1
|
Pérez-Osorio IN, Espinosa-Cerón JA, Álvarez-Gutiérrez C, Gonzalez-Flores R, Besedovsky H, Fragoso G, Torres-Ramos MA, Sciutto E. Combined Use of Intranasal Methylprednisolone and Allopregnanolone: Revisiting Anti-inflammatory and Remyelinating Treatment in a Murine Model of Multiple Sclerosis. FRONT BIOSCI-LANDMRK 2024; 29:420. [PMID: 39735995 DOI: 10.31083/j.fbl2912420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 10/30/2024] [Accepted: 11/06/2024] [Indexed: 12/31/2024]
Abstract
BACKGROUND Multiple sclerosis (MS) is a demyelinating, neuroinflammatory, progressive disease that severely affects human health of young adults. Neuroinflammation (NI) and demyelination, as well as their interactions, are key therapeutic targets to halt or slow disease progression. Potent steroidal anti-inflammatory drugs such as methylprednisolone (MP) and remyelinating neurosteroids such as allopregnanolone (ALLO) could be co-administered intranasally to enhance their efficacy by providing direct access to the central nervous system (CNS). METHODS The individual and combined effects of MP and ALLO to control the clinical score of murine experimental autoimmune encephalitis (EAE), to preserve spinal cord tissue integrity, modulate cellular infiltration and gliosis, promote remyelination, and modify the expression of Aryl hydrocarbon receptor (AhR) were evaluated. In silico studies, to deep insight into the mechanisms involved for the treatments, were also conducted. RESULTS MP was the only treatment that significantly reduced the EAE severity, infiltration of inflammatory cells and ionized calcium-binding adapter molecule 1 (Iba-1) expression respect to those EAE non-treated mice but with no-significant differences between the three treatments. MP, ALLO and MP+ALLO significantly reduced tissue damage, AhR expression, and promoted remyelination. Overall, these results suggest that MP, with or without the co-administration with ALLO is an effective and safe strategy to reduce the inflammatory status and the progression of EAE. Despite the expectations of the use of ALLO to reduce the inflammation in EAE, its effect in the dose-scheme used herein is limited only to improve myelination, an effect that supports its usefulness in demyelinating diseases. These results indicate the interest in exploring different doses of ALLO to recommend its use. CONCLUSIONS ALLO treatment mainly maintain the integrity of the spinal cord tissue and the presence of myelin without affecting NI and the clinical outcome. AhR could be involved in the effect observed in both, MP and ALLO treatments. These results will help in the development of a more efficient therapy for MS patients.
Collapse
Affiliation(s)
- Iván Nicolás Pérez-Osorio
- Department of Immunology, Institute of Biomedical Research Universidad Nacional Autónoma de México, UNAM, 04510 Mexico City, Mexico
| | - José Alejandro Espinosa-Cerón
- Department of Immunology, Institute of Biomedical Research Universidad Nacional Autónoma de México, UNAM, 04510 Mexico City, Mexico
| | - Camila Álvarez-Gutiérrez
- Department of Immunology, Institute of Biomedical Research Universidad Nacional Autónoma de México, UNAM, 04510 Mexico City, Mexico
| | - Rodrigo Gonzalez-Flores
- Department of Immunology, Institute of Biomedical Research Universidad Nacional Autónoma de México, UNAM, 04510 Mexico City, Mexico
| | - Hugo Besedovsky
- Research Group Immunophysiology, Division of Neurophysiology, Institute of Physiology and Pathophysiology, Philipps Universität, 35037 Marburg, Germany
| | - Gladis Fragoso
- Department of Immunology, Institute of Biomedical Research Universidad Nacional Autónoma de México, UNAM, 04510 Mexico City, Mexico
| | - Mónica A Torres-Ramos
- Research Directorate, National Institute of Neurology and Neurosurgery Manuel Velasco Suárez, Tlalpan, 14269 Mexico City, Mexico
- Laboratory 4 Translational Sciences, Center for Research on Aging, CINVESTAV South Headquarters, 14330, Mexico City, Mexico
| | - Edda Sciutto
- Department of Immunology, Institute of Biomedical Research Universidad Nacional Autónoma de México, UNAM, 04510 Mexico City, Mexico
| |
Collapse
|
2
|
Zhang S, Xu W, Liu S, Xu F, Chen X, Qin H, Yao K. Anesthetic effects on electrophysiological responses across the visual pathway. Sci Rep 2024; 14:27825. [PMID: 39537872 PMCID: PMC11561267 DOI: 10.1038/s41598-024-79240-2] [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: 07/01/2024] [Accepted: 11/07/2024] [Indexed: 11/16/2024] Open
Abstract
Anesthetics are widely used in electrophysiological tests to assess retinal and visual system functions to avoid experimental errors caused by movement and stress in experimental animals. To determine the most suitable anesthetic for visual electrophysiological tests, excluding ketamine and chloral hydrate due to regulatory and side effect concerns, this study investigated the effects of ethyl carbamate (EC), avertin (AR), and pentobarbital sodium (PS) on visual signal conduction in the retina and primary visual cortex. Assessments included flash electroretinogram (FERG), pattern electroretinogram (PERG), pattern visual evoked potentials (PVEP), and flash visual evoked potentials (FVEP), FERG and FVEP were used to evaluate the responses of the retina and visual cortex to flash stimuli, respectively, while PERG and PVEP assessed responses to pattern stimuli. The research showed that AR demonstrates the least disruption to the visual signal pathway, as evidenced by consistently high characteristic peaks in the AR group across various tests. In contrast, mice given EC exhibited the lowest peak values in both FERG and FVEP, while subjects anesthetized with PS showed suppressed oscillatory potentials and PERG responses. Notably, substantial PVEP characteristic peaks were observed only in mice anesthetized with AR. Consequently, among the three anesthetics tested, AR is the most suitable for visual electrophysiological studies.
Collapse
Affiliation(s)
- Shiyao Zhang
- Institute of Visual Neuroscience and Stem Cell Engineering, Wuhan University of Science and Technology, Wuhan, 430065, China
- College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan, 430065, China
| | - Weihui Xu
- Institute of Visual Neuroscience and Stem Cell Engineering, Wuhan University of Science and Technology, Wuhan, 430065, China
- College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan, 430065, China
| | - Shanshan Liu
- Institute of Visual Neuroscience and Stem Cell Engineering, Wuhan University of Science and Technology, Wuhan, 430065, China
- College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan, 430065, China
| | - Fang Xu
- Institute of Visual Neuroscience and Stem Cell Engineering, Wuhan University of Science and Technology, Wuhan, 430065, China
- College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan, 430065, China
| | - Xiaopeng Chen
- Institute of Visual Neuroscience and Stem Cell Engineering, Wuhan University of Science and Technology, Wuhan, 430065, China
- College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan, 430065, China
| | - Huan Qin
- Institute of Visual Neuroscience and Stem Cell Engineering, Wuhan University of Science and Technology, Wuhan, 430065, China.
- College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan, 430065, China.
| | - Kai Yao
- Institute of Visual Neuroscience and Stem Cell Engineering, Wuhan University of Science and Technology, Wuhan, 430065, China.
- College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan, 430065, China.
| |
Collapse
|
3
|
Jank L, Singh SS, Lee J, Dhukhwa A, Siavoshi F, Joshi D, Minney V, Gupta K, Ghimire S, Deme P, Schoeps VA, Soman K, Ladakis D, Smith M, Borkowski K, Newman J, Baranzini SE, Waubant EL, Fitzgerald KC, Mangalam A, Haughey N, Kornberg M, Chamling X, Calabresi PA, Bhargava P. Restoring the Multiple Sclerosis Associated Imbalance of Gut Indole Metabolites Promotes Remyelination and Suppresses Neuroinflammation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.27.620437. [PMID: 39554063 PMCID: PMC11565924 DOI: 10.1101/2024.10.27.620437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
In multiple sclerosis (MS) the circulating metabolome is dysregulated, with indole lactate (ILA) being one of the most significantly reduced metabolites. We demonstrate that oral supplementation of ILA impacts key MS disease processes in two preclinical models. ILA reduces neuroinflammation by dampening immune cell activation/ infiltration; and promotes remyelination and in vitro oligodendrocyte differentiation through the aryl hydrocarbon receptor (AhR). Supplementation of ILA, a reductive indole metabolite, restores the gut microbiome's oxidative/reductive metabolic balance by lowering circulating indole acetate (IAA), an oxidative indole metabolite, that blocks remyelination and oligodendrocyte maturation. The ILA-induced reduction in circulating IAA is linked to changes in IAA-producing gut microbiota taxa and pathways that are also dysregulated in MS. Notably, a lower ILA:IAA ratio correlates with worse MS outcomes. Overall, these findings identify ILA as a new potential anti-inflammatory remyelinating agent and provide novel insights into the role of gut dysbiosis-related metabolic alterations in MS progression.
Collapse
|
4
|
Halawani D, Wang Y, Estill M, Sefiani A, Ramakrishnan A, Li J, Ni H, Halperin D, Shen L, Geoffroy CG, Friedel RH, Zou H. Aryl hydrocarbon receptor restricts axon regeneration of DRG neurons in response to injury. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.11.04.565649. [PMID: 37961567 PMCID: PMC10635160 DOI: 10.1101/2023.11.04.565649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Injured neurons sense environmental cues to balance neural protection and axon regeneration, but the mechanisms are unclear. Here, we unveil aryl hydrocarbon receptor (AhR), a ligand-activated bHLH-PAS transcription factor, as a molecular sensor and key regulator of acute stress response at the expense of axon regeneration. We demonstrate responsiveness of DRG sensory neurons to AhR signaling, which functions to inhibit axon regeneration. Conditional Ahr deletion in neurons accelerates axon regeneration after sciatic nerve injury. Ahr deletion partially mimics the conditioning lesion in priming DRG to initiate axonogenesis gene programs; upon peripheral axotomy, Ahr ablation suppresses inflammation and stress signaling while augmenting pro-growth pathways. Moreover, comparative transcriptomics revealed signaling interactions between AhR and HIF-1α, two structurally related bHLH-PAS α units that share the dimerization partner Arnt/HIF-1β. Functional assays showed that the growth advantage of AhR-deficient DRG neurons requires HIF-1α; but in the absence of Arnt, DRG neurons can still mount a regenerative response. We further unveil a link between bHLH-PAS transcription factors and DNA hydroxymethylation in response to peripheral axotomy, while RNA-seq of DRG neurons and neuronal single cell RNA-seq analysis revealed a link of AhR regulon to RNA regulation and integrated stress response (ISR). Altogether, AhR activation favors stress coping and inflammation at the expense of axon regeneration; targeting AhR has the potential to enhance nerve repair.
Collapse
Affiliation(s)
- Dalia Halawani
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Yiqun Wang
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, USA
- Sport Medicine Center, Honghui Hospital, Xi’an Jiaotong University, Xi’an, China
| | - Molly Estill
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Arthur Sefiani
- Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University, USA
| | - Aarthi Ramakrishnan
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Jiaxi Li
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, USA
- Department of Neurosurgery, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Haofei Ni
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, USA
- Division of Spine, Department of Orthopedics, Tongji Hospital, Tongji University School of Medicine, Shanghai, China
| | - Daniel Halperin
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Li Shen
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Cédric G. Geoffroy
- Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University, USA
| | - Roland H. Friedel
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, USA
- Department of Neurosurgery, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Hongyan Zou
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, USA
- Department of Neurosurgery, Icahn School of Medicine at Mount Sinai, New York, USA
| |
Collapse
|
5
|
Lee B, Lee SM, Song JW, Choi JW. Gut Microbiota Metabolite Messengers in Brain Function and Pathology at a View of Cell Type-Based Receptor and Enzyme Reaction. Biomol Ther (Seoul) 2024; 32:403-423. [PMID: 38898687 PMCID: PMC11214962 DOI: 10.4062/biomolther.2024.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 05/02/2024] [Accepted: 05/25/2024] [Indexed: 06/21/2024] Open
Abstract
The human gastrointestinal (GI) tract houses a diverse microbial community, known as the gut microbiome comprising bacteria, viruses, fungi, and protozoa. The gut microbiome plays a crucial role in maintaining the body's equilibrium and has recently been discovered to influence the functioning of the central nervous system (CNS). The communication between the nervous system and the GI tract occurs through a two-way network called the gut-brain axis. The nervous system and the GI tract can modulate each other through activated neuronal cells, the immune system, and metabolites produced by the gut microbiome. Extensive research both in preclinical and clinical realms, has highlighted the complex relationship between the gut and diseases associated with the CNS, such as Alzheimer's disease, Parkinson's disease, and amyotrophic lateral sclerosis. This review aims to delineate receptor and target enzymes linked with gut microbiota metabolites and explore their specific roles within the brain, particularly their impact on CNS-related diseases.
Collapse
Affiliation(s)
- Bada Lee
- Department of Biomedicinal and Pharmaceutical Sciences, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Soo Min Lee
- Department of Biomedicinal and Pharmaceutical Sciences, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Jae Won Song
- Department of Regulatory Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Jin Woo Choi
- Department of Biomedicinal and Pharmaceutical Sciences, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
- Department of Regulatory Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| |
Collapse
|
6
|
Veroni C, Olla S, Brignone MS, Siguri C, Formato A, Marra M, Manzoli R, Macario MC, Ambrosini E, Moro E, Agresti C. The Antioxidant Drug Edaravone Binds to the Aryl Hydrocarbon Receptor (AHR) and Promotes the Downstream Signaling Pathway Activation. Biomolecules 2024; 14:443. [PMID: 38672460 PMCID: PMC11047889 DOI: 10.3390/biom14040443] [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: 01/17/2024] [Revised: 03/27/2024] [Accepted: 03/29/2024] [Indexed: 04/28/2024] Open
Abstract
A considerable effort has been spent in the past decades to develop targeted therapies for the treatment of demyelinating diseases, such as multiple sclerosis (MS). Among drugs with free radical scavenging activity and oligodendrocyte protecting effects, Edaravone (Radicava) has recently received increasing attention because of being able to enhance remyelination in experimental in vitro and in vivo disease models. While its beneficial effects are greatly supported by experimental evidence, there is a current paucity of information regarding its mechanism of action and main molecular targets. By using high-throughput RNA-seq and biochemical experiments in murine oligodendrocyte progenitors and SH-SY5Y neuroblastoma cells combined with molecular docking and molecular dynamics simulation, we here provide evidence that Edaravone triggers the activation of aryl hydrocarbon receptor (AHR) signaling by eliciting AHR nuclear translocation and the transcriptional-mediated induction of key cytoprotective gene expression. We also show that an Edaravone-dependent AHR signaling transduction occurs in the zebrafish experimental model, associated with a downstream upregulation of the NRF2 signaling pathway. We finally demonstrate that its rapid cytoprotective and antioxidant actions boost increased expression of the promyelinating Olig2 protein as well as of an Olig2:GFP transgene in vivo. We therefore shed light on a still undescribed potential mechanism of action for this drug, providing further support to its therapeutic potential in the context of debilitating demyelinating conditions.
Collapse
Affiliation(s)
- Caterina Veroni
- Department of Neuroscience, Istituto Superiore di Sanità, 00161 Rome, Italy; (C.V.); (M.S.B.); (E.A.)
| | - Stefania Olla
- Institute for Genetic and Biomedical Research (IRGB), The National Research Council (CNR), Monserrato, 09042 Cagliari, Italy; (S.O.); (C.S.)
| | - Maria Stefania Brignone
- Department of Neuroscience, Istituto Superiore di Sanità, 00161 Rome, Italy; (C.V.); (M.S.B.); (E.A.)
| | - Chiara Siguri
- Institute for Genetic and Biomedical Research (IRGB), The National Research Council (CNR), Monserrato, 09042 Cagliari, Italy; (S.O.); (C.S.)
| | - Alessia Formato
- Institute of Biochemistry and Cell Biology, IBBC-CNR, Campus Adriano Buzzati Traverso, Monterotondo Scalo, 00015 Rome, Italy;
| | - Manuela Marra
- Core Facilities Technical-Scientific Service, Istituto Superiore di Sanità, 00161 Rome, Italy;
| | - Rosa Manzoli
- Department of Molecular Medicine, University of Padova, 35121 Padova, Italy; (R.M.); (M.C.M.)
| | - Maria Carla Macario
- Department of Molecular Medicine, University of Padova, 35121 Padova, Italy; (R.M.); (M.C.M.)
- Department of Biology, University of Padova, 35121 Padova, Italy
| | - Elena Ambrosini
- Department of Neuroscience, Istituto Superiore di Sanità, 00161 Rome, Italy; (C.V.); (M.S.B.); (E.A.)
| | - Enrico Moro
- Department of Molecular Medicine, University of Padova, 35121 Padova, Italy; (R.M.); (M.C.M.)
| | - Cristina Agresti
- Department of Neuroscience, Istituto Superiore di Sanità, 00161 Rome, Italy; (C.V.); (M.S.B.); (E.A.)
| |
Collapse
|
7
|
Tomkiewicz C, Coumoul X, Nioche P, Barouki R, Blanc EB. Costs of molecular adaptation to the chemical exposome: a focus on xenobiotic metabolism pathways. Philos Trans R Soc Lond B Biol Sci 2024; 379:20220510. [PMID: 38310928 PMCID: PMC10838638 DOI: 10.1098/rstb.2022.0510] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Accepted: 12/04/2023] [Indexed: 02/06/2024] Open
Abstract
Organisms adapt to their environment through different pathways. In vertebrates, xenobiotics are detected, metabolized and eliminated through the inducible xenobiotic-metabolizing pathways (XMP) which can also generate reactive toxic intermediates. In this review, we will discuss the impacts of the chemical exposome complexity on the balance between detoxication and side effects. There is a large discrepancy between the limited number of proteins involved in these pathways (few dozens) and the diversity and complexity of the chemical exposome (tens of thousands of chemicals). Several XMP proteins have a low specificity which allows them to bind and/or metabolize a large number of chemicals. This leads to undesired consequences, such as cross-inhibition, inefficient metabolism, release of toxic intermediates, etc. Furthermore, several XMP proteins have endogenous functions that may be disrupted upon exposure to exogenous chemicals. The gut microbiome produces a very large number of metabolites that enter the body and are part of the chemical exposome. It can metabolize xenobiotics and either eliminate them or lead to toxic derivatives. The complex interactions between chemicals of different origins will be illustrated by the diverse roles of the aryl hydrocarbon receptor which binds and transduces the signals of a large number of xenobiotics, microbiome metabolites, dietary chemicals and endogenous compounds. This article is part of the theme issue 'Endocrine responses to environmental variation: conceptual approaches and recent developments'.
