1
|
Chen C, Xiao Q, Wen Z, Gong F, Zhan H, Liu J, Li H, Jiao Y. Gut microbiome-derived indole-3-carboxaldehyde regulates stress vulnerability in chronic restraint stress by activating aryl hydrocarbon receptors. Pharmacol Res 2025; 213:107654. [PMID: 39946793 DOI: 10.1016/j.phrs.2025.107654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 01/19/2025] [Accepted: 02/11/2025] [Indexed: 03/09/2025]
Abstract
Chronic stress constitutes a major precipitating factor for Major Depressive Disorder (MDD), and comprehending individual differences in stress responses is crucial for the development of effective intervention strategies for MDD. Recent studies indicate that an individual's vulnerability to chronic stress is closely associated with gut microbiota composition, but the underlying mechanisms remain unclear. This study aims to investigate whether the gut microbiota and its metabolites can serve as gut-brain signaling molecules and explores how the gut microbiota affects stress sensitivity. Here, we showed that gut microbiome-derived indole-3-carboxaldehyde (I3C) can act as a gut-brain signaling molecule that links tryptophan metabolism by gut microbes to stress vulnerability in the host. First, we identified a specific reduction in gut microbiome-derived I3C levels in the hippocampus and colon through untargeted and targeted metabolomic analyses. Then, the study of gut microbiota suggested that the relative abundance of lactobacillus was reduced significantly in stress-susceptible rats, whereas fecal microbiota transplantation regulates stress vulnerability. Furthermore, supplementation with I3C and the representative I3C-producing strain, Lactobacillus reuteri, was shown to alleviate depression-like behaviors induced by chronic stress. Further research confirms that I3C can inhibit neuroinflammation and promote hippocampal neurogenesis through the aryl hydrocarbon receptors (AhR) signal pathway, thereby mitigating the host's susceptibility to stress. In conclusion, our findings elucidate that the gut microbiome-derived-I3C can help buffer the host's stress through the AhR/SOCS2/NF-κB/NLRP3 pathway, providing a gut-brain signaling basis for emotional behavior.
Collapse
Affiliation(s)
- Congcong Chen
- Jiangxi Province Key Laboratory of Traditional Chinese Medicine Pharmacology, Institute of Traditional Chinese Medicine Health Industry, China Academy of Chinese Medical Sciences, Nanchang 330115, China; Jiangxi Health Industry Institute of Traditional Chinese Medicine, Nanchang 330115, China.
| | - Qiang Xiao
- Jiangxi Province Key Laboratory of Traditional Chinese Medicine Pharmacology, Institute of Traditional Chinese Medicine Health Industry, China Academy of Chinese Medical Sciences, Nanchang 330115, China; Jiangxi Health Industry Institute of Traditional Chinese Medicine, Nanchang 330115, China
| | - Zhaoyi Wen
- Jiangxi Province Key Laboratory of Traditional Chinese Medicine Pharmacology, Institute of Traditional Chinese Medicine Health Industry, China Academy of Chinese Medical Sciences, Nanchang 330115, China; Jiangxi Health Industry Institute of Traditional Chinese Medicine, Nanchang 330115, China; Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Fenfang Gong
- Jiangxi Province Key Laboratory of Traditional Chinese Medicine Pharmacology, Institute of Traditional Chinese Medicine Health Industry, China Academy of Chinese Medical Sciences, Nanchang 330115, China; Jiangxi Health Industry Institute of Traditional Chinese Medicine, Nanchang 330115, China
| | - Huang Zhan
- Jiangxi Province Key Laboratory of Traditional Chinese Medicine Pharmacology, Institute of Traditional Chinese Medicine Health Industry, China Academy of Chinese Medical Sciences, Nanchang 330115, China; Jiangxi Health Industry Institute of Traditional Chinese Medicine, Nanchang 330115, China
| | - Jian Liu
- Jiangxi Province Key Laboratory of Traditional Chinese Medicine Pharmacology, Institute of Traditional Chinese Medicine Health Industry, China Academy of Chinese Medical Sciences, Nanchang 330115, China; Jiangxi Health Industry Institute of Traditional Chinese Medicine, Nanchang 330115, China
| | - Hui Li
- Jiangxi Province Key Laboratory of Traditional Chinese Medicine Pharmacology, Institute of Traditional Chinese Medicine Health Industry, China Academy of Chinese Medical Sciences, Nanchang 330115, China; Jiangxi Health Industry Institute of Traditional Chinese Medicine, Nanchang 330115, China; Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Yukun Jiao
- Jiangxi Province Key Laboratory of Traditional Chinese Medicine Pharmacology, Institute of Traditional Chinese Medicine Health Industry, China Academy of Chinese Medical Sciences, Nanchang 330115, China; Jiangxi Health Industry Institute of Traditional Chinese Medicine, Nanchang 330115, China.
| |
Collapse
|
2
|
Dallera CA, Placeres-Uray F, Mastromatteo-Alberga P, Dominguez-Torres M, Balleste AF, Gorthy AS, Rahimzadeh TS, Aliancin I, Dietrich WD, de Rivero Vaccari JP, Jacobs IC, Chlipala EA, Benton H, Zeligs MA, Atkins CM. 3,3'-Diindolylmethane improves pathology and neurological outcome following traumatic brain injury. Neurotherapeutics 2025; 22:e00531. [PMID: 39909809 PMCID: PMC12014418 DOI: 10.1016/j.neurot.2025.e00531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 01/15/2025] [Accepted: 01/15/2025] [Indexed: 02/07/2025] Open
Abstract
3,3'-Diindolylmethane (DIM), a naturally occurring bis-indole found in cruciferous vegetables and produced in small amounts in the normal flora of the human gut, has demonstrated neuroprotective benefits in models of CNS hypoxia and stroke. In the CNS, DIM modulates the activation of the aryl hydrocarbon receptor (AhR) and inhibits its pro-inflammatory effects. Although capable of crossing the blood brain barrier, DIM's bioavailability is limited by its low solubility. Dispersed BR4044 provides a nanoscale high-solubility DIM suspension with the potential for treating traumatic brain injury (TBI). The present study aimed to determine whether BR4044 treatment could reduce pathology and improve behavioral recovery following moderate TBI. Male Sprague Dawley rats received moderate fluid percussion injury or sham surgery followed by vehicle or BR4044 treatment in the acute recovery period. TBI BR4044 animals showed significantly reduced cortical and hippocampal edema and lower levels of serum-derived extracellular vesicles compared to TBI Vehicle animals. BR4044 treatment of TBI animals preserved sensorimotor function and associative fear memory. Cortical contusion size and neuronal loss in the parietal cortex and CA3 region of the hippocampus were also significantly reduced with BR4044 treatment. BR4044 also decreased microbleeding and nuclear AhR at the contusion site. This translational study demonstrates that BR4044 ameliorates pathology and improves neurological outcomes following TBI by reducing brain edema, lowering acute extracellular vesicle release, modulating AhR, preserving cortical and hippocampal neurons, reducing red blood cell (RBC) extravasation into the injured brain, and promoting behavioral recovery.
