Anatomic Pathology and Histology, Medical School, University of Perugia, Perugia, Italy
| | - Jose Viña
- Freshage Research Group, Dept. of Physiology, Faculty of Medicine, University of Valencia, CIBERFES, INCLIVA, Valencia, Spain
| | - Manuela Leri
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Italy.,Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, Italy
| | - Monica Bucciantini
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Italy
| | - Pamela Nardiello
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Italy
| | - Fiorella Casamenti
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Italy
| | - Francesco Galli
- Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy
| |
Collapse
|
10
|
Ichkova A, Rodriguez-Grande B, Zub E, Saudi A, Fournier ML, Aussudre J, Sicard P, Obenaus A, Marchi N, Badaut J. Early cerebrovascular and long-term neurological modifications ensue following juvenile mild traumatic brain injury in male mice. Neurobiol Dis 2020; 141:104952. [PMID: 32442681 DOI: 10.1016/j.nbd.2020.104952] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2020] [Revised: 05/05/2020] [Accepted: 05/17/2020] [Indexed: 12/15/2022] Open
Abstract
Clinical evidence suggests that a mild traumatic brain injury occurring at a juvenile age (jmTBI) may be sufficient to elicit pathophysiological modifications. However, clinical reports are not adequately integrated with experimental studies examining brain changes occurring post-jmTBI. We monitored the cerebrovascular modifications and assessed the long-term behavioral and electrographic changes resulting from experimental jmTBI. In vivo photoacoustic imaging demonstrated a decrease of cerebrovascular oxygen saturation levels in the impacted area hours post-jmTBI. Three days post-jmTBI oxygenation returned to pre-jmTBI levels, stabilizing at 7 and 30 days after the injury. At the functional level, cortical arterioles displayed no NMDA vasodilation response, while vasoconstriction induced by thromboxane receptor agonist was enhanced at 1 day post-jmTBI. Arterioles showed abnormal NMDA vasodilation at 3 days post-jmTBI, returning to normality at 7 days post injury. Histology showed changes in vessel diameters from 1 to 30 days post-jmTBI. Neurological evaluation indicated signs of anxiety-like behavior up to 30 days post-jmTBI. EEG recordings performed at the cortical site of impact 30 days post-jmTBI did not indicate seizures activity, although it revealed a reduction of gamma waves as compared to age matched sham. Histology showed decrease of neuronal filament staining. In conclusion, experimental jmTBI triggers an early cerebrovascular hypo‑oxygenation in vivo and faulty vascular reactivity. The exact topographical coherence and the direct casualty between early cerebrovascular changes and the observed long-term neurological modifications remain to be investigated. A potential translational value for cerebro-vascular oxygen monitoring in jmTBI is discussed.
Collapse
Affiliation(s)
| | | | - Emma Zub
- Cerebrovascular and Glia Research Laboratory, Department of Neuroscience, Institute of Functional Genomics (UMR 5203 CNRS-U1191 INSERM, University of Montpellier), Montpellier, France
| | - Amel Saudi
- Cerebrovascular and Glia Research Laboratory, Department of Neuroscience, Institute of Functional Genomics (UMR 5203 CNRS-U1191 INSERM, University of Montpellier), Montpellier, France
| | | | | | - Pierre Sicard
- INSERM, CNRS, Université de Montpellier, PhyMedExp, IPAM, Montpellier, France
| | - André Obenaus
- CNRS UMR5287, University of Bordeaux, Bordeaux, France; Department of Pediatrics, Loma Linda University School of Medicine, Loma Linda, CA, USA; Basic Science Department, Loma Linda University School of Medicine, Loma Linda, CA, USA; Center for Glial-Neuronal Interactions, Division of Biomedical Sciences, UC Riverside, Riverside, CA, USA; Department of Pediatrics, University of California, Irvine, Irvine, CA, USA
| | - Nicola Marchi
- Cerebrovascular and Glia Research Laboratory, Department of Neuroscience, Institute of Functional Genomics (UMR 5203 CNRS-U1191 INSERM, University of Montpellier), Montpellier, France.
| | - Jerome Badaut
- CNRS UMR5287, University of Bordeaux, Bordeaux, France; Basic Science Department, Loma Linda University School of Medicine, Loma Linda, CA, USA.
| |
Collapse
|
11
|
Torres-Vergara P, Ho YS, Espinoza F, Nualart F, Escudero C, Penny J. The constitutive androstane receptor and pregnane X receptor in the brain. Br J Pharmacol 2020; 177:2666-2682. [PMID: 32201941 DOI: 10.1111/bph.15055] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 03/11/2020] [Accepted: 03/12/2020] [Indexed: 12/16/2022] Open
Abstract
Since their discovery, the orphan nuclear receptors constitutive androstane receptor (CAR;NR1I3) and pregnane X receptor (PXR;NR1I2) have been regarded as master regulators of drug disposition and detoxification mechanisms. They regulate the metabolism and transport of endogenous mediators and xenobiotics in organs including the liver, intestine and brain. However, with proposals of new physiological functions for NR1I3 and NR1I2, there is increasing interest in the role of these receptors in influencing brain function. This review will summarise key findings regarding the expression and function of NR1I3 and NR1I2 in the brain, hereby highlighting the need for further research in this field.