Collapse
Affiliation(s)
| | - Xavier Coumoul
- Université Paris Cité, Inserm unit UMRS 1124, 75006 Paris, France
| | - Pierre Nioche
- Université Paris Cité, Inserm unit UMRS 1124, 75006 Paris, France
| | - Robert Barouki
- Université Paris Cité, Inserm unit UMRS 1124, 75006 Paris, France
- Hôpital Necker Enfants malades, AP-HP, 75006 Paris, France
| | - Etienne B. Blanc
- Université Paris Cité, Inserm unit UMRS 1124, 75006 Paris, France
| |
Collapse
|
8
|
Shadboorestan A, Koual M, Dairou J, Coumoul X. The Role of the Kynurenine/AhR Pathway in Diseases Related to Metabolism and Cancer. Int J Tryptophan Res 2023; 16:11786469231185102. [PMID: 37719171 PMCID: PMC10503295 DOI: 10.1177/11786469231185102] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 06/12/2023] [Indexed: 09/19/2023] Open
Abstract
The Aryl hydrocarbon receptor (AhR) is a xenobiotic and endobiotic receptor, which regulates many cellular processes from contaminant metabolism to immunomodulation. Consequently, it is also involved in pathophysiological pathways and now represents a potential therapeutical target. In this review, we will highlight the ancestral function of the protein together with an illustration of its ligand's battery, emphasizing the different responses triggered by these high diverse molecules. Among them, several members of the kynurenine pathway (one key process of tryptophan catabolism) are AhR agonists and are subsequently involved in regulatory functions. We will finally display the interplay between Tryptophan (Trp) catabolism and dysregulation in metabolic pathways drawing hypothesis on the involvement of the AhR pathway in these cancer-related processes.
Collapse
Affiliation(s)
- Amir Shadboorestan
- Department of Toxicology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Meriem Koual
- T3S, INSERM UMR-S 1124, Université Paris Cité, Paris, France
- Assistance Publique-Hôpitaux de Paris, European Hospital Georges-Pompidou, Gynecologic and Breast Oncologic Surgery Department, Paris, France
| | - Julien Dairou
- CNRS, Laboratoire de Chimie et de Biochimie Pharmacologiques et Toxicologiques, Université Paris Cité, Paris, France
| | - Xavier Coumoul
- T3S, INSERM UMR-S 1124, Université Paris Cité, Paris, France
| |
Collapse
|
9
|
Kuang N, Liu Z, Yu G, Wu X, Becker B, Fan H, Peng S, Zhang K, Zhao J, Kang J, Dong G, Zhao X, Sahakian BJ, Robbins TW, Cheng W, Feng J, Schumann G, Palaniyappan L, Zhang J. Neurodevelopmental risk and adaptation as a model for comorbidity among internalizing and externalizing disorders: genomics and cell-specific expression enriched morphometric study. BMC Med 2023; 21:291. [PMID: 37542243 PMCID: PMC10403847 DOI: 10.1186/s12916-023-02920-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 06/01/2023] [Indexed: 08/06/2023] Open
Abstract
BACKGROUND Comorbidity is the rule rather than the exception for childhood and adolescent onset mental disorders, but we cannot predict its occurrence and do not know the neural mechanisms underlying comorbidity. We investigate if the effects of comorbid internalizing and externalizing disorders on anatomical differences represent a simple aggregate of the effects on each disorder and if these comorbidity-associated cortical surface differences relate to a distinct genetic underpinning. METHODS We studied the cortical surface area (SA) and thickness (CT) of 11,878 preadolescents (9-10 years) from the Adolescent Brain and Cognitive Development Study. Linear mixed models were implemented in comparative and association analyses among internalizing (dysthymia, major depressive disorder, disruptive mood dysregulation disorder, agoraphobia, panic disorder, specific phobia, separation anxiety disorder, social anxiety disorder, generalized anxiety disorder, post-traumatic stress disorder), externalizing (attention-deficit/hyperactivity disorder, oppositional defiant disorder, conduct disorder) diagnostic groups, a group with comorbidity of the two and a healthy control group. Genome-wide association analysis (GWAS) and cell type specificity analysis were performed on 4468 unrelated European participants from this cohort. RESULTS Smaller cortical surface area but higher thickness was noted across patient groups when compared to controls. Children with comorbid internalizing and externalizing disorders had more pronounced areal reduction than those without comorbidity, indicating an additive burden. In contrast, cortical thickness had a non-linear effect with comorbidity: the comorbid group had no significant CT differences, while those patient groups without comorbidity had significantly higher thickness compare to healthy controls. Distinct biological pathways were implicated in regional SA and CT differences. Specifically, CT differences were associated with immune-related processes implicating astrocytes and oligodendrocytes, while SA-related differences related mainly to inhibitory neurons. CONCLUSION The emergence of comorbidity across distinct clusters of psychopathology is unlikely to be due to a simple additive neurobiological effect alone. Distinct developmental risk moderated by immune-related adaptation processes, with unique genetic and cell-specific factors, may contribute to underlying SA and CT differences. Children with the highest risk but lowest resilience, both captured in their developmental morphometry, may develop a comorbid illness pattern.
Collapse
Affiliation(s)
- Nanyu Kuang
- Institute of Science and Technology for Brain Inspired Intelligence, Fudan University, Shanghai, People's Republic of China
- Key Laboratory of Computational Neuroscience and Brain Inspired Intelligence, Ministry of Education, Fudan University, Beijing, People's Republic of China
| | - Zhaowen Liu
- School of Computer Science, Northwestern Polytechnical University, Xi'an, Shanxin, People's Republic of China
| | - Gechang Yu
- Institute of Science and Technology for Brain Inspired Intelligence, Fudan University, Shanghai, People's Republic of China
- Key Laboratory of Computational Neuroscience and Brain Inspired Intelligence, Ministry of Education, Fudan University, Beijing, People's Republic of China
| | - Xinran Wu
- Institute of Science and Technology for Brain Inspired Intelligence, Fudan University, Shanghai, People's Republic of China
- Key Laboratory of Computational Neuroscience and Brain Inspired Intelligence, Ministry of Education, Fudan University, Beijing, People's Republic of China
| | - Benjamin Becker
- Clinical Hospital of Chengdu Brain Science Institute, MOE Key Laboratory for Neuroinformation, University of Electronic Science and Technology of China, Chengdu, People's Republic of China
| | - Huaxin Fan
- Institute of Science and Technology for Brain Inspired Intelligence, Fudan University, Shanghai, People's Republic of China
- Key Laboratory of Computational Neuroscience and Brain Inspired Intelligence, Ministry of Education, Fudan University, Beijing, People's Republic of China
| | - Songjun Peng
- Institute of Science and Technology for Brain Inspired Intelligence, Fudan University, Shanghai, People's Republic of China
- Key Laboratory of Computational Neuroscience and Brain Inspired Intelligence, Ministry of Education, Fudan University, Beijing, People's Republic of China
| | - Kai Zhang
- Institute of Computer Science and Technology, East China Normal University, Shanghai, People's Republic of China
| | - Jiajia Zhao
- Institute of Science and Technology for Brain Inspired Intelligence, Fudan University, Shanghai, People's Republic of China
- Key Laboratory of Computational Neuroscience and Brain Inspired Intelligence, Ministry of Education, Fudan University, Beijing, People's Republic of China
| | - Jujiao Kang
- Institute of Science and Technology for Brain Inspired Intelligence, Fudan University, Shanghai, People's Republic of China
- Shanghai Center for Mathematical Sciences, Fudan University, Shanghai, People's Republic of China
| | - Guiying Dong
- Institute of Science and Technology for Brain Inspired Intelligence, Fudan University, Shanghai, People's Republic of China
- MOE Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, People's Republic of China
| | - Xingming Zhao
- Institute of Science and Technology for Brain Inspired Intelligence, Fudan University, Shanghai, People's Republic of China
- MOE Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, People's Republic of China
- Zhangjiang Fudan International Innovation Center, Shanghai, 200433, People's Republic of China
| | - Barbara J Sahakian
- Institute of Science and Technology for Brain Inspired Intelligence, Fudan University, Shanghai, People's Republic of China
- Department of Psychiatry, School of Clinical Medicine, University of Cambridge, Cambridge, UK
| | - Trevor W Robbins
- Institute of Science and Technology for Brain Inspired Intelligence, Fudan University, Shanghai, People's Republic of China
- Department of Psychology, Behavioural and Clinical Neuroscience Institute, University of Cambridge, Cambridge, UK
| | - Wei Cheng
- Institute of Science and Technology for Brain Inspired Intelligence, Fudan University, Shanghai, People's Republic of China
- Key Laboratory of Computational Neuroscience and Brain Inspired Intelligence, Ministry of Education, Fudan University, Beijing, People's Republic of China
- Fudan ISTBI-ZJNU Algorithm Centre for Brain-inspired Intelligence, Zhejiang Normal University, Jinhua, 321004, China
- Shanghai Medical College and Zhongshan Hospital Immunotherapy Technology Transfer Center, Shanghai, 200032, China
| | - Jianfeng Feng
- Institute of Science and Technology for Brain Inspired Intelligence, Fudan University, Shanghai, People's Republic of China
- Key Laboratory of Computational Neuroscience and Brain Inspired Intelligence, Ministry of Education, Fudan University, Beijing, People's Republic of China
- Shanghai Center for Mathematical Sciences, Shanghai, 200433, People's Republic of China
- Department of Computer Science, University of Warwick, Coventry, CV4 7AL, UK
- Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, 200433, People's Republic of China
- Fudan ISTBI-ZJNU Algorithm Centre for Brain-inspired Intelligence, Zhejiang Normal University, Jinhua, People's Republic of China
| | - Gunter Schumann
- Institute of Science and Technology for Brain Inspired Intelligence, Fudan University, Shanghai, People's Republic of China.
- PONS Research Group, Department of Psychiatry and 20 Psychotherapy, Humboldt University, Berlin and Leibniz Institute for Neurobiology, Campus Charite Mitte, Magdeburg, Germany.
| | - Lena Palaniyappan
- Douglas Mental Health University Institute, Department of Psychiatry, McGill University, Montreal, QC, Canada.
- Department of Psychiatry, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada.
- Robarts Research Institute, University of Western Ontario, London, ON, Canada.
- Department of Medical Biophysica, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada.
| | - Jie Zhang
- Institute of Science and Technology for Brain Inspired Intelligence, Fudan University, Shanghai, People's Republic of China.
- Key Laboratory of Computational Neuroscience and Brain Inspired Intelligence, Ministry of Education, Fudan University, Beijing, People's Republic of China.
| |
Collapse
|
10
|
Elson D, Nguyen BD, Bernales S, Chakravarty S, Jang HS, Korjeff NA, Zhang Y, Wilferd SF, Castro DJ, Plaisier CL, Finlay D, Oshima RG, Kolluri SK. Induction of Aryl Hydrocarbon Receptor-Mediated Cancer Cell-Selective Apoptosis in Triple-Negative Breast Cancer Cells by a High-Affinity Benzimidazoisoquinoline. ACS Pharmacol Transl Sci 2023; 6:1028-1042. [PMID: 37470014 PMCID: PMC10353065 DOI: 10.1021/acsptsci.2c00253] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Indexed: 07/21/2023]
Abstract
Triple-negative breast cancer (TNBC) remains a disease with a paucity of targeted treatment opportunities. The aryl hydrocarbon receptor (AhR) is a ligand-activated transcription factor that is involved in a wide range of physiological processes, including the sensing of xenobiotics, immune function, development, and differentiation. Different small-molecule AhR ligands drive strikingly varied cellular and organismal responses. In certain cancers, AhR activation by select small molecules induces cell cycle arrest or apoptosis via activation of tumor-suppressive transcriptional programs. AhR is expressed in triple-negative breast cancers, presenting a tractable therapeutic opportunity. Here, we identify a novel ligand of the aryl hydrocarbon receptor that potently and selectively induces cell death in triple-negative breast cancer cells and TNBC stem cells via the AhR. Importantly, we found that this compound, Analog 523, exhibits minimal cytotoxicity against multiple normal human primary cells. Analog 523 represents a high-affinity AhR ligand with potential for future clinical translation as an anticancer agent.
Collapse
Affiliation(s)
- Daniel
J. Elson
- Cancer
Research Laboratory, Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, Oregon, 97331, United States
| | - Bach D. Nguyen
- Cancer
Research Laboratory, Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, Oregon, 97331, United States
| | - Sebastian Bernales
- Praxis
Biotech, San Francisco, California, 94158, United States
- Centro Ciencia
& Vida, Avda. Del
Valle Norte 725, Santiago, 8580702, Chile
| | | | - Hyo Sang Jang
- Cancer
Research Laboratory, Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, Oregon, 97331, United States
| | - Nicholas A. Korjeff
- Cancer
Research Laboratory, Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, Oregon, 97331, United States
| | - Yi Zhang
- Cancer
Research Laboratory, Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, Oregon, 97331, United States
| | - Sierra F. Wilferd
- School
of Biological and Health Systems Engineering, Arizona State University, Tempe, Arizona 85287, United States
| | - David J. Castro
- Sanford
Burnham Prebys Medical Discovery Institute, NCI Designated Cancer
Center, La Jolla, California, 92037, United States
- Oregon Health
& Science University, Portland, Oregon, 97239, United States
| | - Christopher L. Plaisier
- School
of Biological and Health Systems Engineering, Arizona State University, Tempe, Arizona 85287, United States
| | - Darren Finlay
- Sanford
Burnham Prebys Medical Discovery Institute, NCI Designated Cancer
Center, La Jolla, California, 92037, United States
| | - Robert G. Oshima
- Sanford
Burnham Prebys Medical Discovery Institute, NCI Designated Cancer
Center, La Jolla, California, 92037, United States
| | - Siva K. Kolluri
- Cancer
Research Laboratory, Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, Oregon, 97331, United States
- Linus
Pauling Institute, Oregon State University, Corvallis, Oregon, 97331, United
States
- The
Pacific Northwest Center for Translational Environmental Health Research, Oregon State University, Corvallis, Oregon, 97331, United States
| |
Collapse
|
11
|
Huang YJ, Hung CC, Hsu PC, Lee PY, Tsai YA, Hsin YC, Lee XT, Chou CC, Chen ML, Tarng DC, Lee YH. Astrocytic aryl hydrocarbon receptor mediates chronic kidney disease-associated mental disorders involving GLT1 hypofunction and neuronal activity enhancement in the mouse brain. Glia 2023; 71:1057-1080. [PMID: 36573349 DOI: 10.1002/glia.24326] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 12/09/2022] [Accepted: 12/11/2022] [Indexed: 12/28/2022]
Abstract
Chronic kidney disease (CKD)-associated mental disorders have been attributed to the excessive accumulation of hemodialysis-resistant indoxyl-3-sulfate (I3S) in the brain. I3S not only induces oxidative stress but is also a potent endogenous agonist of the aryl hydrocarbon receptor (AhR). Here, we investigated the role of AhR in CKD-induced brain disorders using a 5/6 nephrectomy-induced CKD mouse model, which showed increased I3S concentration in both blood and brain, anxiety and impaired novelty recognition, and AhR activation in the anterior cortex. GFAP+ reactive astrocytes were increased accompanied with the reduction of glutamate transporter 1 (GLT1) on perineuronal astrocytic processes (PAPs) in the anterior cingulate cortex (ACC) in CKD mice, and these alterations were attenuated in both neural lineage-specific and astrocyte-specific Ahr conditional knockout mice (nAhrCKO and aAhrCKO). By using chronic I3S treatment in primary astrocytes and glia-neuron (GN) mix cultures to mimic the CKD brain microenvironment, we also found significant reduction of GLT1 expression and activity in an AhR-dependent manner. Chronic I3S treatment induced AhR-dependent pro-oxidant Nox1 and AhR-independent anti-oxidant HO-1 expressions. Notably, AhR mediates chronic I3S-induced neuronal activity enhancement and synaptotoxicity in GN mix, not neuron-enriched cortical culture. In CKD mice, neuronal activity enhancement was observed in ACC and hippocampal CA1, and these responses were abrogated by both nAhrCKO and aAhrCKO. Finally, intranasal AhR antagonist CH-223191 administration significantly ameliorated the GLT1/PAPs reduction, increase in c-Fos+ neurons, and memory impairment in the CKD mice. Thus, astrocytic AhR plays a crucial role in the CKD-induced disturbance of neuron-astrocyte interaction and mental disorders.