Collapse
Affiliation(s)
- Carlos A Dallera
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami Miller School of Medicine, 1095 NW 14th Terrace, Miami, FL, USA
| | - Fabiola Placeres-Uray
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami Miller School of Medicine, 1095 NW 14th Terrace, Miami, FL, USA
| | - Patrizzia Mastromatteo-Alberga
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami Miller School of Medicine, 1095 NW 14th Terrace, Miami, FL, USA
| | - Maria Dominguez-Torres
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami Miller School of Medicine, 1095 NW 14th Terrace, Miami, FL, USA
| | - Alyssa F Balleste
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami Miller School of Medicine, 1095 NW 14th Terrace, Miami, FL, USA
| | - Aditi S Gorthy
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami Miller School of Medicine, 1095 NW 14th Terrace, Miami, FL, USA
| | - Tyler S Rahimzadeh
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami Miller School of Medicine, 1095 NW 14th Terrace, Miami, FL, USA
| | - Isabelle Aliancin
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami Miller School of Medicine, 1095 NW 14th Terrace, Miami, FL, USA
| | - W Dalton Dietrich
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami Miller School of Medicine, 1095 NW 14th Terrace, Miami, FL, USA
| | - Juan Pablo de Rivero Vaccari
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami Miller School of Medicine, 1095 NW 14th Terrace, Miami, FL, USA
| | | | | | | | | | - Coleen M Atkins
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami Miller School of Medicine, 1095 NW 14th Terrace, Miami, FL, USA.
| |
Collapse
|
3
|
Procházková J, Kahounová Z, Vondráček J, Souček K. Aryl hydrocarbon receptor as a drug target in advanced prostate cancer therapy - obstacles and perspectives. Transcription 2025; 16:47-66. [PMID: 38547312 PMCID: PMC11970783 DOI: 10.1080/21541264.2024.2334106] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 03/08/2024] [Accepted: 03/19/2024] [Indexed: 04/04/2025] Open
Abstract
Aryl hydrocarbon receptor (AhR) is a transcription factor that is primarily known as an intracellular sensor of environmental pollution. After five decades, the list of synthetic and toxic chemicals that activate AhR signaling has been extended to include a number of endogenous compounds produced by various types of cells via their metabolic activity. AhR signaling is active from the very beginning of embryonal development throughout the life cycle and participates in numerous biological processes such as control of cell proliferation and differentiation, metabolism of aromatic compounds of endogenous and exogenous origin, tissue regeneration and stratification, immune system development and polarization, control of stemness potential, and homeostasis maintenance. AhR signaling can be affected by various pharmaceuticals that may help modulate abnormal AhR signaling and drive pathological states. Given their role in immune system development and regulation, AhR antagonistic ligands are attractive candidates for immunotherapy of disease states such as advanced prostate cancer, where an aberrant immune microenvironment contributes to cancer progression and needs to be reeducated. Advanced stages of prostate cancer are therapeutically challenging and characterized by decreased overall survival (OS) due to the metastatic burden. Therefore, this review addresses the role of AhR signaling in the development and progression of prostate cancer and discusses the potential of AhR as a drug target for the treatment of advanced prostate cancer upon entering the phase of drug resistance and failure of first-line androgen deprivation therapy.Abbreviation: ADC: antibody-drug conjugate; ADT: androgen deprivation therapy; AhR: aryl hydrocarbon receptor; AR: androgen receptor; ARE: androgen response element; ARPI: androgen receptor pathway inhibitor; mCRPC: metastatic castration-resistant prostate cancer; DHT: 5a-dihydrotestosterone; FICZ: 6-formylindolo[3,2-b]carbazole; 3-MC: 3-methylcholanthrene; 6-MCDF: 6-methyl-1,3,8-trichlorodibenzofuran; MDSCs: myeloid-derived suppressor cells; PAHs: polycyclic aromatic hydrocarbons; PCa: prostate cancer; TAMs: tumor-associated macrophages; TF: transcription factor; TCDD, 2,3,7,8-tetrachlorodibenzo-p-dioxin; TME: tumor microenvironment; TRAMP: transgenic adenocarcinoma of the mouse prostate; TROP2: tumor associated calcium signal transducer 2.
Collapse
Affiliation(s)
- Jiřina Procházková
- Department of Cytokinetics, Institute of Biophysics of the Czech Academy of Sciences, Brno, Czech Republic
| | - Zuzana Kahounová
- Department of Cytokinetics, Institute of Biophysics of the Czech Academy of Sciences, Brno, Czech Republic
| | - Jan Vondráček
- Department of Cytokinetics, Institute of Biophysics of the Czech Academy of Sciences, Brno, Czech Republic
| | - Karel Souček
- Department of Cytokinetics, Institute of Biophysics of the Czech Academy of Sciences, Brno, Czech Republic
| |
Collapse
|
4
|
Khandayataray P, Murthy MK. Dietary interventions in mitigating the impact of environmental pollutants on Alzheimer's disease - A review. Neuroscience 2024; 563:148-166. [PMID: 39542342 DOI: 10.1016/j.neuroscience.2024.11.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Revised: 10/23/2024] [Accepted: 11/07/2024] [Indexed: 11/17/2024]
Abstract
Numerous studies linking environmental pollutants to oxidative stress, inflammation, and neurotoxicity have assigned pollutants to several neurodegenerative disorders, including Alzheimer's disease (AD). Heavy metals, pesticides, air pollutants, and endocrine disruptor chemicals have been shown to play important roles in AD development, with some traditional functions in amyloid-β formation, tau kinase action, and neuronal degeneration. However, pharmacological management and supplementation have resulted in limited improvement. This raises the interesting possibility that activities usually considered preventive, including diet, exercise, or mental activity, might be more similar to treatment or therapy for AD. This review focuses on the effects of diet on the effects of environmental pollutants on AD. One of the primary issues addressed in this review is a group of specific diets, including the Mediterranean diet (MeDi), Dietary Approaches to Stop Hypertension (DASH), and Mediterranean-DASH intervention for Neurodegenerative Delay (MIND), which prevent exposure to these toxins. Such diets have been proven to decrease oxidative stress and inflammation, which are unfavorable for neuronal growth. Furthermore, they contribute to positive changes in the composition of the human gut microbiota and thus encourage interactions in the Gut-Brain Axis, reducing inflammation caused by pollutants. This review emphasizes a multi-professional approach with reference to nutritional activities that would lower the neurotoxic load in populations with a high level of exposure to pollutants. Future studies focusing on diet and environment association plans may help identify preventive measures aimed at enhancing current disease deceleration.