Collapse
Affiliation(s)
- Pablo Torres-Vergara
- Departamento de Farmacia, Facultad de Farmacia, Universidad de Concepción, Concepción, Chile.,Centro de Microscopía Avanzada, CMA-BIO BIO, Laboratorio de Neurobiología y Células Madres NeuroCellT, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile.,Group of Research and Innovation in Vascular Health (GRIVAS Health), Universidad del Bío Bío, Chillán, Chile
| | - Yu Siong Ho
- Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology, Health and Medicine, The University of Manchester, Manchester, UK
| | - Francisca Espinoza
- Centro de Microscopía Avanzada, CMA-BIO BIO, Laboratorio de Neurobiología y Células Madres NeuroCellT, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - Francisco Nualart
- Centro de Microscopía Avanzada, CMA-BIO BIO, Laboratorio de Neurobiología y Células Madres NeuroCellT, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - Carlos Escudero
- Laboratorio de FisiologíaVascular, Departamento de Ciencias Básicas, Facultad de Ciencias Básicas, Universidad del Bío-Bío, Chillán, Chile.,Group of Research and Innovation in Vascular Health (GRIVAS Health), Universidad del Bío Bío, Chillán, Chile
| | - Jeffrey Penny
- Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology, Health and Medicine, The University of Manchester, Manchester, UK
| |
Collapse
|
12
|
Williams S, Hossain M, Ferguson L, Busch RM, Marchi N, Gonzalez-Martinez J, Perucca E, Najm IM, Ghosh C. Neurovascular Drug Biotransformation Machinery in Focal Human Epilepsies: Brain CYP3A4 Correlates with Seizure Frequency and Antiepileptic Drug Therapy. Mol Neurobiol 2019; 56:8392-8407. [PMID: 31243719 DOI: 10.1007/s12035-019-01673-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Accepted: 06/07/2019] [Indexed: 01/01/2023]
Abstract
Pharmacoresistance is a major clinical challenge for approximately 30% of patients with epilepsy. Previous studies indicate nuclear receptors (NRs), drug efflux transporters, and cytochrome P450 enzymes (CYPs) control drug passage across the blood-brain barrier (BBB) in drug-resistant epilepsy. Here, we (1) evaluate BBB changes, neurovascular nuclear receptors, and drug transporters in lesional/epileptic (EPI) and non-lesional/non-epileptic (NON-EPI) regions of the same brain, (2) examine regional CYP expression and activity, and (3) investigate the association among CYP brain expression, seizure frequency, duration of epilepsy, and antiepileptic drug (AED) combination. We used surgically resected brain specimens from patients with medically intractable epilepsy (n = 22) where the epileptogenic loci were well-characterized by invasive and non-invasive methods; histology confirmed distinction of small NON-EPI regions from EPI tissues. NRs, transporters, CYPs, and tight-junction proteins were assessed by western blots/immunohistochemistry, and CYP metabolic activity was determined and compared. The relationship of CYP expression with seizure frequency, duration of epilepsy, and prescribed AEDs was evaluated. Decreased BBB tight-junction proteins accompanied IgG leakage in EPI regions and correlated with upregulated NR and efflux transporter levels. CYP expression and activity significantly increased in EPI compared to NON-EPI tissues. Change in EPI and NON-EPI CYP3A4 expression increased in patients taking AEDs that were CYP substrates, was downregulated when CYP- and non-CYP-substrate AEDs were given together, and correlated with seizure frequency. Our studies suggest focal neurovascular CYP-NR-transporter alterations, as demonstrated by the relationship of seizure frequency and AED combination to brain CYP3A4, might together impact biotransformation machinery of human pharmacoresistant epilepsy.
Collapse
Affiliation(s)
- Sherice Williams
- Cerebrovascular Research Laboratory, Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH, 44195, USA
| | - Mohammed Hossain
- Cerebrovascular Research Laboratory, Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH, 44195, USA
| | - Lisa Ferguson
- Epilepsy Center, Neurological Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Robyn M Busch
- Epilepsy Center, Neurological Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Nicola Marchi
- Cerebrovascular Mechanisms of Brain Disorders Laboratory, Department of Neuroscience, Institute of Functional Genomics (CNRS-INSERM), University of Montpellier, Montpellier, France
| | | | - Emilio Perucca
- Department of Internal Medicine and Therapeutics, University of Pavia, Clinical Trial Center, IRCCS Mondino Foundation, Pavia, Italy
| | - Imad M Najm
- Epilepsy Center, Neurological Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Chaitali Ghosh
- Cerebrovascular Research Laboratory, Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH, 44195, USA. .,Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland Clinic, Cleveland, OH, USA.
| |
Collapse
|
13
|
Duszka K, Wahli W. Enteric Microbiota⁻Gut⁻Brain Axis from the Perspective of Nuclear Receptors. Int J Mol Sci 2018; 19:ijms19082210. [PMID: 30060580 PMCID: PMC6121494 DOI: 10.3390/ijms19082210] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Revised: 07/18/2018] [Accepted: 07/23/2018] [Indexed: 12/12/2022] Open
Abstract
Nuclear receptors (NRs) play a key role in regulating virtually all body functions, thus maintaining a healthy operating body with all its complex systems. Recently, gut microbiota emerged as major factor contributing to the health of the whole organism. Enteric bacteria have multiple ways to influence their host and several of them involve communication with the brain. Mounting evidence of cooperation between gut flora and NRs is already available. However, the full potential of the microbiota interconnection with NRs remains to be uncovered. Herewith, we present the current state of knowledge on the multifaceted roles of NRs in the enteric microbiota–gut–brain axis.
Collapse
Affiliation(s)
- Kalina Duszka
- Department of Nutritional Sciences, University of Vienna, Althanstrasse 14, 1090 Vienna, Austria.
| | - Walter Wahli
- Lee Kong Chian School of Medicine, Nanyang Technological, 11 Mandalay Road, Singapore 308232, Singapore.
- Center for Integrative Genomics, University of Lausanne, Génopode, CH-1015 Lausanne, Switzerland.
| |
Collapse
|
14
|
Editorial on the Special Issue: Molecules and Cognition. Neuroscience 2017; 370:1-3. [PMID: 29113927 DOI: 10.1016/j.neuroscience.2017.11.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|