Collapse
Affiliation(s)
- Yu-Jie Huang
- Department and Institute of Physiology, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Chia-Chi Hung
- Department and Institute of Physiology, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan.,Brain Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Pei-Chien Hsu
- Department and Institute of Physiology, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Po-Yi Lee
- Department and Institute of Physiology, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Yen-An Tsai
- Institute of Environmental and Occupational Health Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Yu-Chiao Hsin
- Department and Institute of Physiology, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Xie-Ting Lee
- Department and Institute of Physiology, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Chia-Cheng Chou
- National Laboratory Animal Center, National Applied Research Laboratories, Taipei, Taiwan
| | - Mei-Lien Chen
- Institute of Environmental and Occupational Health Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Der-Cherng Tarng
- Department and Institute of Physiology, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan.,Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan.,Division of Nephrology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Yi-Hsuan Lee
- Department and Institute of Physiology, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan.,Brain Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan
| |
Collapse
|
12
|
Cross-Regulation of the Cellular Redox System, Oxygen, and Sphingolipid Signalling. Metabolites 2023; 13:metabo13030426. [PMID: 36984866 PMCID: PMC10054022 DOI: 10.3390/metabo13030426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 03/09/2023] [Accepted: 03/10/2023] [Indexed: 03/17/2023] Open
Abstract
Redox-active mediators are now appreciated as powerful molecules to regulate cellular dynamics such as viability, proliferation, migration, cell contraction, and relaxation, as well as gene expression under physiological and pathophysiological conditions. These molecules include the various reactive oxygen species (ROS), and the gasotransmitters nitric oxide (NO∙), carbon monoxide (CO), and hydrogen sulfide (H2S). For each of these molecules, direct targets have been identified which transmit the signal from the cellular redox state to a cellular response. Besides these redox mediators, various sphingolipid species have turned out as highly bioactive with strong signalling potential. Recent data suggest that there is a cross-regulation existing between the redox mediators and sphingolipid molecules that have a fundamental impact on a cell’s fate and organ function. This review will summarize the effects of the different redox-active mediators on sphingolipid signalling and metabolism, and the impact of this cross-talk on pathophysiological processes. The relevance of therapeutic approaches will be highlighted.
Collapse
|
13
|
Martin NR, Patel R, Kossack ME, Tian L, Camarillo MA, Cintrón-Rivera LG, Gawdzik JC, Yue MS, Nwagugo FO, Elemans LMH, Plavicki JS. Proper modulation of AHR signaling is necessary for establishing neural connectivity and oligodendrocyte precursor cell development in the embryonic zebrafish brain. Front Mol Neurosci 2022; 15:1032302. [PMID: 36523606 PMCID: PMC9745199 DOI: 10.3389/fnmol.2022.1032302] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 10/24/2022] [Indexed: 12/03/2022] Open
Abstract
2,3,7,8-tetrachlorodibenzo-[p]-dioxin (TCDD) is a persistent global pollutant that exhibits a high affinity for the aryl hydrocarbon receptor (AHR), a ligand activated transcription factor. Epidemiological studies have associated AHR agonist exposure with multiple human neuropathologies. Consistent with the human data, research studies using laboratory models have linked pollutant-induced AHR activation to disruptions in learning and memory as well as motor impairments. Our understanding of endogenous AHR functions in brain development is limited and, correspondingly, scientists are still determining which cell types and brain regions are sensitive to AHR modulation. To identify novel phenotypes resulting from pollutant-induced AHR activation and ahr2 loss of function, we utilized the optically transparent zebrafish model. Early embryonic TCDD exposure impaired embryonic brain morphogenesis, resulted in ventriculomegaly, and disrupted neural connectivity in the optic tectum, habenula, cerebellum, and olfactory bulb. Altered neural network formation was accompanied by reduced expression of synaptic vesicle 2. Loss of ahr2 function also impaired nascent network development, but did not affect gross brain or ventricular morphology. To determine whether neural AHR activation was sufficient to disrupt connectivity, we used the Gal4/UAS system to express a constitutively active AHR specifically in differentiated neurons and observed disruptions only in the cerebellum; thus, suggesting that the phenotypes resulting from global AHR activation likely involve multiple cell types. Consistent with this hypothesis, we found that TCDD exposure reduced the number of oligodendrocyte precursor cells and their derivatives. Together, our findings indicate that proper modulation of AHR signaling is necessary for the growth and maturation of the embryonic zebrafish brain.
Collapse
Affiliation(s)
- Nathan R. Martin
- Department of Pathology and Laboratory Medicine, Brown University, Providence, RI, United States
| | - Ratna Patel
- Department of Pathology and Laboratory Medicine, Brown University, Providence, RI, United States
| | - Michelle E. Kossack
- Department of Pathology and Laboratory Medicine, Brown University, Providence, RI, United States
| | - Lucy Tian
- Department of Pathology and Laboratory Medicine, Brown University, Providence, RI, United States
| | - Manuel A. Camarillo
- Department of Pathology and Laboratory Medicine, Brown University, Providence, RI, United States
| | - Layra G. Cintrón-Rivera
- Department of Pathology and Laboratory Medicine, Brown University, Providence, RI, United States
| | - Joseph C. Gawdzik
- Molecular and Environmental Toxicology Center, University of Wisconsin at Madison, Madison, WI, United States,Division of Pharmaceutical Sciences, University of Wisconsin at Madison, Madison, WI, United States
| | - Monica S. Yue
- Molecular and Environmental Toxicology Center, University of Wisconsin at Madison, Madison, WI, United States,Division of Pharmaceutical Sciences, University of Wisconsin at Madison, Madison, WI, United States
| | - Favour O. Nwagugo
- Department of Biology, University of Maryland Baltimore County, Baltimore, MD, United States
| | - Loes M. H. Elemans
- Division of Toxicology, Institute for Risk Assessment Sciences (IRAS), Utrecht University, Utrecht, Netherlands
| | - Jessica S. Plavicki
- Department of Pathology and Laboratory Medicine, Brown University, Providence, RI, United States,*Correspondence: Jessica S. Plavicki,
| |
Collapse
|
14
|
Hu J, Baydyuk M, Huang JK. Impact of amino acids on microglial activation and CNS remyelination. Curr Opin Pharmacol 2022; 66:102287. [PMID: 36067684 DOI: 10.1016/j.coph.2022.102287] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 07/20/2022] [Accepted: 08/01/2022] [Indexed: 11/27/2022]
Abstract
Amino acids and their derivatives function as building blocks as well as signaling molecules to modulate various cellular processes in living organisms. In mice, amino acids accumulate in demyelinated lesions and return to basal levels during remyelination. Studies have found that amino acids and their metabolites modulate immune activity in the central nervous system (CNS) and influence oligodendrocyte differentiation and remyelination efficiency. In this review, we discuss current studies on amino acid metabolism in the context of CNS remyelination. By understanding the mechanisms of amino acid signaling and metabolism in demyelinated lesions, we may deepen our understanding of compartmentalized CNS inflammation in demyelinating disease like multiple sclerosis (MS) and provide evidence to develop novel pharmacological therapies targeting amino acid metabolism to prevent disease worsening.
Collapse
Affiliation(s)
- Jingwen Hu
- Department of Biology, Georgetown University, 37th and O St., NW, Washington, DC, 20057, USA
| | - Maryna Baydyuk
- Department of Biology, Georgetown University, 37th and O St., NW, Washington, DC, 20057, USA; Center for Cell Reprogramming, Georgetown University Medical Center, 37th and O St., NW, Washington, DC, 20057, USA
| | - Jeffrey K Huang
- Department of Biology, Georgetown University, 37th and O St., NW, Washington, DC, 20057, USA; Center for Cell Reprogramming, Georgetown University Medical Center, 37th and O St., NW, Washington, DC, 20057, USA.
| |
Collapse
|
15
|
Larigot L, Bui LC, de Bouvier M, Pierre O, Pinon G, Fiocca J, Ozeir M, Tourette C, Ottolenghi C, Imbeaud S, Pontoizeau C, Blaise BJ, Chevallier A, Tomkiewicz C, Legrand B, Elena-Herrmann B, Néri C, Brinkmann V, Nioche P, Barouki R, Ventura N, Dairou J, Coumoul X. Identification of Modulators of the C. elegans Aryl Hydrocarbon Receptor and Characterization of Transcriptomic and Metabolic AhR-1 Profiles. Antioxidants (Basel) 2022; 11:antiox11051030. [PMID: 35624894 PMCID: PMC9137885 DOI: 10.3390/antiox11051030] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 05/06/2022] [Accepted: 05/18/2022] [Indexed: 02/02/2023] Open
Abstract
The Aryl hydrocarbon Receptor (AhR) is a xenobiotic sensor in vertebrates, regulating the metabolism of its own ligands. However, no ligand has been identified to date for any AhR in invertebrates. In C. elegans, the AhR ortholog, AHR-1, displays physiological functions. Therefore, we compared the transcriptomic and metabolic profiles of worms expressing AHR-1 or not and investigated the putative panel of chemical AHR-1 modulators. The metabolomic profiling indicated a role for AHR-1 in amino acids, carbohydrates, and fatty acids metabolism. The transcriptional profiling in neurons expressing AHR-1, identified 95 down-regulated genes and 76 up-regulated genes associated with neuronal and metabolic functions in the nervous system. A gene reporter system allowed us to identify several AHR-1 modulators including bacterial, dietary, or environmental compounds. These results shed new light on the biological functions of AHR-1 in C. elegans and perspectives on the evolution of the AhR functions across species.
Collapse
Affiliation(s)
- Lucie Larigot
- INSERM UMR-S1124, T3S, Toxicologie Environnementale, Cibles Thérapeutiques, Signalisation Cellulaire et Biomarqueurs, Université Paris Cité, 75006 Paris, France; (L.L.); (L.-C.B.); (M.d.B.); (O.P.); (G.P.); (J.F.); (M.O.); (C.O.); (A.C.); (C.T.); (B.L.); (P.N.); (R.B.)
- CNRS UMR 8601, Metabolism, Pharmacochemistry and Neurochemistry, Université Paris Cité, 75006 Paris, France
| | - Linh-Chi Bui
- INSERM UMR-S1124, T3S, Toxicologie Environnementale, Cibles Thérapeutiques, Signalisation Cellulaire et Biomarqueurs, Université Paris Cité, 75006 Paris, France; (L.L.); (L.-C.B.); (M.d.B.); (O.P.); (G.P.); (J.F.); (M.O.); (C.O.); (A.C.); (C.T.); (B.L.); (P.N.); (R.B.)
- Unité de biologie fonctionnelle et adaptative, UMR 8251, CNRS, Université Paris Cité, 75013 Paris, France
| | - Marine de Bouvier
- INSERM UMR-S1124, T3S, Toxicologie Environnementale, Cibles Thérapeutiques, Signalisation Cellulaire et Biomarqueurs, Université Paris Cité, 75006 Paris, France; (L.L.); (L.-C.B.); (M.d.B.); (O.P.); (G.P.); (J.F.); (M.O.); (C.O.); (A.C.); (C.T.); (B.L.); (P.N.); (R.B.)
| | - Ophélie Pierre
- INSERM UMR-S1124, T3S, Toxicologie Environnementale, Cibles Thérapeutiques, Signalisation Cellulaire et Biomarqueurs, Université Paris Cité, 75006 Paris, France; (L.L.); (L.-C.B.); (M.d.B.); (O.P.); (G.P.); (J.F.); (M.O.); (C.O.); (A.C.); (C.T.); (B.L.); (P.N.); (R.B.)
- Laboratoire Interactions Epithéliums-Neurones (LIEN), Université de Brest, EA4685, 29200 Brest, France
| | - Grégory Pinon
- INSERM UMR-S1124, T3S, Toxicologie Environnementale, Cibles Thérapeutiques, Signalisation Cellulaire et Biomarqueurs, Université Paris Cité, 75006 Paris, France; (L.L.); (L.-C.B.); (M.d.B.); (O.P.); (G.P.); (J.F.); (M.O.); (C.O.); (A.C.); (C.T.); (B.L.); (P.N.); (R.B.)
- Structural and Molecular Analysis Platform, Biomedtech Facilities, Université Paris Cité, 75006 Paris, France
| | - Justine Fiocca
- INSERM UMR-S1124, T3S, Toxicologie Environnementale, Cibles Thérapeutiques, Signalisation Cellulaire et Biomarqueurs, Université Paris Cité, 75006 Paris, France; (L.L.); (L.-C.B.); (M.d.B.); (O.P.); (G.P.); (J.F.); (M.O.); (C.O.); (A.C.); (C.T.); (B.L.); (P.N.); (R.B.)
- Structural and Molecular Analysis Platform, Biomedtech Facilities, Université Paris Cité, 75006 Paris, France
| | - Mohammad Ozeir
- INSERM UMR-S1124, T3S, Toxicologie Environnementale, Cibles Thérapeutiques, Signalisation Cellulaire et Biomarqueurs, Université Paris Cité, 75006 Paris, France; (L.L.); (L.-C.B.); (M.d.B.); (O.P.); (G.P.); (J.F.); (M.O.); (C.O.); (A.C.); (C.T.); (B.L.); (P.N.); (R.B.)
- Structural and Molecular Analysis Platform, Biomedtech Facilities, Université Paris Cité, 75006 Paris, France
| | - Cendrine Tourette
- Centre Paul Broca, INSERM U894 Neuronal Cell Biology & Pathology & EA Université Paris Cité, 75014 Paris, France;
| | - Chris Ottolenghi
- INSERM UMR-S1124, T3S, Toxicologie Environnementale, Cibles Thérapeutiques, Signalisation Cellulaire et Biomarqueurs, Université Paris Cité, 75006 Paris, France; (L.L.); (L.-C.B.); (M.d.B.); (O.P.); (G.P.); (J.F.); (M.O.); (C.O.); (A.C.); (C.T.); (B.L.); (P.N.); (R.B.)
- AP-HP, Hôpital Necker-Enfants Malades, Service de Biochimie Métabolique, 75015 Paris, France;
| | - Sandrine Imbeaud
- Gif/Orsay DNA MicroArray Platform, 91190 Gif sur Yvette, France;
| | - Clément Pontoizeau
- AP-HP, Hôpital Necker-Enfants Malades, Service de Biochimie Métabolique, 75015 Paris, France;
- Centre de Résonance Magnétique Nucléaire à Très Hauts Champs, Univ. Lyon, CNRS, UCBL, ENS Lyon, 69100 Villeurbanne, France; (B.J.B.); (B.E.-H.)
| | - Benjamin J. Blaise
- Centre de Résonance Magnétique Nucléaire à Très Hauts Champs, Univ. Lyon, CNRS, UCBL, ENS Lyon, 69100 Villeurbanne, France; (B.J.B.); (B.E.-H.)
| | - Aline Chevallier
- INSERM UMR-S1124, T3S, Toxicologie Environnementale, Cibles Thérapeutiques, Signalisation Cellulaire et Biomarqueurs, Université Paris Cité, 75006 Paris, France; (L.L.); (L.-C.B.); (M.d.B.); (O.P.); (G.P.); (J.F.); (M.O.); (C.O.); (A.C.); (C.T.); (B.L.); (P.N.); (R.B.)
| | - Céline Tomkiewicz
- INSERM UMR-S1124, T3S, Toxicologie Environnementale, Cibles Thérapeutiques, Signalisation Cellulaire et Biomarqueurs, Université Paris Cité, 75006 Paris, France; (L.L.); (L.-C.B.); (M.d.B.); (O.P.); (G.P.); (J.F.); (M.O.); (C.O.); (A.C.); (C.T.); (B.L.); (P.N.); (R.B.)
| | - Béatrice Legrand
- INSERM UMR-S1124, T3S, Toxicologie Environnementale, Cibles Thérapeutiques, Signalisation Cellulaire et Biomarqueurs, Université Paris Cité, 75006 Paris, France; (L.L.); (L.-C.B.); (M.d.B.); (O.P.); (G.P.); (J.F.); (M.O.); (C.O.); (A.C.); (C.T.); (B.L.); (P.N.); (R.B.)
| | - Bénédicte Elena-Herrmann
- Centre de Résonance Magnétique Nucléaire à Très Hauts Champs, Univ. Lyon, CNRS, UCBL, ENS Lyon, 69100 Villeurbanne, France; (B.J.B.); (B.E.-H.)
- Institute for Advanced Biosciences, Univ. Grenoble Alpes, CNRS, INSERM, 38000 Grenoble, France
| | - Christian Néri
- CNRS UMR 8256, Inserm ERL U1164, Sorbonne Université, 75005 Paris, France;
| | - Vanessa Brinkmann
- Institute of Clinical Chemistry and Laboratory Diagnostic, Medical Faculty, Heinrich Heine University, Düsseldorf, Moorenstr 5, 40225 Düsseldorf, Germany; (V.B.); (N.V.)
- Leibniz Institute for Environmental Medicine (IUF), Auf’m Hennekamp 50, 40225 Düsseldorf, Germany
| | - Pierre Nioche
- INSERM UMR-S1124, T3S, Toxicologie Environnementale, Cibles Thérapeutiques, Signalisation Cellulaire et Biomarqueurs, Université Paris Cité, 75006 Paris, France; (L.L.); (L.-C.B.); (M.d.B.); (O.P.); (G.P.); (J.F.); (M.O.); (C.O.); (A.C.); (C.T.); (B.L.); (P.N.); (R.B.)