Collapse
Affiliation(s)
- Pratima Khandayataray
- Department of Biotechnology, Academy of Management and Information Technology, Utkal University, Bhubaneswar, Odisha 752057, India
| | - Meesala Krishna Murthy
- Department of Allied Health Sciences, Chitkara School of Health Sciences, Chitkara University, Rajpura, Punjab 140401, India.
| |
Collapse
|
5
|
Zhang J, Dong X, Pang Q, Zhang A. Irisin Alleviates Cognitive Impairment by Inhibiting AhR/NF- κB-NLRP3-Mediated Pyroptosis of Hippocampal Neurons in Chronic Kidney Disease. Mediators Inflamm 2024; 2024:2662362. [PMID: 39698584 PMCID: PMC11655147 DOI: 10.1155/mi/2662362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Accepted: 11/29/2024] [Indexed: 12/20/2024] Open
Abstract
Introduction: Cognitive impairment is a vital complication of chronic kidney disease (CKD). The effect of irisin on CKD-induced cognitive impairment remains unclear. In the present study, we aimed to investigate the role of Irisin in mitigating cognitive impairment and explore the underlying mechanisms in CKD. Methods: A CKD mice model was established by adenine. Cognitive function was assessed via the novel object recognition (NOR). Interleukin-1β (IL-1β) levels were measured by enzyme-linked immunosorbent assay (ELISA), while pyroptosis-related protein expression was analyzed using western blotting. Results: Our data showed an upregulation of cell pyroptosis in hippocampus tissues of CKD mice, accompanied by significant cognitive impairment. Pyroptosis and cognitive impairment was both improved by Irisin treatment in vivo. Additionally, irisin markedly downregulated pyroptosis levels through aryl hydrocarbon receptor (AhR)/NF-κB p65 signaling in HT-22 cells pretreated with indoxyl sulfate (IS). In vitro experiments further confirmed that pyroptosis was inhibited by AhR and NF-κB p65 inhibitors. Conclusions: We first demonstrated that irisin alleviated cognitive impairment by inhibiting AhR/NF-κB-NLRP3-mediated pyroptosis of hippocampal neurons in CKD. Overall, irisin may have the potential to serve as a critical antipyroptotic agent for improving CKD-induced cognitive impairment.
Collapse
Affiliation(s)
- Jialing Zhang
- Department of Nephrology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Xingtong Dong
- Department of Nephrology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Qi Pang
- Department of Nephrology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Aihua Zhang
- Department of Nephrology, Xuanwu Hospital, Capital Medical University, Beijing, China
- National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
6
|
Holme JA, Myhre O, Øvrevik J. Adverse neurodevelopment in children associated with prenatal exposure to fine particulate matter (PM 2.5) - Possible roles of polycyclic aromatic hydrocarbons (PAHs) and mechanisms involved. Reprod Toxicol 2024; 130:108718. [PMID: 39276806 DOI: 10.1016/j.reprotox.2024.108718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 09/11/2024] [Accepted: 09/11/2024] [Indexed: 09/17/2024]
Abstract
Prenatal exposure to ambient fine particles (PM2.5) and polycyclic aromatic hydrocarbons (PAHs) has been associated with adverse birth outcomes including neurodevelopmental effects with cognitive and/or behavioral implications in early childhood. As a background we first briefly summarize human studies on PM2.5 and PAHs associated with adverse birth outcomes and modified neurodevelopment. Next, we add more specific information from animal studies and in vitro studies and elucidate possible biological mechanisms. More specifically we focus on the potential role of PAHs attached to PM2.5 and explore whether effects of these compounds may arise from disturbance of placental function or more directly by interfering with neurodevelopmental processes in the fetal brain. Possible molecular initiating events (MIEs) include interactions with cellular receptors such as the aryl hydrocarbon receptor (AhR), beta-adrenergic receptors (βAR) and transient receptor potential (TRP)-channels resulting in altered gene expression. MIE linked to the binding of PAHs to cytochrome P450 (CYP) enzymes and formation of reactive electrophilic metabolites are likely less important. The experimental animal and in vitro studies support the epidemiological findings and suggest steps involved in mechanistic pathways explaining the associations. An overall evaluation of the doses/concentrations used in experimental studies combined with the mechanistic understanding further supports the hypothesis that prenatal PAHs exposure may cause adverse outcomes (AOs) linked to human neurodevelopment. Several MIEs will likely occur simultaneously in various cells/tissues involving several key events (KEs) which relative importance will depend on dose, time, tissue, genetics, other environmental factors, and neurodevelopmental endpoint in study.
Collapse
Affiliation(s)
- Jørn A Holme
- Department of Air quality and Noise, Division of Climate and Environmental Health, Norwegian Institute of Public Health, PO Box PO Box 222 Skøyen, Oslo 0213, Norway.
| | - Oddvar Myhre
- Department of Chemical Toxicology, Division of Climate and Environmental Health, Norwegian Institute of Public Health, PO Box 222 Skøyen, Oslo 0213, Norway
| | - Johan Øvrevik
- Department of Biosciences, Faculty of Mathematics and Natural Sciences, University of Oslo, PO Box 1066 Blindern, Oslo 0316, Norway; Division of Climate and Environmental Health, Norwegian Institute of Public Health, PO Box 222 Skøyen, Oslo 0213, Norway
| |
Collapse
|
7
|
Meyer DN, Silva I, Vo B, Paquette A, Blount JR, George SE, Gonzalez G, Cavaneau E, Khalaf A, Petriv AM, Wu CC, Haimbaugh A, Baker TR. Juvenile exposure to low-level 2,3,7,8-tetrachlorodibenzo- p-dioxin (TCDD) alters behavior and longitudinal morphometrics in zebrafish and F 1 offspring. J Dev Orig Health Dis 2024; 15:e22. [PMID: 39397699 DOI: 10.1017/s2040174424000229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/15/2024]
Abstract
Exposure to 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), an environmental endocrine disruptor and model AhR agonist, is linked to skeletal abnormalities, cardiac edema, stunted growth rate, altered metabolism, and neurobehavioral deficits. We have previously reported transgenerational reproductive outcomes of developmental TCDD exposure in adult zebrafish (Danio rerio), an NIH-validated model for developmental and generational toxicology. Using the same paradigm of sublethal TCDD exposure (50 pg/ml) at both 3 and 7 weeks post fertilization (wpf), we investigated several novel endpoints, including longitudinal morphometrics and anxiety-linked behavior, in fish exposed as juveniles. We also assessed developmental abnormalities and neurobehavior in their F1 larval offspring. TCDD exposure induced timepoint-dependent decreases in several craniofacial and trunk morphometrics across juvenile development. In early adulthood, however, only exposed males underwent a transient period of compensatory growth, ending between 7 and 12 months post fertilization (mpf). At 12 mpf, exposed adult fish of both sexes displayed increased exploratory behaviors in a novel tank test. The F1 offspring of parents exposed at both 3 and 7 wpf were hyperactive, but neurobehavioral outcomes diverged depending on parental exposure window. F1 exposure-lineage larvae had increased rates of edema and skeletal abnormalities, but fewer unhatched larvae compared to controls. Parent- and timepoint-specific effects of exposure on abnormality rate were also evaluated; these outcomes were considerably less severe. Our novel behavioral findings expand current knowledge of the long-term and intergenerational consequences of early-life TCDD exposure in a zebrafish model, in addition to delineating minor longitudinal morphometric changes in exposed fish and abnormalities in larval offspring.
Collapse
Affiliation(s)
- Danielle N Meyer
- Department of Environmental and Global Health, University of Florida, Gainesville, FL, USA
- Department of Pharmacology, Wayne State University, Detroit, MI, USA
- Institute of Environmental Health Sciences, Wayne State University, Detroit, MI, USA
| | - Isabela Silva
- Department of Environmental and Global Health, University of Florida, Gainesville, FL, USA
| | - Brianna Vo
- Department of Environmental and Global Health, University of Florida, Gainesville, FL, USA
| | - Amelia Paquette
- Department of Environmental and Global Health, University of Florida, Gainesville, FL, USA
| | - Jessica R Blount
- Institute of Environmental Health Sciences, Wayne State University, Detroit, MI, USA
| | - Serena E George
- School of Veterinary Medicine, University of Madison-Wisconsin, Madison, WI, USA
| | - Gabrielle Gonzalez
- Department of Environmental and Global Health, University of Florida, Gainesville, FL, USA
| | - Emma Cavaneau
- Department of Environmental and Global Health, University of Florida, Gainesville, FL, USA
| | - Aicha Khalaf
- Institute of Environmental Health Sciences, Wayne State University, Detroit, MI, USA
| | - Anna-Maria Petriv
- Institute of Environmental Health Sciences, Wayne State University, Detroit, MI, USA
| | - Chia-Chen Wu
- Department of Environmental and Global Health, University of Florida, Gainesville, FL, USA
- Institute of Environmental Health Sciences, Wayne State University, Detroit, MI, USA
| | - Alex Haimbaugh
- Department of Pharmacology, Wayne State University, Detroit, MI, USA
- Institute of Environmental Health Sciences, Wayne State University, Detroit, MI, USA
| | - Tracie R Baker
- Department of Environmental and Global Health, University of Florida, Gainesville, FL, USA
- Department of Pharmacology, Wayne State University, Detroit, MI, USA
- Institute of Environmental Health Sciences, Wayne State University, Detroit, MI, USA
| |
Collapse
|
8
|
Martins-Macedo J, Araújo B, Anjo SI, Silveira-Rosa T, Patrício P, Alves ND, Silva JM, Teixeira FG, Manadas B, Rodrigues AJ, Lepore AC, Salgado AJ, Gomes ED, Pinto L. Glial-restricted precursors stimulate endogenous cytogenesis and effectively recover emotional deficits in a model of cytogenesis ablation. Mol Psychiatry 2024; 29:2185-2198. [PMID: 38454085 PMCID: PMC11632613 DOI: 10.1038/s41380-024-02490-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 02/08/2024] [Accepted: 02/15/2024] [Indexed: 03/09/2024]
Abstract
Adult cytogenesis, the continuous generation of newly-born neurons (neurogenesis) and glial cells (gliogenesis) throughout life, is highly impaired in several neuropsychiatric disorders, such as Major Depressive Disorder (MDD), impacting negatively on cognitive and emotional domains. Despite playing a critical role in brain homeostasis, the importance of gliogenesis has been overlooked, both in healthy and diseased states. To examine the role of newly formed glia, we transplanted Glial Restricted Precursors (GRPs) into the adult hippocampal dentate gyrus (DG), or injected their secreted factors (secretome), into a previously validated transgenic GFAP-tk rat line, in which cytogenesis is transiently compromised. We explored the long-term effects of both treatments on physiological and behavioral outcomes. Grafted GRPs reversed anxiety-like deficits and demonstrated an antidepressant-like effect, while the secretome promoted recovery of only anxiety-like behavior. Furthermore, GRPs elicited a recovery of neurogenic and gliogenic levels in the ventral DG, highlighting the unique involvement of these cells in the regulation of brain cytogenesis. Both GRPs and their secretome induced significant alterations in the DG proteome, directly influencing proteins and pathways related to cytogenesis, regulation of neural plasticity and neuronal development. With this work, we demonstrate a valuable and specific contribution of glial progenitors to normalizing gliogenic levels, rescuing neurogenesis and, importantly, promoting recovery of emotional deficits characteristic of disorders such as MDD.