- Structural and Molecular Analysis Platform, Biomedtech Facilities, Université Paris Cité, 75006 Paris, France
| | - Robert Barouki
- INSERM UMR-S1124, T3S, Toxicologie Environnementale, Cibles Thérapeutiques, Signalisation Cellulaire et Biomarqueurs, Université Paris Cité, 75006 Paris, France; (L.L.); (L.-C.B.); (M.d.B.); (O.P.); (G.P.); (J.F.); (M.O.); (C.O.); (A.C.); (C.T.); (B.L.); (P.N.); (R.B.)
- Assistance Publique-Hôpitaux de Paris, Hôpital Necker, 75015 Paris, France
| | - Natascia Ventura
- Institute of Clinical Chemistry and Laboratory Diagnostic, Medical Faculty, Heinrich Heine University, Düsseldorf, Moorenstr 5, 40225 Düsseldorf, Germany; (V.B.); (N.V.)
- Leibniz Institute for Environmental Medicine (IUF), Auf’m Hennekamp 50, 40225 Düsseldorf, Germany
| | - Julien Dairou
- CNRS UMR 8601, Metabolism, Pharmacochemistry and Neurochemistry, Université Paris Cité, 75006 Paris, France
- Correspondence: (J.D.); (X.C.); Tel.: +33-1-42-86-91-21 (J.D.); +33-1-42-86-33-59 (X.C.)
| | - Xavier Coumoul
- INSERM UMR-S1124, T3S, Toxicologie Environnementale, Cibles Thérapeutiques, Signalisation Cellulaire et Biomarqueurs, Université Paris Cité, 75006 Paris, France; (L.L.); (L.-C.B.); (M.d.B.); (O.P.); (G.P.); (J.F.); (M.O.); (C.O.); (A.C.); (C.T.); (B.L.); (P.N.); (R.B.)
- Correspondence: (J.D.); (X.C.); Tel.: +33-1-42-86-91-21 (J.D.); +33-1-42-86-33-59 (X.C.)
| |
Collapse
|
16
|
CCR1 antagonist ameliorates experimental autoimmune encephalomyelitis by inhibition of Th9/Th22-related markers in the brain and periphery. Mol Immunol 2022; 144:127-137. [DOI: 10.1016/j.molimm.2022.02.017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 01/20/2022] [Accepted: 02/17/2022] [Indexed: 12/19/2022]
|
17
|
Hammond CL, Roztocil E, Gupta V, Feldon SE, Woeller CF. More than Meets the Eye: The Aryl Hydrocarbon Receptor is an Environmental Sensor, Physiological Regulator and a Therapeutic Target in Ocular Disease. FRONTIERS IN TOXICOLOGY 2022; 4:791082. [PMID: 35295218 PMCID: PMC8915869 DOI: 10.3389/ftox.2022.791082] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 02/08/2022] [Indexed: 12/22/2022] Open
Abstract
The aryl hydrocarbon receptor (AHR) is a ligand activated transcription factor originally identified as an environmental sensor of xenobiotic chemicals. However, studies have revealed that the AHR regulates crucial aspects of cell growth and metabolism, development and the immune system. The importance of the AHR and AHR signaling in eye development, toxicology and disease is now being uncovered. The AHR is expressed in many ocular tissues including the retina, choroid, cornea and the orbit. A significant role for the AHR in age-related macular degeneration (AMD), autoimmune uveitis, and other ocular diseases has been identified. Ligands for the AHR are structurally diverse organic molecules from exogenous and endogenous sources. Natural AHR ligands include metabolites of tryptophan and byproducts of the microbiome. Xenobiotic AHR ligands include persistent environmental pollutants such as dioxins, benzo (a) pyrene [B (a) P] and polychlorinated biphenyls (PCBs). Pharmaceutical agents including the proton pump inhibitors, esomeprazole and lansoprazole, and the immunosuppressive drug, leflunomide, activate the AHR. In this review, we highlight the role of the AHR in the eye and discuss how AHR signaling is involved in responding to endogenous and environmental stimuli. We also present the emerging concept that the AHR is a promising therapeutic target for eye disease.
Collapse
Affiliation(s)
| | | | | | | | - Collynn F. Woeller
- Flaum Eye Institute, Rochester, NY, United States
- Department of Environmental Medicine, School of Medicine and Dentistry, University of Rochester, Rochester, NY, United States
- *Correspondence: Collynn F. Woeller,
| |
Collapse
|
18
|
Lowery R, Latchney S, Peer R, Lamantia C, Lordy K, Opanashuk L, McCall M, Majewska A. Gestational and lactational exposure to 2,3,7,8-tetrachlorodibenzo-p-dioxin primes cortical microglia to tissue injury. Brain Behav Immun 2022; 101:288-303. [PMID: 35065196 PMCID: PMC9007156 DOI: 10.1016/j.bbi.2022.01.013] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 12/22/2021] [Accepted: 01/16/2022] [Indexed: 11/16/2022] Open
Abstract
Recent studies have shown that the aryl hydrocarbon receptor (AhR) is expressed in the brain's native immune cells, known as microglia. However, while the impact of exposure to AhR ligands is well studied in the peripheral immune system, the impact of such exposure on immune function in the brain is less well defined. Microglia serve dual roles in providing synaptic and immunological support for neighboring neurons and in mediating responses to environmental stimuli, including exposure to environmental chemicals. Because of their dual roles in regulating physiological and pathological processes, cortical microglia are well positioned to translate toxic stimuli into defects in cortical function via aberrant synaptic and immunological functioning, mediated either through direct microglial AhR activation or in response to AhR activation in neighboring cells. Here, we use gene expression studies, histology, and two-photon in vivo imaging to investigate how developmental exposure to 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), a high-affinity and persistent AhR agonist, modulates microglial characteristics and function in the intact brain. Whole cortical RT-qPCR analysis and RNA-sequencing of isolated microglia revealed that gestational and lactational TCDD exposure produced subtle, but durable, changes in microglia transcripts. Histological examination and two-photon in vivo imaging revealed that while microglia density, distribution, morphology, and motility were unaffected by TCDD exposure, exposure resulted in microglia that responded more robustly to focal tissue injury. However, this effect was rectified with depletion and repopulation of microglia. These results suggest that gestational and lactational exposure to AhR ligands can result in long-term priming of microglia to produce heightened responses towards tissue injury which can be restored to normal function through microglial repopulation.
Collapse
Affiliation(s)
- R.L. Lowery
- Department of Neuroscience, Center for Visual Science, University of Rochester, Rochester, NY 14642
| | - S.E. Latchney
- Department of Neuroscience, Center for Visual Science, University of Rochester, Rochester, NY 14642
| | - R.P. Peer
- Department of Neuroscience, Center for Visual Science, University of Rochester, Rochester, NY 14642
| | - C.E. Lamantia
- Department of Neuroscience, Center for Visual Science, University of Rochester, Rochester, NY 14642
| | - K.A. Lordy
- Department of Neuroscience, Center for Visual Science, University of Rochester, Rochester, NY 14642
| | | | - M. McCall
- Department of Biostatistics and Computational Biology, University of Rochester, NY 14642,Department of Biomedical Genetics, University of Rochester, NY 14642
| | - A.K Majewska
- Department of Neuroscience, Center for Visual Science, University of Rochester, Rochester, NY 14642,Corresponding Author: Ania K. Majewska, University of Rochester, School of Medicine and Dentistry, Department of Neuroscience, Center for Visual Science, 601 Elmwood Avenue, Box 603, Rochester, New York 14642, , Phone: (585) 276-2254
| |
Collapse
|
19
|
Chen Y, Dong Y, Lu X, Li W, Zhang Y, Mao B, Pan X, Li X, Zhou Y, An Q, Xie F, Wang S, Xue Y, Cai X, Lai M, Zhou Q, Yan Y, Fu R, Wang H, Nakahata T, An X, Shi L, Zhang Y, Ma F. Inhibition of aryl hydrocarbon receptor signaling promotes the terminal differentiation of human erythroblasts. J Mol Cell Biol 2022; 14:6504013. [PMID: 35022784 PMCID: PMC9122643 DOI: 10.1093/jmcb/mjac001] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 11/06/2021] [Indexed: 11/12/2022] Open
Abstract
The aryl hydrocarbon receptor (AHR) plays an important role during mammalian embryo development. Inhibition of AHR signaling promotes the development of hematopoietic stem/progenitor cells. AHR also regulates the functional maturation of blood cells, such as T cells and megakaryocytes. However, little is known about the role of AHR modulation during the development of erythroid cells. In this study, we used the AHR antagonist StemRegenin 1 (SR1) and the AHR agonist 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) during different stages of human erythropoiesis to elucidate the function of AHR. We found that antagonizing AHR signaling improved the production of human embryonic stem cell (hESC)-derived erythrocytes and enhanced erythroid terminal differentiation. RNA-sequencing showed that SR1 treatment of proerythroblasts upregulated the expression of erythrocyte differentiation-related genes and downregulated actin organization-associated genes. We found that SR1 accelerated F-actin remodeling in terminally differentiated erythrocytes, favoring their maturation of the cytoskeleton and enucleation. We demonstrated that the effects of AHR inhibition on erythroid maturation were associated with F-actin remodeling. Our findings help uncover the mechanism for AHR-mediated human erythroid cell differentiation. We also provide a new approach toward the large-scale production of functionally mature human pluripotent stem cell-derived erythrocytes for use in translational applications.
Collapse
Affiliation(s)
- Yijin Chen
- Institute of Blood Transfusion, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Chengdu, China
| | - Yong Dong
- Institute of Blood Transfusion, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Chengdu, China
| | - Xulin Lu
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, CAMS & PUMC, Tianjin, China
| | - Wanjing Li
- Institute of Blood Transfusion, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Chengdu, China
| | - Yimeng Zhang
- Institute of Blood Transfusion, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Chengdu, China
| | - Bin Mao
- Institute of Blood Transfusion, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Chengdu, China
| | - Xu Pan
- Institute of Blood Transfusion, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Chengdu, China
| | - Xiaohong Li
- Institute of Blood Transfusion, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Chengdu, China
| | - Ya Zhou
- Institute of Blood Transfusion, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Chengdu, China
| | - Quanming An
- Institute of Blood Transfusion, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Chengdu, China
| | - Fangxin Xie
- Institute of Blood Transfusion, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Chengdu, China
| | | | - Yuan Xue
- Institute of Blood Transfusion, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Chengdu, China
| | - Xinping Cai
- Institute of Blood Transfusion, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Chengdu, China
| | - Mowen Lai
- Institute of Blood Transfusion, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Chengdu, China
| | - Qiongxiu Zhou
- Institute of Blood Transfusion, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Chengdu, China
| | - Yan Yan
- Jinjiang Maternity and child health hospital, Chengdu, China
| | - Ruohan Fu
- Jinjiang Maternity and child health hospital, Chengdu, China
| | - Hong Wang
- Institute of Blood Transfusion, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Chengdu, China
| | - Tatsutoshi Nakahata
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Xiuli An
- Laboratory of Membrane Biology, New York Blood Center, New York, NY
| | - Lihong Shi
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, CAMS & PUMC, Tianjin, China
| | - Yonggang Zhang
- Institute of Blood Transfusion, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Chengdu, China
| | - Feng Ma
- Institute of Blood Transfusion, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Chengdu, China.,State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, CAMS & PUMC, Tianjin, China
| |
Collapse
|
20
|
Qian C, Yang C, Lu M, Bao J, Shen H, Deng B, Li S, Li W, Zhang M, Cao C. Activating AhR alleviates cognitive deficits of Alzheimer's disease model mice by upregulating endogenous Aβ catabolic enzyme Neprilysin. Theranostics 2021; 11:8797-8812. [PMID: 34522212 PMCID: PMC8419060 DOI: 10.7150/thno.61601] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Accepted: 08/02/2021] [Indexed: 02/07/2023] Open
Abstract
Rationale: Neprilysin (NEP) is a major endogenous catabolic enzyme of amyloid β (Aβ). Previous studies have suggested that increasing NEP expression in animal models of Alzheimer's disease had an ameliorative effect. However, the underlying signaling pathway that regulates NEP expression remains unclear. The aryl hydrocarbon receptor (AhR) is a ligand-activated cytoplasmic receptor and transcription factor. Recent studies have shown that AhR plays essential roles in the central nervous system (CNS), but its physiological and pathological roles in regulating NEP are not entirely known. Methods: Western blotting, immunofluorescence, quantitative RT-PCR and enzyme activity assay were used to verify the effects of AhR agonists on NEP in a cell model (N2a) and a mouse model (APP/PS1). Luciferase reporter assay and chromatin immunoprecipitation (ChIP) assay were conducted to investigate the roles of AhR in regulating NEP transcription. Object recognition test and the Morris water maze task were performed to assess the cognitive capacity of the mice. Results: Activating AhR by the endogenous ligand L-Kynurenine (L-KN) or FICZ, or by the exogenous ligand diosmin or indole-3-carbinol (I3C) significantly increases NEP expression and enzyme activity in N2a cells and APP/PS1 mice. We also found that AhR is a direct transcription factor of NEP. Diosmin treatment effectively ameliorated the cognitive disorder and memory deficit of APP/PS1 transgenic mice. By knocking down AhR or using a small molecular inhibitor targeting AhR or NEP, we found that diosmin enhanced Aβ degradation through activated AhR and increased NEP expression. Conclusions: These results indicate a novel pathway for regulating NEP expression in neurons and that AhR may be a potential therapeutic target for the treatment of Alzheimer's disease.
Collapse
|
21
|
Larigot L, Benoit L, Koual M, Tomkiewicz C, Barouki R, Coumoul X. Aryl Hydrocarbon Receptor and Its Diverse Ligands and Functions: An Exposome Receptor. Annu Rev Pharmacol Toxicol 2021; 62:383-404. [PMID: 34499523 DOI: 10.1146/annurev-pharmtox-052220-115707] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The aryl hydrocarbon receptor (AhR) is a transcriptional factor that regulates multiple functions following its activation by a variety of ligands, including xenobiotics, natural products, microbiome metabolites, and endogenous molecules. Because of this diversity, the AhR constitutes an exposome receptor. One of its main functions is to regulate several lines of defense against chemical insults and bacterial infections. Indeed, in addition to its well-established detoxication function, it has several functions at physiological barriers, and it plays a critical role in immunomodulation. The AhR is also involved in the development of several organs and their homeostatic maintenance. Its activity depends on the type of ligand and on the time frame of the receptor activation, which can be either sustained or transient, leading in some cases to opposite modes of regulations as illustrated in the regulation of different cancer pathways. The development of selective modulators and their pharmacological characterization are important areas of research. Expected final online publication date for the Annual Review of Pharmacology and Toxicology, Volume 62 is January 2022. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Lucie Larigot
- INSERM UMR-S1124, T3S, Toxicologie Environnementale, Cibles thérapeutiques, Signalisation cellulaire et Biomarqueurs, and Université de Paris, 75006 Paris, France;
| | - Louise Benoit
- INSERM UMR-S1124, T3S, Toxicologie Environnementale, Cibles thérapeutiques, Signalisation cellulaire et Biomarqueurs, and Université de Paris, 75006 Paris, France; .,Service de Chirurgie Cancérologique Gynécologique et du Sein, Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges-Pompidou, 75015 Paris, France
| | - Meriem Koual
- INSERM UMR-S1124, T3S, Toxicologie Environnementale, Cibles thérapeutiques, Signalisation cellulaire et Biomarqueurs, and Université de Paris, 75006 Paris, France; .,Service de Chirurgie Cancérologique Gynécologique et du Sein, Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges-Pompidou, 75015 Paris, France
| | - Céline Tomkiewicz
- INSERM UMR-S1124, T3S, Toxicologie Environnementale, Cibles thérapeutiques, Signalisation cellulaire et Biomarqueurs, and Université de Paris, 75006 Paris, France;
| | - Robert Barouki
- INSERM UMR-S1124, T3S, Toxicologie Environnementale, Cibles thérapeutiques, Signalisation cellulaire et Biomarqueurs, and Université de Paris, 75006 Paris, France; .,Service de Chirurgie Cancérologique Gynécologique et du Sein, Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges-Pompidou, 75015 Paris, France
| | - Xavier Coumoul
- INSERM UMR-S1124, T3S, Toxicologie Environnementale, Cibles thérapeutiques, Signalisation cellulaire et Biomarqueurs, and Université de Paris, 75006 Paris, France;
| |
Collapse
|
22
|
Gileadi TE, Swamy AK, Hore Z, Horswell S, Ellegood J, Mohan C, Mizuno K, Lundebye AK, Giese KP, Stockinger B, Hogstrand C, Lerch JP, Fernandes C, Basson MA. Effects of Low-Dose Gestational TCDD Exposure on Behavior and on Hippocampal Neuron Morphology and Gene Expression in Mice. ENVIRONMENTAL HEALTH PERSPECTIVES 2021; 129:57002. [PMID: 33956508 PMCID: PMC8101924 DOI: 10.1289/ehp7352] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 02/19/2021] [Accepted: 03/29/2021] [Indexed: 05/03/2023]
Abstract
BACKGROUND 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) is a persistent and toxic environmental pollutant. Gestational exposure to TCDD has been linked to cognitive and motor deficits, and increased incidence of autism spectrum disorder (ASD) traits in children. Most animal studies of these neurodevelopmental effects involve acute TCDD exposure, which does not model typical exposure in humans. OBJECTIVES The aim of the study was to establish a dietary low-dose gestational TCDD exposure protocol and performed an initial characterization of the effects on offspring behavior, neurodevelopmental phenotypes, and gene expression. METHODS Throughout gestation, pregnant C57BL/6J mice were fed a diet containing a low dose of TCDD (9 ng TCDD/kg body weight per day) or a control diet. The offspring were tested in a battery of behavioral tests, and structural brain alterations were investigated by magnetic resonance imaging. The dendritic morphology of pyramidal neurons in the hippocampal Cornu Ammonis (CA)1 area was analyzed. RNA sequencing was performed on hippocampi of postnatal day 14 TCDD-exposed and control offspring. RESULTS TCDD-exposed females displayed subtle deficits in motor coordination and reversal learning. Volumetric difference between diet groups were observed in regions of the hippocampal formation, mammillary bodies, and cerebellum, alongside higher dendritic arborization of pyramidal neurons in the hippocampal CA1 region of TCDD-exposed females. RNA-seq analysis identified 405 differentially expressed genes in the hippocampus, enriched for genes with functions in regulation of microtubules, axon guidance, extracellular matrix, and genes regulated by SMAD3. DISCUSSION Exposure to 9 ng TCDD/kg body weight per day throughout gestation was sufficient to cause specific behavioral and structural brain phenotypes in offspring. Our data suggest that alterations in SMAD3-regulated microtubule polymerization in the developing postnatal hippocampus may lead to an abnormal morphology of neuronal dendrites that persists into adulthood. These findings show that environmental low-dose gestational exposure to TCDD can have significant, long-term impacts on brain development and function. https://doi.org/10.1289/EHP7352.