Collapse
Affiliation(s)
- Joana Martins-Macedo
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
- Center for Translational Health and Medical Biotechnology Research (TBIO), School of Health (ESS), Polytechnic of Porto, Porto, Portugal
- I3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135, Porto, Portugal
| | - Bruna Araújo
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
- Center for Translational Health and Medical Biotechnology Research (TBIO), School of Health (ESS), Polytechnic of Porto, Porto, Portugal
- I3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135, Porto, Portugal
| | - Sandra I Anjo
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- Institute for Interdisciplinary Research, University of Coimbra (IIIUC), Coimbra, Portugal
- Centre for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal
| | - Tiago Silveira-Rosa
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Patrícia Patrício
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Nuno Dinis Alves
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Joana M Silva
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Fábio G Teixeira
- Center for Translational Health and Medical Biotechnology Research (TBIO), School of Health (ESS), Polytechnic of Porto, Porto, Portugal
- I3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135, Porto, Portugal
| | - Bruno Manadas
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- Institute for Interdisciplinary Research, University of Coimbra (IIIUC), Coimbra, Portugal
- Centre for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal
| | - Ana J Rodrigues
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Angelo C Lepore
- Department of Neuroscience, Vickie and Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
| | - António J Salgado
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Eduardo D Gomes
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
- Center for Translational Health and Medical Biotechnology Research (TBIO), School of Health (ESS), Polytechnic of Porto, Porto, Portugal
- I3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135, Porto, Portugal
| | - Luísa Pinto
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal.
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal.
| |
Collapse
|
9
|
Bajic D, Wiens F, Wintergerst E, Deyaert S, Baudot A, den Abbeele PV. HMOs Impact the Gut Microbiome of Children and Adults Starting from Low Predicted Daily Doses. Metabolites 2024; 14:239. [PMID: 38668367 PMCID: PMC11052010 DOI: 10.3390/metabo14040239] [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: 02/29/2024] [Revised: 03/29/2024] [Accepted: 04/16/2024] [Indexed: 04/28/2024] Open
Abstract
Recent studies suggest that the dietary intake of human milk oligosaccharides (HMOs) provides health benefits from infancy up to adulthood. Thus far, beneficial changes in the adult gut microbiome have been observed at oral doses of 5-20 g/day of HMOs. Efficacy of lower doses has rarely been tested. We assessed four HMO molecular species-2'Fucosyllactose (2'FL), Lacto-N-neotetraose (LNnT), 3'Sialyllactose (3'SL), and 6'Sialyllactose (6'SL)-at predicted doses from 0.3 to 5 g/day for 6-year-old children and adults (n = 6 each), using ex vivo SIFR® technology (Cryptobiotix, Ghent, Belgium). This technology employing bioreactor fermentation on fecal samples enables us to investigate microbial fermentation products that are intractable in vivo given their rapid absorption/consumption in the human gut. We found that HMOs significantly increased short-chain fatty acids (SCFAs), acetate, propionate (in children/adults), and butyrate (in adults) from predicted doses of 0.3-0.5 g/day onwards, with stronger effects as dosing increased. The fermentation of 6'SL had the greatest effect on propionate, LNnT most strongly increased butyrate, and 2'FL and 3'SL most strongly increased acetate. An untargeted metabolomic analysis revealed that HMOs enhanced immune-related metabolites beyond SCFAs, such as aromatic lactic acids (indole-3-lactic acid/3-phenyllactic acid) and 2-hydroxyisocaproic acid, as well as gut-brain-axis-related metabolites (γ-aminobutyric acid/3-hydroxybutyric acid/acetylcholine) and vitamins. The effects of low doses of HMOs potentially originate from the highly specific stimulation of keystone species belonging to, for example, the Bifidobacteriaceae family, which had already significantly increased at doses of only 0.5 g/day LNnT (adults) and 1 g/day 2'FL (children/adults).
Collapse
Affiliation(s)
- Danica Bajic
- DSM Nutritional Products Ltd., Wurmisweg 576, 4303 Kaiseraugst, Switzerland
| | - Frank Wiens
- DSM Nutritional Products Ltd., Wurmisweg 576, 4303 Kaiseraugst, Switzerland
| | - Eva Wintergerst
- DSM Nutritional Products Ltd., Wurmisweg 576, 4303 Kaiseraugst, Switzerland
| | - Stef Deyaert
- Cryptobiotix SA, Technologiepark-Zwijnaarde 82, 9052 Ghent, Belgium
| | - Aurélien Baudot
- Cryptobiotix SA, Technologiepark-Zwijnaarde 82, 9052 Ghent, Belgium
| | | |
Collapse
|
10
|
Cintrón-Rivera LG, Burns N, Patel R, Plavicki JS. Exposure to the aryl hydrocarbon receptor agonist dioxin disrupts formation of the muscle, nerves, and vasculature in the developing jaw. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2023; 337:122499. [PMID: 37660771 DOI: 10.1016/j.envpol.2023.122499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 08/14/2023] [Accepted: 08/31/2023] [Indexed: 09/05/2023]
Abstract
Human exposure to environmental pollutants can disrupt embryonic development and impact juvenile and adult health outcomes by adversely affecting cell and organ function. Notwithstanding, environmental contamination continues to increase due to industrial development, insufficient regulations, and the mobilization of pollutants as a result of extreme weather events. Dioxins are a class of structurally related persistent organic pollutants that are highly toxic, carcinogenic, and teratogenic. 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) is the most potent dioxin compound and has been shown to induce toxic effects in developing organisms by activating the aryl hydrocarbon receptor (AHR), a ligand activated transcription factor targeted by multiple persistent organic pollutants. Contaminant-induced AHR activation results in malformations of the craniofacial cartilages and neurocranium; however, the mechanisms mediating these phenotypes are not well understood. In this study, we utilized the optically transparent zebrafish model to elucidate novel cellular targets and potential transcriptional targets underlying TCDD-induced craniofacial malformations. To this end, we exposed zebrafish embryos at 4 h post fertilization to TCDD and employed a mixed-methods approach utilizing immunohistochemistry staining, transgenic reporter lines, fixed and in vivo confocal imaging, and timelapse microscopy to determine the targets mediating TCDD-induced craniofacial phenotypes. Our data indicate that embryonic TCDD exposure reduced jaw and pharyngeal arch Sox10+ chondrocytes and Tcf21+ pharyngeal mesoderm progenitors. Exposure to TCDD correspondingly led to a reduction in collagen type II deposition in Sox10+ domains. Embryonic TCDD exposure impaired development of tissues derived from or guided by Tcf21+ progenitors, namely: nerves, muscle, and vasculature. Specifically, TCDD exposure disrupted development of the hyoid and mandibular arch muscles, decreased neural innervation of the jaw, resulted in compression of cranial nerves V and VII, and led to jaw vasculature malformations. Collectively, these findings reveal novel structural targets and potential transcriptional targets of TCDD-induced toxicity, showcasing how contaminant exposures lead to congenital craniofacial malformations.
Collapse
Affiliation(s)
- Layra G Cintrón-Rivera
- Department of Pathology and Laboratory Medicine, Brown University, 70 Ship St, Providence, RI, 02903, USA
| | - Nicole Burns
- Department of Pathology and Laboratory Medicine, Brown University, 70 Ship St, Providence, RI, 02903, USA
| | - Ratna Patel
- Department of Pathology and Laboratory Medicine, Brown University, 70 Ship St, Providence, RI, 02903, USA
| | - Jessica S Plavicki
- Department of Pathology and Laboratory Medicine, Brown University, 70 Ship St, Providence, RI, 02903, USA.
| |
Collapse
|
11
|
Tumova S, Dolezel D, Jindra M. Conserved and Unique Roles of bHLH-PAS Transcription Factors in Insects - From Clock to Hormone Reception. J Mol Biol 2023; 436:168332. [PMID: 39491146 DOI: 10.1016/j.jmb.2023.168332] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 10/20/2023] [Accepted: 10/23/2023] [Indexed: 11/05/2024]
Abstract
A dozen bHLH-PAS transcription factors have evolved since the dawn of the animal kingdom; nine of them have mutual orthologs between arthropods and vertebrates. These proteins are master regulators in a range of developmental processes from organogenesis, nervous system formation and functioning, to cell fate decisions defining identity of limbs or photoreceptors for color vision. Among the functionally best conserved are bHLH-PAS proteins acting in the animal circadian clock. On the other side of the spectrum are fundamental physiological mechanisms such as those underlying xenobiotic detoxification, oxygen homeostasis, and metabolic adaptation to hypoxia, infection or tumor progression. Predictably, malfunctioning of bHLH-PAS regulators leads to pathologies. Performance of the individual bHLH-PAS proteins is modulated at multiple levels including dimerization and other protein-protein interactions, proteasomal degradation, and by binding low-molecular weight ligands. Despite the vast evolutionary gap dividing arthropods and vertebrates, and the differences in their anatomy, many functions of orthologous bHLH-PAS proteins are remarkably similar, including at the molecular level. Our phylogenetic analysis shows that one bHLH-PAS protein type has been lost during vertebrate evolution. This protein has a unique function as a receptor of the sesquiterpenoid juvenile hormones of insects and crustaceans. Although some other bHLH-PAS proteins are regulated by binding small molecules, the juvenile hormone receptor presents an unprecedented case, since all other non-peptide animal hormones activate members of the nuclear receptor family. The purpose of this review is to compare and highlight parallels and differences in functioning of bHLH-PAS proteins between insects and vertebrates.
Collapse
Affiliation(s)
- Sarka Tumova
- Institute of Entomology, Biology Center of the Czech Academy of Sciences, Ceske Budejovice 37005, Czech Republic
| | - David Dolezel
- Institute of Entomology, Biology Center of the Czech Academy of Sciences, Ceske Budejovice 37005, Czech Republic
| | - Marek Jindra
- Institute of Entomology, Biology Center of the Czech Academy of Sciences, Ceske Budejovice 37005, Czech Republic.
| |
Collapse
|
12
|
Alluli A, Rijnbout St James W, Eidelman DH, Baglole CJ. Dynamic relationship between the aryl hydrocarbon receptor and long noncoding RNA balances cellular and toxicological responses. Biochem Pharmacol 2023; 216:115745. [PMID: 37597813 DOI: 10.1016/j.bcp.2023.115745] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 08/10/2023] [Accepted: 08/11/2023] [Indexed: 08/21/2023]
Abstract
The aryl hydrocarbon receptor (AhR) is a cytosolic transcription factor activated by endogenous ligands and xenobiotic chemicals. Once the AhR is activated, it translocates to the nucleus, dimerizes with the AhR nuclear translator (ARNT) and binds to xenobiotic response elements (XRE) to promote gene transcription, notably the cytochrome P450 CYP1A1. The AhR not only mediates the toxic effects of environmental chemicals, but also has numerous putative physiological functions. This dichotomy in AhR biology may be related to reciprocal regulation of long non-coding RNA (lncRNA). lncRNA are defined as transcripts more than 200 nucleotides in length that do not encode a protein but are implicated in many physiological processes such as cell differentiation, cell proliferation, and apoptosis. lncRNA are also linked to disease pathogenesis, particularly the development of cancer. Recent studies have revealed that AhR activation by environmental chemicals affects the expression and function of lncRNA. In this article, we provide an overview of AhR signaling pathways activated by diverse ligands and highlight key differences in the putative biological versus toxicological response of AhR activation. We also detail the functions of lncRNA and provide current data on their regulation by the AhR. Finally, we outline how overlap in function between AhR and lncRNA may be one way in which AhR can be both a regulator of endogenous functions but also a mediator of toxicological responses to environmental chemicals. Overall, more research is still needed to fully understand the dynamic interplay between the AhR and lncRNA.