Collapse
Affiliation(s)
- Talia E. Gileadi
- Centre for Craniofacial and Regenerative Biology, King’s College London, London, UK
| | - Abhyuday K. Swamy
- Centre for Craniofacial and Regenerative Biology, King’s College London, London, UK
| | - Zoe Hore
- Social, Genetic & Developmental Psychiatry Centre, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, UK
| | - Stuart Horswell
- Department of Bioinformatics and Biostatistics, The Francis Crick Institute, London, UK
| | - Jacob Ellegood
- Mouse Imaging Centre (MICe), Hospital for Sick Children, Toronto, Ontario, Canada
| | - Conor Mohan
- Centre for Craniofacial and Regenerative Biology, King’s College London, London, UK
| | - Keiko Mizuno
- Department of Basic and Clinical Neuroscience, King’s College London, London, UK
| | | | - K. Peter Giese
- Department of Basic and Clinical Neuroscience, King’s College London, London, UK
| | | | | | - Jason P. Lerch
- Mouse Imaging Centre (MICe), Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
- Wellcome Centre for Integrative Neuroimaging, University of Oxford, Oxford, UK
| | - Cathy Fernandes
- Social, Genetic & Developmental Psychiatry Centre, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, UK
- MRC Centre for Neurodevelopmental Disorders, King’s College London, London, UK
| | - M. Albert Basson
- Centre for Craniofacial and Regenerative Biology, King’s College London, London, UK
- MRC Centre for Neurodevelopmental Disorders, King’s College London, London, UK
| |
Collapse
|
23
|
Esteban J, Sánchez-Pérez I, Hamscher G, Miettinen HM, Korkalainen M, Viluksela M, Pohjanvirta R, Håkansson H. Role of aryl hydrocarbon receptor (AHR) in overall retinoid metabolism: Response comparisons to 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) exposure between wild-type and AHR knockout mice. Reprod Toxicol 2021; 101:33-49. [PMID: 33607186 DOI: 10.1016/j.reprotox.2021.02.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 01/20/2021] [Accepted: 02/10/2021] [Indexed: 02/06/2023]
Abstract
Young adult wild-type and aryl hydrocarbon receptor knockout (AHRKO) mice of both sexes and the C57BL/6J background were exposed to 10 weekly oral doses of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD; total dose of 200 μg/kg bw) to further characterize the observed impacts of AHR as well as TCDD on the retinoid system. Unexposed AHRKO mice harboured heavier kidneys, lighter livers and lower serum all-trans retinoic acid (ATRA) and retinol (REOH) concentrations than wild-type mice. Results from the present study also point to a role for the murine AHR in the control of circulating REOH and ATRA concentrations. In wild-type mice, TCDD elevated liver weight and reduced thymus weight, and drastically reduced the hepatic concentrations of 9-cis-4-oxo-13,14-dihydro-retinoic acid (CORA) and retinyl palmitate (REPA). In female wild-type mice, TCDD increased the hepatic concentration of ATRA as well as the renal and circulating REOH concentrations. Renal CORA concentrations were substantially diminished in wild-type male mice exclusively following TCDD-exposure, with a similar tendency in serum. In contrast, TCDD did not affect any of these toxicity or retinoid system parameters in AHRKO mice. Finally, a distinct sex difference occurred in kidney concentrations of all the analysed retinoid forms. Together, these results strengthen the evidence of a mandatory role of AHR in TCDD-induced retinoid disruption, and suggest that the previously reported accumulation of several retinoid forms in the liver of AHRKO mice is a line-specific phenomenon. Our data further support participation of AHR in the control of liver and kidney development in mice.
Collapse
Affiliation(s)
- Javier Esteban
- Instituto De Bioingeniería, Universidad Miguel Hernández De Elche, Elche, Alicante, Spain.
| | - Ismael Sánchez-Pérez
- Instituto De Bioingeniería, Universidad Miguel Hernández De Elche, Elche, Alicante, Spain.
| | - Gerd Hamscher
- Institute of Food Chemistry and Food Biotechnology, Justus Liebig University Giessen, Giessen, Germany.
| | - Hanna M Miettinen
- School of Pharmacy (Toxicology) and Department of Environmental and Biological Sciences, University of Eastern Finland, Kuopio, Finland.
| | - Merja Korkalainen
- Environmental Health Unit, Finnish Insitute for Health and Welfare (THL), Kuopio, Finland.
| | - Matti Viluksela
- School of Pharmacy (Toxicology) and Department of Environmental and Biological Sciences, University of Eastern Finland, Kuopio, Finland; Environmental Health Unit, Finnish Insitute for Health and Welfare (THL), Kuopio, Finland.
| | - Raimo Pohjanvirta
- Department of Food Hygiene & Environmental Health, Faculty of Veterinary Medicine, University of Helsinki, Mustialankatu 1, FI-00790 Helsinki, Finland.
| | - Helen Håkansson
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
24
|
Kou Z, Dai W. Aryl hydrocarbon receptor: Its roles in physiology. Biochem Pharmacol 2021; 185:114428. [PMID: 33515530 PMCID: PMC8862184 DOI: 10.1016/j.bcp.2021.114428] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Revised: 01/15/2021] [Accepted: 01/19/2021] [Indexed: 12/27/2022]
Abstract
Aryl hydrocarbon receptor (AHR) was initially discovered as a cellular protein involved in mediating the detoxification of xenobiotic compounds. Extensive research in the past two decades has identified several families of physiological ligands and uncovered important functions of AHR in normal development and homeostasis. Deficiency in AHR expression disrupts major signaling systems and transcriptional programs, which appear to be responsible for the development of numerous developmental abnormalities including cardiac hypertrophy and epidermal hyperplasia. This mini review primarily summarizes recent advances in our understanding of AHR functions in normal physiology with an emphasis on the cardiovascular, gastrointestinal, integumentary, nervous, and immunomodulatory systems.
Collapse
Affiliation(s)
- Ziyue Kou
- Department of Environmental Medicine, New York University Langone Medical Center, NY 10010, United States
| | - Wei Dai
- Department of Environmental Medicine, New York University Langone Medical Center, NY 10010, United States.
| |
Collapse
|
25
|
Naffaa V, Laprévote O, Schang AL. Effects of endocrine disrupting chemicals on myelin development and diseases. Neurotoxicology 2020; 83:51-68. [PMID: 33352275 DOI: 10.1016/j.neuro.2020.12.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 12/10/2020] [Accepted: 12/16/2020] [Indexed: 12/14/2022]
Abstract
In the central and peripheral nervous systems, myelin is essential for efficient conduction of action potentials. During development, oligodendrocytes and Schwann cells differentiate and ensure axon myelination, and disruption of these processes can contribute to neurodevelopmental disorders. In adults, demyelination can lead to important disabilities, and recovery capacities by remyelination often decrease with disease progression. Among environmental chemical pollutants, endocrine disrupting chemicals (EDCs) are of major concern for human health and are notably suspected to participate in neurodevelopmental and neurodegenerative diseases. In this review, we have combined the current knowledge on EDCs impacts on myelin including several persistent organic pollutants, bisphenol A, triclosan, heavy metals, pesticides, and nicotine. Besides, we presented several other endocrine modulators, including pharmaceuticals and the phytoestrogen genistein, some of which are candidates for treating demyelinating conditions but could also be deleterious as contaminants. The direct impacts of EDCs on myelinating cells were considered as well as their indirect consequences on myelin, particularly on immune mechanisms associated with demyelinating conditions. More studies are needed to describe the effects of these compounds and to further understand the underlying mechanisms in relation to the potential for endocrine disruption.
Collapse
Affiliation(s)
- Vanessa Naffaa
- Université de Paris, UMR 8038 (CiTCoM), CNRS, Faculté de Pharmacie de Paris, 4 avenue de l'Observatoire, 75006 Paris, France.
| | - Olivier Laprévote
- Université de Paris, UMR 8038 (CiTCoM), CNRS, Faculté de Pharmacie de Paris, 4 avenue de l'Observatoire, 75006 Paris, France; Hôpital Européen Georges Pompidou, AP-HP, Service de Biochimie, 20 rue Leblanc, 75015 Paris, France.
| | - Anne-Laure Schang
- Université de Paris, UMR 1153 (CRESS), Faculté de Pharmacie de Paris, 4 avenue de l'Observatoire, 75006 Paris, France.
| |
Collapse
|
26
|
Armstrong TD, Suwannasual U, Kennedy CL, Thasma A, Schneider LJ, Phillippi D, Lund AK. Exposure to Traffic-Generated Pollutants Exacerbates the Expression of Factors Associated with the Pathophysiology of Alzheimer’s Disease in Aged C57BL/6 Wild-Type Mice. J Alzheimers Dis 2020; 78:1453-1471. [DOI: 10.3233/jad-200929] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Background: Multiple studies report a strong correlation between traffic-generated air pollution-exposure and detrimental outcomes in the central nervous system (CNS), including Alzheimer’s disease (AD). Incidence of AD is rapidly increasing and, worldwide, many live in regions where pollutants exceed regulatory standards. Thus, it is imperative to identify environmental pollutants that contribute to AD, and the mechanisms involved. Objective: We investigated the effects of mixed gasoline and diesel engine emissions (MVE) on the expression of factors involved in progression of AD in the hippocampus and cerebrum in a young versus aged mouse model. Methods: Young (2 months old) and aged (18 months old) male C57BL/6 mice were exposed to either MVE (300μg/m3 PM) or filtered air (FA) for 6 h/d, 7 d/wk, for 50 d. Immunofluorescence and RT-qPCR were used to quantify oxidative stress (8-OHdG) and expression of amyloid-β protein precursor (AβPP), β secretase (BACE1), amyloid-β (Aβ), aryl hydrocarbon receptor (AhR), cytochrome P450 (CYP) 1B1, angiotensin-converting enzyme (ACE1), and angiotensin II type 1 (AT1) receptor in the cerebrum and hippocampus, in addition to cerebral microvascular tight junction (TJ) protein expression. Results: We observed age-related increases in oxidative stress, AhR, CYP1B1, Aβ, BACE1, and AT1 receptor in the CA1 region of the hippocampus, and elevation of cerebral AβPP, AhR, and CYP1B1 mRNA, associated with decreased cerebral microvascular TJ protein claudin-5. MVE-exposure resulted in further promotion of oxidative stress, and significant increases in AhR, CYP1B1, BACE1, ACE1, and Aβ, compared to the young and aged FA-exposed mice. Conclusion: Such findings suggest that MVE-exposure exacerbates the expression of factors in the CNS associated with AD pathogenesis in aged populations.
Collapse
Affiliation(s)
- Tyler D. Armstrong
- Advanced Environmental Research Institute, Department of Biological Sciences, University of North Texas, Denton, TX, USA
| | - Usa Suwannasual
- Advanced Environmental Research Institute, Department of Biological Sciences, University of North Texas, Denton, TX, USA
| | - Conner L. Kennedy
- Advanced Environmental Research Institute, Department of Biological Sciences, University of North Texas, Denton, TX, USA
| | - Akshaykumar Thasma
- Advanced Environmental Research Institute, Department of Biological Sciences, University of North Texas, Denton, TX, USA
| | - Leah J. Schneider
- Advanced Environmental Research Institute, Department of Biological Sciences, University of North Texas, Denton, TX, USA
| | - Danielle Phillippi
- Advanced Environmental Research Institute, Department of Biological Sciences, University of North Texas, Denton, TX, USA
| | - Amie K. Lund
- Advanced Environmental Research Institute, Department of Biological Sciences, University of North Texas, Denton, TX, USA
| |
Collapse
|
27
|
Borovok N, Weiss C, Sharkia R, Reichenstein M, Wissinger B, Azem A, Mahajnah M. Gene and Protein Expression in Subjects With a Nystagmus-Associated AHR Mutation. Front Genet 2020; 11:582796. [PMID: 33193710 PMCID: PMC7542227 DOI: 10.3389/fgene.2020.582796] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 09/02/2020] [Indexed: 11/24/2022] Open
Abstract
Recently, a consanguineous family was identified in Israel with three children affected by Infantile Nystagmus and Foveal Hypoplasia, following an autosomal recessive mode of inheritance. A homozygous stop mutation c.1861C > T; p.Q621∗ in the aryl hydrocarbon receptor (AHR) gene (AHR; MIM 600253) was identified that co-segregated with the disease in the larger family. AHR is the first gene to be identified causing an autosomal recessive Infantile Nystagmus-related disease in humans. The goal of this study is to delineate the molecular basis of this newly discovered human genetic disorder associated with a rare AHR gene mutation. The gene and protein expression levels of AHR and selected AHR targets from leukocyte cultures of healthy subjects and the patients were analyzed. We observed significant variation between mRNA and protein expression of CYP1A1, CYP1B1, and TiPARP under rest and AHR-induced conditions. The CYP1A1 enzymatic activity in induced leukocytes also differs significantly between the patients and healthy volunteers. Intriguingly, the heterozygous subjects demonstrate CYP1A1 and TiPARP gene and protein expression similar to homozygous patients. In contrast, CYP1B1 inducibility and expression vary between hetero- and homozygous subjects. Similarity and differences in gene and protein expression between heterozygotes and homozygous patients can give us a hint as to which metabolic pathway/s might be involved in the Nystagmus etiology. Thus, we have a unique human model for AHR deficiency that will allow us the opportunity to study the biochemical basis of this rare human mutation, as well as the involvement of AHR in other physiological processes.
Collapse
Affiliation(s)
- Natalia Borovok
- Faculty of Life Sciences, School of Neurobiology, Biochemistry and Biophysics, Tel Aviv University, Tel Aviv, Israel
| | - Celeste Weiss
- Faculty of Life Sciences, School of Neurobiology, Biochemistry and Biophysics, Tel Aviv University, Tel Aviv, Israel
| | - Rajech Sharkia
- Triangle Research and Development Center, Kafr Qara, Israel.,Beit Berl College, Beit Berl, Israel
| | - Michal Reichenstein
- Faculty of Life Sciences, School of Neurobiology, Biochemistry and Biophysics, Tel Aviv University, Tel Aviv, Israel
| | - Bernd Wissinger
- Institute for Ophthalmic Research Centre for Ophthalmology, Eberhard Karls University of Tübingen, Tübingen, Germany
| | - Abdussalam Azem
- Faculty of Life Sciences, School of Neurobiology, Biochemistry and Biophysics, Tel Aviv University, Tel Aviv, Israel
| | - Muhammad Mahajnah
- Hillel Yaffe Medical Center, Hadera, Israel.,The Ruth and Bruce Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel
| |
Collapse
|
28
|
Khan AS, Langmann T. Indole-3-carbinol regulates microglia homeostasis and protects the retina from degeneration. J Neuroinflammation 2020; 17:327. [PMID: 33143743 PMCID: PMC7640677 DOI: 10.1186/s12974-020-01999-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 10/14/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Retinal degenerative diseases significantly contribute to visual impairment and blindness. Microglia reactivity is a hallmark of neurodegenerative diseases including retinal cell death and immunomodulation emerges as a therapeutic option. Indole-3-carbinol (I3C) is a natural ligand of aryl hydrocarbon receptor (AhR), with potent immunomodulatory properties. Here, we hypothesized that I3C may inhibit microglia reactivity and exert neuroprotective effects in the light-damaged murine retina mimicking important immunological aspects of retinal degeneration. METHODS BV-2 microglia were treated in vitro with I3C followed by lipopolysaccharide (LPS) stimulation to analyze pro-inflammatory and anti-oxidant responses by quantitative real-time PCR (qRT-PCR) and Western blots. Nitric oxide (NO) secretion, caspase 3/7 levels, phagocytosis rates, migration, and morphology were analyzed in control and AhR knockdown cells. I3C or vehicle was systemically applied to light-treated BALB/cJ mice as an experimental model of retinal degeneration. Pro-inflammatory and anti-oxidant responses in the retina were examined by qRT-PCR, ELISA, and Western blots. Immunohistochemical staining of retinal flat mounts and cryosections were performed. The retinal thickness and structure were evaluated by in vivo imaging using spectral domain-optical coherence tomography (SD-OCT). RESULTS The in vitro data showed that I3C potently diminished LPS-induced pro-inflammatory gene expression of I-NOS, IL-1ß, NLRP3, IL-6, and CCL2 and induced anti-oxidants gene levels of NQO1, HMOX1, and CAT1 in BV-2 cells. I3C also reduced LPS-induced NO secretion, phagocytosis, and migration as important functional microglia parameters. siRNA-mediated knockdown of AhR partially prevented the previously observed gene regulatory events. The in vivo experiments revealed that I3C treatment diminished light-damage induced I-NOS, IL-1ß, NLRP3, IL-6, and CCL2 transcripts and also reduced CCL2, I-NOS, IL-1ß, p-NFkBp65 protein levels in mice. Moreover, I3C increased anti-oxidant NQO1 and HMOX1 protein levels in light-exposed retinas. Finally, I3C therapy prevented the accumulation of amoeboid microglia in the subretinal space and protected from retinal degeneration. CONCLUSIONS The AhR ligand I3C potently counter-acts microgliosis and light-induced retinal damage, highlighting a potential treatment concept for retinal degeneration.