Collapse
Affiliation(s)
- Aeshah Alluli
- Meakins-Christie Laboratories, McGill University, Canada; Translational Research in Respiratory Diseases Program at the Research Institute of the McGill University Health Centre, Canada; Department of Pathology, McGill University, Canada
| | - Willem Rijnbout St James
- Meakins-Christie Laboratories, McGill University, Canada; Translational Research in Respiratory Diseases Program at the Research Institute of the McGill University Health Centre, Canada; Department of Pathology, McGill University, Canada
| | - David H Eidelman
- Meakins-Christie Laboratories, McGill University, Canada; Department of Medicine, McGill University, Canada
| | - Carolyn J Baglole
- Meakins-Christie Laboratories, McGill University, Canada; Translational Research in Respiratory Diseases Program at the Research Institute of the McGill University Health Centre, Canada; Department of Pathology, McGill University, Canada; Department of Medicine, McGill University, Canada; Department of Pharmacology and Therapeutics, McGill University, Canada.
| |
Collapse
|
13
|
Sun N, Wang H, Wang XY, Yu Q, Han JY, Huang Y, Zhou WX. Deletion of AhR attenuates fear memory leaving other types of memory intact. Behav Brain Res 2023; 451:114505. [PMID: 37217138 DOI: 10.1016/j.bbr.2023.114505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Revised: 05/11/2023] [Accepted: 05/19/2023] [Indexed: 05/24/2023]
Abstract
The aryl hydrocarbon receptor (AhR), a classic "environmental sensor", has been found to play an important role in cognitive and emotional function. Recent studies showed AhR deletion led to an attenuated fear memory, providing a potential target against fear memory, whether it is the consequence of attenuated sense of fear or memory ability deficit or both is unclear. Here this study aims to work this out. The freezing time in contextual fear conditioning (CFC) reduced significantly in AhR knockout mice, indicating an attenuated fear memory. Hot plate test and acoustic startle reflex showed that AhR knockout did not change the pain threshold and hearing, excluded the possibility of sensory impairments. Results from NORT, MWM and SBT showed that deletion of AhR had little effects on other types of memory. But the anxiety-like behaviors reduced both in naïve or suffered (tested after CFC) AhR knockout mice, showing that AhR-deficient mice have a reduced basal and stressful emotional response. The basal low-frequency to high-frequency (LF/HF) ratio of the AhR knockout mice was significantly lower than that of the control group, indicating lower sympathetic excitability in the basal state, suggesting a low level of basal stress in the knockout mice. Before and after CFC, the LF/HF ratio of AhR-KO mice tended to be significantly lower than that of WT mice, and their heart rate was significantly lower; and the AhR-KO mice also has a decreased serum corticosterone level after CFC, suggesting a reduced stress response in AhR knockout mice. Altogether, the basal stress level and stress response were significant reduced in AhR knockout mice, which might contribute to the attenuated fear memory with little impairment on other types of memory, suggesting AhR as a "psychologic sensor" additional to "environmental sensor".
Collapse
Affiliation(s)
- Na Sun
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Hao Wang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Xin-Yue Wang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Qi Yu
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Jin-Yuan Han
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Yan Huang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China.
| | - Wen-Xia Zhou
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China.
| |
Collapse
|
14
|
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
|
15
|
Pinto L, Macedo J, Araújo B, Anjo S, Silveira-Rosa T, Patrício P, Teixeira F, Manadas B, Rodrigues AJ, Lepore A, Salgado A, Gomes E. Glial-Restricted Precursors stimulate endogenous cytogenesis and effectively recover emotional deficits in a model of cytogenesis ablation. RESEARCH SQUARE 2023:rs.3.rs-2747462. [PMID: 37034743 PMCID: PMC10081440 DOI: 10.21203/rs.3.rs-2747462/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/30/2023]
Abstract
Adult cytogenesis, the continuous generation of newly-born neurons (neurogenesis) and glial cells (gliogenesis) throughout life, is highly impaired in several neuropsychiatric disorders, such as Major Depressive Disorder (MDD), impacting negatively on cognitive and emotional domains. Despite playing a critical role in brain homeostasis, the importance of gliogenesis has been overlooked, both in healthy and diseased states. To examine the role of newly formed glia, we transplanted Glial Restricted Precursors (GRPs) into the adult hippocampal dentate gyrus (DG), or injected their secreted factors (secretome), into a previously validated transgenic GFAP-tk rat line, in which cytogenesis is transiently compromised. We explored the long-term effects of both treatments on physiological and behavioral outcomes. Grafted GRPs reversed anxiety-like and depressive-like deficits, while the secretome promoted recovery of only anxiety-like behavior. Furthermore, GRPs elicited a recovery of neurogenic and gliogenic levels in the ventral DG, highlighting the unique involvement of these cells in the regulation of brain cytogenesis. Both GRPs and their secretome induced significant alterations in the DG proteome, directly influencing proteins and pathways related to cytogenesis, regulation of neural plasticity and neuronal development. With this work, we demonstrate a valuable and specific contribution of glial progenitors to normalizing gliogenic levels, rescueing neurogenesis and, importantly, promoting recovery of emotional deficits characteristic of disorders such as MDD.
Collapse
Affiliation(s)
| | | | | | - Sandra Anjo
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra
| | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Bajic D, Wiens F, Wintergerst E, Deyaert S, Baudot A, Van den Abbeele P. HMOs Exert Marked Bifidogenic Effects on Children's Gut Microbiota Ex Vivo, Due to Age-Related Bifidobacterium Species Composition. Nutrients 2023; 15:1701. [PMID: 37049541 PMCID: PMC10097135 DOI: 10.3390/nu15071701] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Revised: 03/25/2023] [Accepted: 03/28/2023] [Indexed: 04/03/2023] Open
Abstract
Prebiotics are substrates that are selectively utilized by host microorganisms, thus conferring a health benefit. There is a growing awareness that interpersonal and age-dependent differences in gut microbiota composition impact prebiotic effects. Due to the interest in using human milk oligosaccharides (HMOs) beyond infancy, this study evaluated how HMOs [2'Fucosyllactose (2'FL), Lacto-N-neotetraose (LNnT), 3'Sialyllactose (3'SL), 6'Sialyllactose (6'SL)] and blends thereof affect the microbiota of 6-year-old children (n = 6) and adults (n = 6), compared to prebiotics inulin (IN) and fructooligosaccharides (FOS). The ex vivo SIFR® technology was used, given its demonstrated predictivity in clinical findings. First, HMOs and HMO blends seemed to maintain a higher α-diversity compared to FOS/IN. Further, while 2'FL/LNnT were bifidogenic for both age groups, 3'SL/6'SL and FOS/IN were exclusively bifidogenic for children and adults, respectively. This originated from age-related differences in microbiota composition because while 3'SL/6'SL stimulated B. pseudocatenulatum (abundant in children), FOS/IN enhanced B. adolescentis (abundant in adults). Moreover, all treatments significantly increased acetate, propionate and butyrate (only in adults) with product- and age-dependent differences. Among the HMOs, 6'SL specifically stimulated propionate (linked to Bacteroides fragilis in children and Phocaeicola massiliensis in adults), while LNnT stimulated butyrate (linked to Anaerobutyricum hallii in adults). Indole-3-lactic acid and 3-phenyllactic acid (linked to immune health) and gamma-aminobutyric acid (linked to gut-brain axis) were most profoundly stimulated by 2'FL and HMO blends in both children and adults, correlating with specific Bifidobacteriaceae. Finally, 2'FL/LNnT increased melatonin in children, while 3'SL remarkably increased folic acid in adults. Overall, age-dependent differences in microbiota composition greatly impacted prebiotic outcomes, advocating for the development of age-specific nutritional supplements. HMOs were shown to be promising modulators in the adult, and particularly the children's microbiota. The observed HMO-specific effects, likely originating from their structural heterogeneity, suggest that blends of different HMOs could maximize treatment effects.