Collapse
Affiliation(s)
- Amir Saeed Khan
- Laboratory for Experimental Immunology of the Eye, Department of Ophthalmology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Joseph-Stelzmann-Str. 9, D-50931, Cologne, Germany
| | - Thomas Langmann
- Laboratory for Experimental Immunology of the Eye, Department of Ophthalmology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Joseph-Stelzmann-Str. 9, D-50931, Cologne, Germany.
- Center for Molecular Medicine Cologne, Cologne, Germany.
| |
Collapse
|
29
|
Choudhary M, Malek G. The Aryl Hydrocarbon Receptor: A Mediator and Potential Therapeutic Target for Ocular and Non-Ocular Neurodegenerative Diseases. Int J Mol Sci 2020; 21:ijms21186777. [PMID: 32947781 PMCID: PMC7555571 DOI: 10.3390/ijms21186777] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 09/10/2020] [Accepted: 09/11/2020] [Indexed: 02/06/2023] Open
Abstract
The aryl hydrocarbon receptor (AHR) is a ligand-activated transcription factor, which senses environmental, dietary or metabolic signals to mount a transcriptional response, vital in health and disease. As environmental stimuli and metabolic products have been shown to impact the central nervous system (CNS), a burgeoning area of research has been on the role of the AHR in ocular and non-ocular neurodegenerative diseases. Herein, we summarize our current knowledge, of AHR-controlled cellular processes and their impact on regulating pathobiology of select ocular and neurodegenerative diseases. We catalogue animal models generated to study the role of the AHR in tissue homeostasis and disease pathogenesis. Finally, we discuss the potential of targeting the AHR pathway as a therapeutic strategy, in the context of the maladies of the eye and brain.
Collapse
Affiliation(s)
- Mayur Choudhary
- Department of Ophthalmology, Duke University School of Medicine, 2351 Erwin Road, P.O. Box 3802, Durham, NC 27705, USA
- Correspondence: (M.C.); (G.M.)
| | - Goldis Malek
- Department of Ophthalmology, Duke University School of Medicine, 2351 Erwin Road, P.O. Box 3802, Durham, NC 27705, USA
- Department of Pathology, Duke University School of Medicine, Durham, NC 27705, USA
- Correspondence: (M.C.); (G.M.)
| |
Collapse
|
30
|
Aryl Hydrocarbon Receptor (AHR) Ligands as Selective AHR Modulators (SAhRMs). Int J Mol Sci 2020; 21:ijms21186654. [PMID: 32932962 PMCID: PMC7555580 DOI: 10.3390/ijms21186654] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 09/01/2020] [Accepted: 09/09/2020] [Indexed: 02/06/2023] Open
Abstract
The aryl hydrocarbon receptor (AhR) was first identified as the intracellular protein that bound and mediated the toxic effects of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD, dioxin) and dioxin-like compounds (DLCs). Subsequent studies show that the AhR plays an important role in maintaining cellular homeostasis and in pathophysiology, and there is increasing evidence that the AhR is an important drug target. The AhR binds structurally diverse compounds, including pharmaceuticals, phytochemicals and endogenous biochemicals, some of which may serve as endogenous ligands. Classification of DLCs and non-DLCs based on their persistence (metabolism), toxicities, binding to wild-type/mutant AhR and structural similarities have been reported. This review provides data suggesting that ligands for the AhR are selective AhR modulators (SAhRMs) that exhibit tissue/cell-specific AhR agonist and antagonist activities, and that their functional diversity is similar to selective receptor modulators that target steroid hormone and other nuclear receptors.
Collapse
|
31
|
Giovannoni F, Quintana FJ. The Role of Astrocytes in CNS Inflammation. Trends Immunol 2020; 41:805-819. [PMID: 32800705 DOI: 10.1016/j.it.2020.07.007] [Citation(s) in RCA: 328] [Impact Index Per Article: 65.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 07/10/2020] [Accepted: 07/10/2020] [Indexed: 01/18/2023]
Abstract
Astrocytes are the most abundant cell type in the central nervous system (CNS), performing complex functions in health and disease. It is now clear that multiple astrocyte subsets or activation states (plastic phenotypes driven by intrinsic and extrinsic cues) can be identified, associated to specific genomic programs and functions. The characterization of these subsets and the mechanisms that control them may provide unique insights into the pathogenesis of neurologic diseases, and identify potential targets for therapeutic intervention. In this article, we provide an overview of the role of astrocytes in CNS inflammation, highlighting recent discoveries on astrocyte subsets and the mechanisms that control them.
Collapse
Affiliation(s)
- Federico Giovannoni
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Francisco J Quintana
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|
32
|
Brown EM, Kenny DJ, Xavier RJ. Gut Microbiota Regulation of T Cells During Inflammation and Autoimmunity. Annu Rev Immunol 2020; 37:599-624. [PMID: 31026411 DOI: 10.1146/annurev-immunol-042718-041841] [Citation(s) in RCA: 199] [Impact Index Per Article: 39.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
The intestinal microbiota plays a crucial role in influencing the development of host immunity, and in turn the immune system also acts to regulate the microbiota through intestinal barrier maintenance and immune exclusion. Normally, these interactions are homeostatic, tightly controlled, and organized by both innate and adaptive immune responses. However, a combination of environmental exposures and genetic defects can result in a break in tolerance and intestinal homeostasis. The outcomes of these interactions at the mucosal interface have broad, systemic effects on host immunity and the development of chronic inflammatory or autoimmune disease. The underlying mechanisms and pathways the microbiota can utilize to regulate these diseases are just starting to emerge. Here, we discuss the recent evidence in this area describing the impact of microbiota-immune interactions during inflammation and autoimmunity, with a focus on barrier function and CD4+ T cell regulation.
Collapse
Affiliation(s)
- Eric M Brown
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, USA; , .,Center for Microbiome Informatics and Therapeutics, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
| | - Douglas J Kenny
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, USA; , .,Center for Microbiome Informatics and Therapeutics, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
| | - Ramnik J Xavier
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, USA; , .,Center for Microbiome Informatics and Therapeutics, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA.,Gastrointestinal Unit, Center for the Study of Inflammatory Bowel Disease, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114, USA;
| |
Collapse
|
33
|
Concentration and Duration of Indoxyl Sulfate Exposure Affects Osteoclastogenesis by Regulating NFATc1 via Aryl Hydrocarbon Receptor. Int J Mol Sci 2020; 21:ijms21103486. [PMID: 32429048 PMCID: PMC7278944 DOI: 10.3390/ijms21103486] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Revised: 05/09/2020] [Accepted: 05/12/2020] [Indexed: 12/19/2022] Open
Abstract
Indoxyl sulfate (IS) is a chronic kidney disease (CKD)-specific renal osteodystrophy metabolite that affects the nuclear factor of activated T-cells, cytoplasmic 1 (NFATc1), a transcription factor promoting osteoclastogenesis. However, the mechanisms underlying the regulation of NFATc1 by IS remain unknown. It is intriguing that the Aryl hydrocarbon receptor (AhR) plays a key role in osteoclastogenesis, since IS is an endogenous AhR agonist. This study investigates the relationship between IS concentration and osteoclast differentiation in Raw 264.7 cells, and examines the effects of different IS concentrations on NFATc1 expression through AhR signaling. Our data suggest that both osteoclastogenesis and NFATc1 are affected by IS through AhR signaling in both dose- and time-dependent manners. Osteoclast differentiation increases with short-term, low-dose IS exposure and decreases with long-term, high-dose IS exposure. Different IS levels switch the role of AhR from that of a ligand-activated transcription factor to that of an E3 ubiquitin ligase. We found that the AhR nuclear translocator may play an important role in the regulation of these dual functions of AhR under IS treatment. Altogether, this study demonstrates that the IS/AhR/NFATc1 signaling axis plays a critical role in osteoclastogenesis, indicating a potential role of AhR in the pathology and abnormality of bone turnover in CKD patients.
Collapse
|
34
|
Majumder S, Kono M, Lee YT, Byrnes C, Li C, Tuymetova G, Proia RL. A genome-wide CRISPR/Cas9 screen reveals that the aryl hydrocarbon receptor stimulates sphingolipid levels. J Biol Chem 2020; 295:4341-4349. [PMID: 32029474 PMCID: PMC7105297 DOI: 10.1074/jbc.ac119.011170] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 01/28/2020] [Indexed: 01/12/2023] Open
Abstract
Sphingolipid biosynthesis generates lipids for membranes and signaling that are crucial for many developmental and physiological processes. In some cases, large amounts of specific sphingolipids must be synthesized for specialized physiological functions, such as during axon myelination. How sphingolipid synthesis is regulated to fulfill these physiological requirements is not known. To identify genes that positively regulate membrane sphingolipid levels, here we employed a genome-wide CRISPR/Cas9 loss-of-function screen in HeLa cells using selection for resistance to Shiga toxin, which uses a plasma membrane-associated glycosphingolipid, globotriaosylceramide (Gb3), for its uptake. The screen identified several genes in the sphingolipid biosynthetic pathway that are required for Gb3 synthesis, and it also identified the aryl hydrocarbon receptor (AHR), a ligand-activated transcription factor widely involved in development and physiology, as being required for Gb3 biosynthesis. AHR bound and activated the gene promoter of serine palmitoyltransferase small subunit A (SPTSSA), which encodes a subunit of the serine palmitoyltransferase that catalyzes the first and rate-limiting step in de novo sphingolipid biosynthesis. AHR knockout HeLa cells exhibited significantly reduced levels of cell-surface Gb3, and both AHR knockout HeLa cells and tissues from Ahr knockout mice displayed decreased sphingolipid content as well as significantly reduced expression of several key genes in the sphingolipid biosynthetic pathway. The sciatic nerve of Ahr knockout mice exhibited both reduced ceramide content and reduced myelin thickness. These results indicate that AHR up-regulates sphingolipid levels and is important for full axon myelination, which requires elevated levels of membrane sphingolipids.
Collapse
Affiliation(s)
- Saurav Majumder
- Genetics of Development and Disease Branch, NIDDK, National Institutes of Health, Bethesda, Maryland 20892
| | - Mari Kono
- Genetics of Development and Disease Branch, NIDDK, National Institutes of Health, Bethesda, Maryland 20892
| | - Y Terry Lee
- Genetics of Development and Disease Branch, NIDDK, National Institutes of Health, Bethesda, Maryland 20892
| | - Colleen Byrnes
- Genetics of Development and Disease Branch, NIDDK, National Institutes of Health, Bethesda, Maryland 20892
| | - Cuiling Li
- Genetics of Development and Disease Branch, NIDDK, National Institutes of Health, Bethesda, Maryland 20892
| | - Galina Tuymetova
- Genetics of Development and Disease Branch, NIDDK, National Institutes of Health, Bethesda, Maryland 20892
| | - Richard L Proia
- Genetics of Development and Disease Branch, NIDDK, National Institutes of Health, Bethesda, Maryland 20892.
| |
Collapse
|
35
|
Ramos-García NA, Orozco-Ibarra M, Estudillo E, Elizondo G, Gómez Apo E, Chávez Macías LG, Sosa-Ortiz AL, Torres-Ramos MA. Aryl Hydrocarbon Receptor in Post-Mortem Hippocampus and in Serum from Young, Elder, and Alzheimer's Patients. Int J Mol Sci 2020; 21:ijms21061983. [PMID: 32183254 PMCID: PMC7139760 DOI: 10.3390/ijms21061983] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 03/09/2020] [Accepted: 03/10/2020] [Indexed: 11/16/2022] Open
Abstract
One of the characteristics of the cerebral aging process is the presence of chronic inflammation through glial cells, which is particularly significant in neurodegeneration. On the other hand, it has been demonstrated that the aryl hydrocarbon receptor (AHR) participates in the inflammatory response. Currently, evidence in animal models shows that the hallmarks of aging are associated with changes in the AHR levels. However, there is no information concerning the behavior and participation of AHR in the human aging brain or in Alzheimer’s disease (AD). We evaluated the expression of AHR in human hippocampal post-mortem tissue and its association with reactive astrocytes by immunohistochemistry. Besides this, we analyzed through ELISA the AHR levels in blood serum from young and elder participants, and from AD patients. The levels of AHR and glial fibrillar acid protein were higher in elder than in young post-mortem brain samples. AHR was localized mainly in the cytosol of astrocytes and displayed a pattern that resembles extracellular vesicles; this latter feature was more conspicuous in AD subjects. We found higher serum levels of AHR in AD patients than in the other participants. These results suggest that AHR participates in the aging process, and probably in the development of neurodegenerative diseases like AD.
Collapse
Affiliation(s)
- Nicte Alaide Ramos-García
- Unidad Periférica de Neurociencias, Instituto Nacional de Neurología y Neurocirugía/Universidad Nacional Autónoma de México. Av. Insurgentes Sur No. 3877 Col. La Fama, Tlalpan, C.P. 14269, Ciudad de México, Mexico;
- Departamento de Biología Celular, CINVESTAV-IPN, Av. Instituto Politécnico Nacional No. 2508, Col. San Pedro Zacatenco, Gustavo A. Madero, C.P. 07360, Ciudad de México, Mexico;
| | - Marisol Orozco-Ibarra
- Laboratorio de Neurobiología Molecular y Celular, Instituto Nacional de Neurología y Neurocirugía. Av. Insurgentes Sur No. 3877 Col. La Fama, Tlalpan, C.P. 14269, Ciudad de México, Mexico;
| | - Enrique Estudillo
- Laboratorio de Reprogramación Celular, Instituto Nacional de Neurología y Neurocirugía/Universidad Nacional Autónoma de México. Av. Insurgentes Sur No. 3877 Col. La Fama, Tlalpan, C.P. 14269, Ciudad de México, Mexico;
| | - Guillermo Elizondo
- Departamento de Biología Celular, CINVESTAV-IPN, Av. Instituto Politécnico Nacional No. 2508, Col. San Pedro Zacatenco, Gustavo A. Madero, C.P. 07360, Ciudad de México, Mexico;
| | - Erick Gómez Apo
- Hospital General de México, “Dr. Eduardo Liceaga”. Dr. Balmis No. 148, Col. Doctores, Cuauhtémoc, C.P. 06720, Ciudad de México, Mexico; (E.G.A.); (L.G.C.M.)
| | - Laura Graciela Chávez Macías
- Hospital General de México, “Dr. Eduardo Liceaga”. Dr. Balmis No. 148, Col. Doctores, Cuauhtémoc, C.P. 06720, Ciudad de México, Mexico; (E.G.A.); (L.G.C.M.)
| | - Ana Luisa Sosa-Ortiz
- Laboratorio de Demencias, Instituto Nacional de Neurología y Neurocirugía, Av. Insurgentes Sur No. 3877 Col. La Fama, Tlalpan, C.P. 14269, Ciudad de México, Mexico;
| | - Mónica Adriana Torres-Ramos
- Unidad Periférica de Neurociencias, Instituto Nacional de Neurología y Neurocirugía/Universidad Nacional Autónoma de México. Av. Insurgentes Sur No. 3877 Col. La Fama, Tlalpan, C.P. 14269, Ciudad de México, Mexico;
- Correspondence: ; Tel.: +52-55-56063822 (ext. 3045)
| |
Collapse
|
36
|
Mayer AK, Mahajnah M, Thomas MG, Cohen Y, Habib A, Schulze M, Maconachie GDE, AlMoallem B, De Baere E, Lorenz B, Traboulsi EI, Kohl S, Azem A, Bauer P, Gottlob I, Sharkia R, Wissinger B. Homozygous stop mutation in AHR causes autosomal recessive foveal hypoplasia and infantile nystagmus. Brain 2020; 142:1528-1534. [PMID: 31009037 DOI: 10.1093/brain/awz098] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Revised: 02/04/2019] [Accepted: 02/16/2019] [Indexed: 01/27/2023] Open
Abstract
Herein we present a consanguineous family with three children affected by foveal hypoplasia with infantile nystagmus, following an autosomal recessive mode of inheritance. The patients showed normal electroretinography responses, no signs of albinism, and no anterior segment or brain abnormalities. Upon whole exome sequencing, we identified a homozygous mutation (c.1861C>T;p.Q621*) in the aryl hydrocarbon receptor (AHR) gene that perfectly co-segregated with the disease in the larger family. AHR is a ligand-activated transcription factor that has been intensively studied in xenobiotic-induced toxicity. Further, it has been shown to play a physiological role under normal cellular conditions, such as in immunity, inflammatory response and neurogenesis. Notably, knockout of the Ahr gene in mouse impairs optic nerve myelin sheath formation and results in oculomotor deficits sharing many features with our patients: the eye movement disorder in Ahr-/- mice appears early in development and presents as conjugate horizontal pendular nystagmus. We therefore propose AHR to be a novel disease gene for a new, recessively inherited disorder in humans, characterized by infantile nystagmus and foveal hypoplasia.
Collapse
Affiliation(s)
- Anja K Mayer
- Molecular Genetics Laboratory, Institute for Ophthalmic Research, University of Tuebingen, Tuebingen, Germany
| | - Muhammad Mahajnah
- Child Neurology and Development Center, Hillel-Yaffe Medical Center, Hadera, Israel.,The Ruth and Bruce Rappaport Faculty of Medicine, Technion, Haifa, Israel
| | - Mervyn G Thomas
- Ulverscroft Eye Unit, Department of Neuroscience, Psychology and Behaviour, University of Leicester, Leicester, UK
| | - Yuval Cohen
- The Ruth and Bruce Rappaport Faculty of Medicine, Technion, Haifa, Israel.,Ophthalmology Department, Hillel Yaffe Medical Center, Hadera, Israel
| | - Adib Habib
- Pediatric Department, St. Vincent French Hospital, Nazareth, Israel
| | - Martin Schulze
- Institute of Medical Genetics and Applied Genomics, University of Tuebingen, Tuebingen, Germany
| | - Gail D E Maconachie
- Ulverscroft Eye Unit, Department of Neuroscience, Psychology and Behaviour, University of Leicester, Leicester, UK
| | - Basamat AlMoallem
- Center for Medical Genetics Ghent, Ghent University and Ghent University Hospital, Ghent, Belgium.,Department of Ophthalmology, King Abdul-Aziz University Hospital, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Elfride De Baere
- Center for Medical Genetics Ghent, Ghent University and Ghent University Hospital, Ghent, Belgium
| | - Birgit Lorenz
- Department of Ophthalmology, Justus Liebig University Giessen, Giessen, Germany
| | | | - Susanne Kohl
- Molecular Genetics Laboratory, Institute for Ophthalmic Research, University of Tuebingen, Tuebingen, Germany
| | - Abdussalam Azem
- Department of Biochemistry and Molecular Biology, Tel Aviv University, Tel Aviv, Israel
| | - Peter Bauer
- Institute of Medical Genetics and Applied Genomics, University of Tuebingen, Tuebingen, Germany
| | - Irene Gottlob
- Ulverscroft Eye Unit, Department of Neuroscience, Psychology and Behaviour, University of Leicester, Leicester, UK
| | - Rajech Sharkia
- The Triangle Regional Research and Development Center, Kfar Qari', Israel.,Beit-Berl Academic College, Beit-Berl, Israel
| | - Bernd Wissinger
- Molecular Genetics Laboratory, Institute for Ophthalmic Research, University of Tuebingen, Tuebingen, Germany
| |
Collapse
|
37
|
Celarain N, Tomas-Roig J. Aberrant DNA methylation profile exacerbates inflammation and neurodegeneration in multiple sclerosis patients. J Neuroinflammation 2020; 17:21. [PMID: 31937331 PMCID: PMC6961290 DOI: 10.1186/s12974-019-1667-1] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2019] [Accepted: 11/27/2019] [Indexed: 12/12/2022] Open
Abstract
Multiple sclerosis (MS) is an autoimmune and demyelinating disease of the central nervous system characterised by incoordination, sensory loss, weakness, changes in bladder capacity and bowel function, fatigue and cognitive impairment, creating a significant socioeconomic burden. The pathogenesis of MS involves both genetic susceptibility and exposure to distinct environmental risk factors. The gene x environment interaction is regulated by epigenetic mechanisms. Epigenetics refers to a complex system that modifies gene expression without altering the DNA sequence. The most studied epigenetic mechanism is DNA methylation. This epigenetic mark participates in distinct MS pathophysiological processes, including blood-brain barrier breakdown, inflammatory response, demyelination, remyelination failure and neurodegeneration. In this study, we also accurately summarised a list of environmental factors involved in the MS pathogenesis and its clinical course. A literature search was conducted using MEDLINE through PubMED and Scopus. In conclusion, an exhaustive study of DNA methylation might contribute towards new pharmacological interventions in MS by use of epigenetic drugs.
Collapse
Affiliation(s)
- Naiara Celarain
- Girona Neuroimmunology and Multiple Sclerosis Unit (UNIEM), Dr. Josep Trueta University Hospital and Girona Biomedical Research Institute (IDIBGI), Girona, Spain.
| | - Jordi Tomas-Roig
- Girona Neuroimmunology and Multiple Sclerosis Unit (UNIEM), Dr. Josep Trueta University Hospital and Girona Biomedical Research Institute (IDIBGI), Girona, Spain.
| |
Collapse
|
38
|
Mayer AK, Balousha G, Sharkia R, Mahajnah M, Ayesh S, Schulze M, Buchert R, Zobor D, Azem A, Schöls L, Bauer P, Wissinger B. Unraveling the genetic cause of hereditary ophthalmic disorders in Arab societies from Israel and the Palestinian Authority. Eur J Hum Genet 2020; 28:742-753. [PMID: 31896775 DOI: 10.1038/s41431-019-0566-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Revised: 11/12/2019] [Accepted: 12/10/2019] [Indexed: 11/09/2022] Open
Abstract
Visual impairment due to inherited ophthalmic disorders is amongst the most common disabilities observed in populations practicing consanguineous marriages. Here we investigated the molecular genetic basis of an unselected broad range of ophthalmic disorders in 20 consanguineous families from Arab villages of Israel and the Palestinian Authority. Most patients had little or very poor prior clinical workup and were recruited in a field study. Homozygosity mapping followed by candidate gene sequencing applying conventional Sanger sequencing or targeted next generation sequencing was performed in six families. In the remaining 14 families, one affected subject per family was chosen for whole exome sequencing. We discovered likely disease-causing variants, all homozygous, in 19 of 20 independent families (95%) including a previously reported novel disease gene for congenital nystagmus associated with foveal hypoplasia. Moreover, we found a family in which disease-causing variants for two collagenopathies - Stickler and Knobloch syndrome - segregate within a large sibship. Nine of the 19 distinct variants observed in this study were novel. Our study demonstrated a very high molecular diagnostic yield for a highly diverse spectrum of rare ophthalmic disorders in Arab patients from Israel and the Palestinian Authority, even with very limited prior clinical investigation. We conclude that 'genetic testing first' may be an economic way to direct clinical care and to support proper genetic counseling and risk assessment in these families.
Collapse
Affiliation(s)
- Anja K Mayer
- Institute for Ophthalmic Research, Molecular Genetics Laboratory, Tuebingen, Germany.,Praxis fuer Humangenetik Tuebingen, Tuebingen, Germany
| | - Ghassan Balousha
- Department of Pathology and Histology, Al-Quds University, Eastern Jerusalem, Palestinian Authority, Jerusalem, Israel
| | - Rajech Sharkia
- The Triangle Regional Research and Development Center, Kfar Qari', Israel.,Beit-Berl Academic College, Beit-Berl, Israel
| | - Muhammad Mahajnah
- Child Neurology and Development Center, Hillel-Yaffe Medical Center, Hadera, Israel.,The Ruth and Bruce Rappaport Faculty of Medicine, Technion, Haifa, Israel
| | - Suhail Ayesh
- Molecular Genetic Laboratory, Al-Makassed Islamic Charitable Society Hospital, Jerusalem, Israel
| | - Martin Schulze
- Institute of Medical Genetics and Applied Genomics, University of Tuebingen, Tuebingen, Germany.,Praxis fuer Humangenetik Tuebingen, Tuebingen, Germany
| | - Rebecca Buchert
- Institute of Medical Genetics and Applied Genomics, University of Tuebingen, Tuebingen, Germany
| | - Ditta Zobor
- University Eye Hospital, University of Tuebingen, Tuebingen, Germany
| | - Abdussalam Azem
- The School of Neurobiology, Biochemistry and Biophysics, George S. Wise faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Ludger Schöls
- Hertie Institute for Brain Research, University of Tuebingen, Tuebingen, Germany.,German Center of Neurodegenerative Diseases (DZNE), Tuebingen, Germany
| | - Peter Bauer
- Institute of Medical Genetics and Applied Genomics, University of Tuebingen, Tuebingen, Germany
| | - Bernd Wissinger
- Institute for Ophthalmic Research, Molecular Genetics Laboratory, Tuebingen, Germany.
| |
Collapse
|
39
|
The aryl hydrocarbon receptor: an environmental sensor integrating immune responses in health and disease. Nat Rev Immunol 2019; 19:184-197. [PMID: 30718831 DOI: 10.1038/s41577-019-0125-8] [Citation(s) in RCA: 700] [Impact Index Per Article: 116.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The environment, diet, microbiota and body's metabolism shape complex biological processes in health and disease. However, our understanding of the molecular pathways involved in these processes is still limited. The aryl hydrocarbon receptor (AHR) is a ligand-activated transcription factor that integrates environmental, dietary, microbial and metabolic cues to control complex transcriptional programmes in a ligand-specific, cell-type-specific and context-specific manner. In this Review, we summarize our current knowledge of AHR and the transcriptional programmes it controls in the immune system. Finally, we discuss the role of AHR in autoimmune and neoplastic diseases of the central nervous system, with a special focus on the gut immune system, the gut-brain axis and the therapeutic potential of targeting AHR in neurological disorders.
Collapse
|
40
|
Qiu J, Yang X, Wang L, Zhang Q, Ma W, Huang Z, Bao Y, Zhong L, Sun H, Ding F. Isoquercitrin promotes peripheral nerve regeneration through inhibiting oxidative stress following sciatic crush injury in mice. ANNALS OF TRANSLATIONAL MEDICINE 2019; 7:680. [PMID: 31930081 DOI: 10.21037/atm.2019.11.18] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Background Oxidative stress has been recognized to play a crucial role in the pathogenesis of peripheral nerve injury. Isoquercitrin (quercetin-3-glucoside) is a flavonoid that exhibited many biological activities, including anti-oxidative effect. However, it is unclear whether isoquercitrin has protective effects on peripheral nerve injury. Methods Mice treated by isoquercitrin were used as a case group, and mice injected with saline was the control group. Sciatic behavioral function was assessed using SFI and CMAPs were measured by electrophysiology. Schwann cells proliferation and migration were tested using EdU staining and Transwell migration chambers respectively. The expression of oxidative stress related factors were detected by qRT-PCR and Western blotting. Results In present study, our results demonstrated that isoquercitrin (20 mg/kg/day) treatment achieved significantly higher SFI and higher amplitude of CMAP, promoted the nerve regeneration and remyelination, increased the production of GAP43, NF200, MAG and PMP22, alleviated target muscle atrophy and autophagy, and suppressed the expression of ATG7, PINK1 and Beclin1 in soleus muscles after sciatic nerve crush. In vitro studies found that isoquercitrin promoted the axonal regeneration of DRGs neurons, the proliferation and migration of Schwann cells, and the expression of proliferating cell nuclear antigen (PCNA) in Schwann cells. The administration of isoquercitrin at 40 and 320 µM showed a dose dependent, and high doses of isoquercitrin (160 and 320 µM) showed better performance in promoting axonal regeneration of DRGs neurons, and the proliferation and migration of Schwann cells than low dose of isoquercitrin (40 µM). Furthermore, isoquercitrin significantly inhibited oxidative stress through reducing the production of Nox4 and Duox1, and promoting the expression of Nrf2 and SOD2 in soleus muscles after sciatic nerve crush. Conclusions Isoquercitrin may promote motor functional recovery and nerve regeneration following peripheral nerve injury though inhibition of oxidative stress, which highlighted the therapeutic values of isoquercitrin as a neuroprotective drug for peripheral nerve repair applications.
Collapse
Affiliation(s)
- Jiaying Qiu
- School of Biology and Basic Medical Sciences, Medical College of Soochow University, Suzhou 215123, China.,Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226001, China
| | - Xiaoming Yang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226001, China
| | - Lingbin Wang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226001, China
| | - Qiuyu Zhang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226001, China
| | - Wenjing Ma
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226001, China
| | - Ziwei Huang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226001, China
| | - Yuhua Bao
- Affiliated Hospital of Nantong University, Nantong University, Nantong 226001, China
| | - Lou Zhong
- Affiliated Hospital of Nantong University, Nantong University, Nantong 226001, China
| | - Hualin Sun
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226001, China
| | - Fei Ding
- School of Biology and Basic Medical Sciences, Medical College of Soochow University, Suzhou 215123, China.,Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226001, China
| |
Collapse
|
41
|
Wu PY, Chuang PY, Chang GD, Chan YY, Tsai TC, Wang BJ, Lin KH, Hsu WM, Liao YF, Lee H. Novel Endogenous Ligands of Aryl Hydrocarbon Receptor Mediate Neural Development and Differentiation of Neuroblastoma. ACS Chem Neurosci 2019; 10:4031-4042. [PMID: 31404492 DOI: 10.1021/acschemneuro.9b00273] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Aryl hydrocarbon receptor (AHR) signaling has been suggested to play roles in various physiological functions independent of its xenobiotic activity, including cell cycle regulation, immune response, and embryonic development. Several endogenous ligands were also identified by high-throughput screening techniques. However, the mechanism by which these molecules mediate AHR signaling in certain functions is still elusive. In this study, we investigated the possible pathway through which AHR and its endogenous ligands regulate neural development. We first identified two neuroactive steroids, 3α,5α-tetrahydrocorticosterone and 3α,5β-tetrahydrocorticosterone (5α- and 5β-THB), as novel AHR endogenous ligands through the use of an ultrasensitive dioxin-like compound bioassay and liquid chromatography-electrospray ionization-tandem mass spectrometry (LC-ESI-MS). We then treated zebrafish embryos with 5α- and 5β-THB, which enhance the expression of neurogenesis marker HuC. Furthermore, 5α- and 5β-THB both enhanced the expression of myelinating glial cell markers, sex determining region Y-box 10 (Sox10), and myelin-associated proteins myelin basic protein (Mbp) and improved the mobility of zebrafish larvae via the Ahr2 pathway. These results indicated that AHR mediates zebrafish neurogenesis and gliogenesis, especially the differentiation of oligodendrocyte or Schwann cells. Additionally, we showed that these molecules may induce neuroblastoma (NB) cell differentiation suggesting therapeutic potential of 5α- and 5β-THB in NB treatment. In summary, our results reveal that 5α- and 5β-THB are endogenous ligands of AHR and have therapeutic potential for NB treatment. By the interaction with THB, AHR signaling regulates various aspects of neural development.
Collapse
Affiliation(s)
- Pei-Yi Wu
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei 115, Taiwan
| | - Pei-Yun Chuang
- Department of Life Science, National Taiwan University, Taipei 10617, Taiwan
| | - Geen-Dong Chang
- Institute of Biochemical Science, National Taiwan University, Taipei 106, Taiwan
| | - Ya-Yun Chan
- Department of Life Science, National Taiwan University, Taipei 10617, Taiwan
| | - Tzu-Ching Tsai
- Department of Life Science, National Taiwan University, Taipei 10617, Taiwan
| | - Bo-Jeng Wang
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei 115, Taiwan
| | - Kuan-Hung Lin
- Department of Life Science, National Taiwan University, Taipei 10617, Taiwan
| | - Wen-Ming Hsu
- Department of Surgery, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei 100, Taiwan
| | - Yung-Feng Liao
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei 115, Taiwan
- Department of Life Science, National Taiwan University, Taipei 10617, Taiwan
| | - Hsinyu Lee
- Department of Life Science, National Taiwan University, Taipei 10617, Taiwan
- Department of Electrical Engineering, National Taiwan University, Taipei 10617, Taiwan
- Institute of Biomedical Electronics and Bioinformatics, National Taiwan University, Taipei 10617, Taiwan
| |
Collapse
|
42
|
Bravo-Ferrer I, Cuartero MI, Medina V, Ahedo-Quero D, Peña-Martínez C, Pérez-Ruíz A, Fernández-Valle ME, Hernández-Sánchez C, Fernández-Salguero PM, Lizasoain I, Moro MA. Lack of the aryl hydrocarbon receptor accelerates aging in mice. FASEB J 2019; 33:12644-12654. [PMID: 31483997 DOI: 10.1096/fj.201901333r] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The aryl hydrocarbon receptor (AhR) is a ligand-activated transcription factor, largely known for its role in xenobiotic metabolism and detoxification as well as its crucial role as a regulator of inflammation. Here, we have compared a cohort wild-type and AhR-null mice along aging to study the relationship between this receptor and age-associated inflammation, termed as "inflammaging," both at a systemic and the CNS level. Our results show that AhR deficiency is associated with a premature aged phenotype, characterized by early inflammaging, as shown by an increase in plasma cytokines levels. The absence of AhR also promotes the appearance of brain aging anatomic features, such as the loss of the white matter integrity. In addition, AhR-/- mice present an earlier spatial memory impairment and an enhanced astrogliosis in the hippocampus when compared with their age-matched AhR+/+ controls. Importantly, we have found that AhR protein levels decrease with age in this brain structure, strongly suggesting a link between AhR and aging.-Bravo-Ferrer, I., Cuartero, M. I., Medina, V., Ahedo-Quero, D., Peña-Martínez, C., Pérez-Ruíz, A., Fernández-Valle, M. E., Hernández-Sánchez, C., Fernández-Salguero, P. M., Lizasoain, I., Moro, M. A. Lack of the aryl hydrocarbon receptor accelerates aging in mice.
Collapse
Affiliation(s)
- Isabel Bravo-Ferrer
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Madrid, Spain.,Instituto Universitario de Investigación en Neuroquímica (IUIN), Madrid, Spain.,Instituto de Investigación Hospital 12 de Octubre (i+12), Madrid, Spain
| | - María I Cuartero
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Madrid, Spain.,Instituto Universitario de Investigación en Neuroquímica (IUIN), Madrid, Spain.,Instituto de Investigación Hospital 12 de Octubre (i+12), Madrid, Spain
| | - Violeta Medina
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Madrid, Spain.,Instituto Universitario de Investigación en Neuroquímica (IUIN), Madrid, Spain.,Instituto de Investigación Hospital 12 de Octubre (i+12), Madrid, Spain
| | - Dalia Ahedo-Quero
- Escuela Superior de Medicina del Instituto Politécnico Nacional, Ciudad de México, México
| | - Carolina Peña-Martínez
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Madrid, Spain.,Instituto Universitario de Investigación en Neuroquímica (IUIN), Madrid, Spain.,Instituto de Investigación Hospital 12 de Octubre (i+12), Madrid, Spain
| | - Alberto Pérez-Ruíz
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Madrid, Spain.,Instituto Universitario de Investigación en Neuroquímica (IUIN), Madrid, Spain.,Instituto de Investigación Hospital 12 de Octubre (i+12), Madrid, Spain
| | - M Encarnación Fernández-Valle
- Unidad de Resonancia Magnética Nuclear (RMN), Centro de Apoyo a la Investigación (CAI) de Bioimagen, Universidad Complutense de Madrid (UCM), Madrid, Spain
| | - Catalina Hernández-Sánchez
- Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain.,Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Pedro M Fernández-Salguero
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias, Universidad de Extremadura, Badajoz, Badajoz, Spain
| | - Ignacio Lizasoain
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Madrid, Spain.,Instituto Universitario de Investigación en Neuroquímica (IUIN), Madrid, Spain.,Instituto de Investigación Hospital 12 de Octubre (i+12), Madrid, Spain
| | - María A Moro
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Madrid, Spain.,Instituto Universitario de Investigación en Neuroquímica (IUIN), Madrid, Spain.,Instituto de Investigación Hospital 12 de Octubre (i+12), Madrid, Spain
| |
Collapse
|
43
|
Nutritional Modulation of Immune and Central Nervous System Homeostasis: The Role of Diet in Development of Neuroinflammation and Neurological Disease. Nutrients 2019; 11:nu11051076. [PMID: 31096592 PMCID: PMC6566411 DOI: 10.3390/nu11051076] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2019] [Revised: 05/02/2019] [Accepted: 05/13/2019] [Indexed: 02/06/2023] Open
Abstract
The gut-microbiome-brain axis is now recognized as an essential part in the regulation of systemic metabolism and homeostasis. Accumulating evidence has demonstrated that dietary patterns can influence the development of metabolic alterations and inflammation through the effects of nutrients on a multitude of variables, including microbiome composition, release of microbial products, gastrointestinal signaling molecules, and neurotransmitters. These signaling molecules are, in turn, implicated in the regulation of the immune system, either promoting or inhibiting the production of pro-inflammatory cytokines and the expansion of specific leukocyte subpopulations, such as Th17 and Treg cells, which are relevant in the development of neuroinflammatory and neurodegenerative conditions. Metabolic diseases, like obesity and type 2 diabetes mellitus, are related to inadequate dietary patterns and promote variations in the aforementioned signaling pathways in patients with these conditions, which have been linked to alterations in neurological functions and mental health. Thus, maintenance of adequate dietary patterns should be an essential component of any strategy aiming to prevent neurological pathologies derived from systemic metabolic alterations. The present review summarizes current knowledge on the role of nutrition in the modulation of the immune system and its impact in the development of neuroinflammation and neurological disease.
Collapse
|
44
|
Mohammadi-Bardbori A, Omidi M, Arabnezhad MR. Impact of CH223191-Induced Mitochondrial Dysfunction on Its Aryl Hydrocarbon Receptor Agonistic and Antagonistic Activities. Chem Res Toxicol 2019; 32:691-697. [DOI: 10.1021/acs.chemrestox.8b00371] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Affiliation(s)
- Afshin Mohammadi-Bardbori
- Department of Pharmacology and Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mahmoud Omidi
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Mohammad-Reza Arabnezhad
- Department of Pharmacology and Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
45
|
Guerrina N, Traboulsi H, Eidelman DH, Baglole CJ. The Aryl Hydrocarbon Receptor and the Maintenance of Lung Health. Int J Mol Sci 2018; 19:E3882. [PMID: 30563036 PMCID: PMC6320801 DOI: 10.3390/ijms19123882] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Revised: 11/27/2018] [Accepted: 11/29/2018] [Indexed: 01/09/2023] Open
Abstract
Much of what is known about the Aryl Hydrocarbon Receptor (AhR) centers on its ability to mediate the deleterious effects of the environmental toxicant 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD; dioxin). However, the AhR is both ubiquitously-expressed and evolutionarily-conserved, suggesting that it evolved for purposes beyond strictly mediating responses to man-made environmental toxicants. There is growing evidence that the AhR is required for the maintenance of health, as it is implicated in physiological processes such as xenobiotic metabolism, organ development and immunity. Dysregulation of AhR expression and activity is also associated with a variety of disease states, particularly those at barrier organs such as the skin, gut and lungs. The lungs are particularly vulnerable to inhaled toxicants such as cigarette smoke. However, the role of the AhR in diseases such as chronic obstructive pulmonary disease (COPD)-a respiratory illness caused predominately by cigarette smoking-and lung cancer remains largely unexplored. This review will discuss the growing body of literature that provides evidence that the AhR protects the lungs against the damaging effects of cigarette smoke.
Collapse
Affiliation(s)
- Necola Guerrina
- Research Institute of the McGill University Health Centre, Montreal, QC H4A 3J1, Canada.
- Department of Pathology, McGill University, Montreal, QC H3A 2B4, Canada.
| | - Hussein Traboulsi
- Research Institute of the McGill University Health Centre, Montreal, QC H4A 3J1, Canada.
| | - David H Eidelman
- Research Institute of the McGill University Health Centre, Montreal, QC H4A 3J1, Canada.
- Department of Medicine, McGill University, Montreal, QC H4A 3J1, Canada.
| | - Carolyn J Baglole
- Research Institute of the McGill University Health Centre, Montreal, QC H4A 3J1, Canada.
- Department of Pathology, McGill University, Montreal, QC H3A 2B4, Canada.
- Department of Medicine, McGill University, Montreal, QC H4A 3J1, Canada.
- Department of Pharmacology and Therapeutics, McGill University, Montreal, QC H3G 1Y6, Canada.
| |
Collapse
|
46
|
Neavin DR, Liu D, Ray B, Weinshilboum RM. The Role of the Aryl Hydrocarbon Receptor (AHR) in Immune and Inflammatory Diseases. Int J Mol Sci 2018; 19:ijms19123851. [PMID: 30513921 PMCID: PMC6321643 DOI: 10.3390/ijms19123851] [Citation(s) in RCA: 184] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Revised: 11/27/2018] [Accepted: 11/29/2018] [Indexed: 12/17/2022] Open
Abstract
The aryl hydrocarbon receptor (AHR) is a nuclear receptor that modulates the response to environmental stimuli. It was recognized historically for its role in toxicology but, in recent decades, it has been increasingly recognized as an important modulator of disease—especially for its role in modulating immune and inflammatory responses. AHR has been implicated in many diseases that are driven by immune/inflammatory processes, including major depressive disorder, multiple sclerosis, rheumatoid arthritis, asthma, and allergic responses, among others. The mechanisms by which AHR has been suggested to impact immune/inflammatory diseases include targeted gene expression and altered immune differentiation. It has been suggested that single nucleotide polymorphisms (SNPs) that are near AHR-regulated genes may contribute to AHR-dependent disease mechanisms/pathways. Further, we have found that SNPs that are outside of nuclear receptor binding sites (i.e., outside of AHR response elements (AHREs)) may contribute to AHR-dependent gene regulation in a SNP- and ligand-dependent manner. This review will discuss the evidence and mechanisms of AHR contributions to immune/inflammatory diseases and will consider the possibility that SNPs that are outside of AHR binding sites might contribute to AHR ligand-dependent inter-individual variation in disease pathophysiology and response to pharmacotherapeutics.
Collapse
Affiliation(s)
- Drew R Neavin
- Mayo Clinic Graduate School of Biomedical Sciences, Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN 55902, USA.
| | - Duan Liu
- Division of Clinical Pharmacology, Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN 55902, USA.
| | - Balmiki Ray
- Division of Clinical Pharmacology, Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN 55902, USA.
| | - Richard M Weinshilboum
- Division of Clinical Pharmacology, Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN 55902, USA.
| |
Collapse
|
47
|
Expression, Localization, and Activity of the Aryl Hydrocarbon Receptor in the Human Placenta. Int J Mol Sci 2018; 19:ijms19123762. [PMID: 30486367 PMCID: PMC6321474 DOI: 10.3390/ijms19123762] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Revised: 11/23/2018] [Accepted: 11/23/2018] [Indexed: 12/12/2022] Open
Abstract
The human placenta is an organ between the blood of the mother and the fetus, which is essential for fetal development. It also plays a role as a selective barrier against environmental pollutants that may bypass epithelial barriers and reach the placenta, with implications for the outcome of pregnancy. The aryl hydrocarbon receptor (AhR) is one of the most important environmental-sensor transcription factors and mediates the metabolism of a wide variety of xenobiotics. Nevertheless, the identification of dietary and endogenous ligands of AhR suggest that it may also fulfil physiological functions with which pollutants may interfere. Placental AhR expression and activity is largely unknown. We established the cartography of AhR expression at transcript and protein levels, its cellular distribution, and its transcriptional activity toward the expression of its main target genes. We studied the profile of AhR expression and activity during different pregnancy periods, during trophoblasts differentiation in vitro, and in a trophoblast cell line. Using diverse methods, such as cell fractionation and immunofluorescence microscopy, we found a constitutive nuclear localization of AhR in every placental model, in the absence of any voluntarily-added exogenous activator. Our data suggest an intrinsic activation of AhR due to the presence of endogenous placental ligands.
Collapse
|
48
|
The Aryl Hydrocarbon Receptor and the Nervous System. Int J Mol Sci 2018; 19:ijms19092504. [PMID: 30149528 PMCID: PMC6163841 DOI: 10.3390/ijms19092504] [Citation(s) in RCA: 93] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Revised: 08/17/2018] [Accepted: 08/21/2018] [Indexed: 12/12/2022] Open
Abstract
The aryl hydrocarbon receptor (or AhR) is a cytoplasmic receptor of pollutants. It translocates into the nucleus upon binding to its ligands, and forms a heterodimer with ARNT (AhR nuclear translocator). The heterodimer is a transcription factor, which regulates the transcription of xenobiotic metabolizing enzymes. Expressed in many cells in vertebrates, it is mostly present in neuronal cell types in invertebrates, where it regulates dendritic morphology or feeding behavior. Surprisingly, few investigations have been conducted to unravel the function of the AhR in the central or peripheral nervous systems of vertebrates. In this review, we will present how the AhR regulates neural functions in both invertebrates and vertebrates as deduced mainly from the effects of xenobiotics. We will introduce some of the molecular mechanisms triggered by the well-known AhR ligand, 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), which impact on neuronal proliferation, differentiation, and survival. Finally, we will point out the common features found in mice that are exposed to pollutants, and in AhR knockout mice.
Collapse
|
49
|
Huang Y, He J, Liang H, Hu K, Jiang S, Yang L, Mei S, Zhu X, Yu J, Kijlstra A, Yang P, Hou S. Aryl Hydrocarbon Receptor Regulates Apoptosis and Inflammation in a Murine Model of Experimental Autoimmune Uveitis. Front Immunol 2018; 9:1713. [PMID: 30090104 PMCID: PMC6068235 DOI: 10.3389/fimmu.2018.01713] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Accepted: 07/12/2018] [Indexed: 12/14/2022] Open
Abstract
Uveitis is characterized as a common cause of blindness worldwide. Aryl hydrocarbon receptor (AhR), a ligand-activated nuclear receptor, has been implicated to play a role in human uveitis, although the exact mechanisms remain poorly understood. The purpose of this study was to enhance our knowledge concerning the role of AhR during intraocular inflammation. We immunized wild-type and AhR-knockout C57BL/6J mice with IRBP651–670 to induce experimental autoimmune uveitis (EAU). Disease severity was evaluated with both clinical and histopathological grading. Blood–retinal barrier (BRB) integrity was tested by Evans blue and tight junction proteins qualifications. Apoptosis was measured using TdT-mediated dUTP nick end labeling staining. Macrophage/microglia activation and polarization were studied by immunofluorescence and Western blot. Following EAU induction, AhR−/− mice had more severe clinical and histopathological manifestations of uveitis than AhR+/+ mice. Increased vascular permeability and apoptotic cells were observed in AhR−/− EAU mice when compared with AhR+/+ EAU mice. In addition, AhR−/− EAU mice showed evidence of a significantly increased macrophage/microglia cells and a stronger polarization from the M2 to the M1 phenotype as compared to AhR+/+ EAU mice. The levels of pro-inflammatory cytokines including tumor necrosis factor-α (TNF-α), interleukin (IL)-6, and IL-1β were increased in AhR−/− EAU mice, which was associated with the activation of NF-κB and signal transducers and activators of transcription (STAT) pathways. 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), an agonist of AhR, caused a significant decrease in the clinical and histopathological manifestations, preserved BRB integrity, reduced apoptotic cells, inhibited macrophage/microglia activation, and shifted their polarization from M1 toward M2. Moreover, decreased expression of pro-inflammatory cytokines including TNF-α, IL-6, and IL-1β and inhibition of NF-κB and STAT pathways were found in EAU mice following TCDD treatment. In conclusion, AhR activation with TCDD exhibits an immunomodulatory effect by reducing BRB breakdown, inhibiting retinal cell apoptosis, and reducing pro-inflammatory cytokine expression during EAU. The underlying mechanism may involve the modulation of macrophages/microglia polarization and the downregulation of NF-κB and STAT pathways.
Collapse
Affiliation(s)
- Yike Huang
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Eye Institute, Chongqing Key Laboratory of Ophthalmology, Chongqing, China
| | - Junchi He
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Eye Institute, Chongqing Key Laboratory of Ophthalmology, Chongqing, China
| | - Huaping Liang
- State Key Laboratory of Trauma, Burns and Combined Injury, Research Institute of Surgery, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Ke Hu
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Eye Institute, Chongqing Key Laboratory of Ophthalmology, Chongqing, China
| | - Shaoqiu Jiang
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Eye Institute, Chongqing Key Laboratory of Ophthalmology, Chongqing, China
| | - Lu Yang
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Eye Institute, Chongqing Key Laboratory of Ophthalmology, Chongqing, China
| | - Suyin Mei
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Eye Institute, Chongqing Key Laboratory of Ophthalmology, Chongqing, China
| | - Xiao Zhu
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Dongguan Scientific Research Center, Guangdong Medical University, Dongguan, China
| | - Jing Yu
- State Key Laboratory of Trauma, Burns and Combined Injury, Research Institute of Surgery, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Aize Kijlstra
- University Eye Clinic Maastricht, Maastricht, Netherlands
| | - Peizeng Yang
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Eye Institute, Chongqing Key Laboratory of Ophthalmology, Chongqing, China
| | - Shengping Hou
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Eye Institute, Chongqing Key Laboratory of Ophthalmology, Chongqing, China
| |
Collapse
|
50
|
Larigot L, Juricek L, Dairou J, Coumoul X. AhR signaling pathways and regulatory functions. BIOCHIMIE OPEN 2018; 7:1-9. [PMID: 30003042 PMCID: PMC6039966 DOI: 10.1016/j.biopen.2018.05.001] [Citation(s) in RCA: 375] [Impact Index Per Article: 53.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/11/2018] [Accepted: 05/28/2018] [Indexed: 12/12/2022]
Abstract
Animals and humans are exposed each day to a multitude of chemicals in the air, water and food. They have developed a battery of enzymes and transporters that facilitate the biotransformation and elimination of these compounds. Moreover, a majority of these enzymes and transporters are inducible due to the activation of xenobiotic receptors which act as transcription factors for the regulation of their target genes (such as xenobiotic metabolizing enzymes, see below §4 for the AhR). These receptors include several members of the nuclear/steroid receptor family (CAR for Constitutive Androstane Receptor, PXR for Pregnane X Receptor) but also the Aryl hydrocarbon Receptor or AhR, a member of the bHLH-PAS family (basic Helix-Loop-Helix - Period/ARNT/Single minded). In addition to the regulation of xenobiotic metabolism, numerous alternative functions have been characterized for the AhR since its discovery. These alternative functions will be described in this review along with its endogenous functions as revealed by experiments performed on knock-out animals.
Collapse
Affiliation(s)
- Lucie Larigot
- INSERM UMR-S 1124, Toxicologie Pharmacologie et Signalisation Cellulaire, 45 rue des Saints-Pères, 75006 Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, 45 rue des Saints-Pères, 75006 Paris, France
| | - Ludmila Juricek
- INSERM UMR-S 1124, Toxicologie Pharmacologie et Signalisation Cellulaire, 45 rue des Saints-Pères, 75006 Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, 45 rue des Saints-Pères, 75006 Paris, France
| | - Julien Dairou
- CNRS 8601, 45 rue des Saints-Pères, 75006 Paris, France
| | - Xavier Coumoul
- INSERM UMR-S 1124, Toxicologie Pharmacologie et Signalisation Cellulaire, 45 rue des Saints-Pères, 75006 Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, 45 rue des Saints-Pères, 75006 Paris, France
| |
Collapse
|