Collapse
Affiliation(s)
- Danica Bajic
- Glycom A/S-DSM Nutritional Products Ltd., Kogle Allé 4, 2970 Hørsholm, Denmark
| | - Frank Wiens
- DSM Nutritional Products Ltd., Wurmisweg 576, 4303 Kaiseraugst, Switzerland
| | - Eva Wintergerst
- DSM Nutritional Products Ltd., Wurmisweg 576, 4303 Kaiseraugst, Switzerland
| | - Stef Deyaert
- Cryptobiotix SA, Technologiepark-Zwijnaarde 82, 9052 Ghent, Belgium
| | - Aurélien Baudot
- Cryptobiotix SA, Technologiepark-Zwijnaarde 82, 9052 Ghent, Belgium
| | | |
Collapse
|
17
|
Wang Y, Sun J, Zhu K, Wang D, Zhao X, Zhang H, Wu S, Wang Y, Wang J. Microglial aryl hydrocarbon receptor enhances phagocytic function via SYK and promotes remyelination in the cuprizone mouse model of demyelination. J Neuroinflammation 2023; 20:83. [PMID: 36966295 PMCID: PMC10040134 DOI: 10.1186/s12974-023-02764-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 03/11/2023] [Indexed: 03/27/2023] Open
Abstract
Multiple sclerosis (MS) is an inflammatory-mediated demyelinating disease of the central nervous system (CNS). Although studies have demonstrated that microglia facilitate remyelination in demyelinating diseases, the underlying mechanisms are still not fully characterized. We found that aryl hydrocarbon receptor (AhR), an environment sensor, was upregulated within the corpus callosum in the cuprizone model of CNS demyelination, and upregulated AhR was mainly confined to microglia. Deletion of AhR in adult microglia inhibited efficient remyelination. Transcriptome analysis using RNA-seq revealed that AhR-deficient microglia displayed impaired gene expression signatures associated with lysosome and phagocytotic pathways. Furthermore, AhR-deficient microglia showed impaired clearance of myelin debris and defected phagocytic capacity. Further investigation of target genes of AhR revealed that spleen tyrosine kinase (SYK) is the downstream effector of AhR and mediated the phagocytic capacity of microglia. Additionally, AhR deficiency in microglia aggravated CNS inflammation during demyelination. Altogether, our study highlights an essential role for AhR in microglial phagocytic function and suggests the therapeutic potential of AhR in demyelinating diseases.
Collapse
Affiliation(s)
- Yumeng Wang
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Science, Institutes of Integrative Medicine, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jingxian Sun
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Science, Institutes of Integrative Medicine, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Keying Zhu
- Department of Clinical Neuroscience, Karolinska Institute, Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Danjie Wang
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Science, Institutes of Integrative Medicine, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xiaoqiang Zhao
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Science, Institutes of Integrative Medicine, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Hongyu Zhang
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Science, Institutes of Integrative Medicine, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Shuai Wu
- Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yanqing Wang
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Science, Institutes of Integrative Medicine, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Shanghai Medical College, Fudan University, Shanghai, China.
| | - Jun Wang
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Science, Institutes of Integrative Medicine, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Shanghai Medical College, Fudan University, Shanghai, China.
| |
Collapse
|
18
|
Vijay A, Boyle NR, Kumar SM, Perdew GH, Srinivasan S, Patterson AD. Aryl hydrocarbon receptor activation affects nitrergic neuronal survival and delays intestinal motility in mice. Toxicol Sci 2023; 192:117-128. [PMID: 36782369 PMCID: PMC10025877 DOI: 10.1093/toxsci/kfad014] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/15/2023] Open
Abstract
Despite progress describing the effects of persistent organic pollutants (POPs) on the central nervous system, the effect of POPs on enteric nervous system (ENS) function remains underexplored. We studied the effects of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), a POP, and a potent aryl hydrocarbon receptor (AHR) ligand, on the ENS and intestinal motility in mice. C57Bl/6J mice treated with TCDD (2.4 µg/kg body weight) for 8 weeks (once per week) exhibited significant delay in intestinal motility as shown by reduced stool frequency, prolonged intestinal transit time, and a persistence of dye in the jejunum compared to control mice with maximal dye retention in the ileum. TCDD significantly increased Cyp1a1 expression, an AHR target gene, and reduced the total number of neurons and affected nitrergic neurons in cells isolated from WT mice, but not Ahr-/- mice. In immortalized fetal enteric neuronal cells, TCDD-induced nuclear translocation of AHR as well as increased Cyp1a1 expression. AHR activation did not affect neuronal proliferation. However, AHR activation resulted in enteric neuronal toxicity, specifically, nitrergic neurons. Our results demonstrate that TCDD adversely affects nitrergic neurons and thereby contributes to delayed intestinal motility. These findings suggest that AHR signaling in the ENS may play a role in modulating TCDD-induced gastrointestinal pathophysiology.
Collapse
Affiliation(s)
- Anitha Vijay
- Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, Pennsylvania 16802, USA
| | - Nina R Boyle
- Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, Pennsylvania 16802, USA
| | - Supriya M Kumar
- Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, Pennsylvania 16802, USA
| | - Gary H Perdew
- Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, Pennsylvania 16802, USA
| | - Shanthi Srinivasan
- Department of Digestive Diseases, Emory University School of Medicine, Atlanta, Georgia, USA
- Atlanta VA Medical Center, Decatur, Georgia, USA
| | - Andrew D Patterson
- Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, Pennsylvania 16802, USA
| |
Collapse
|
19
|
The impact of amino acid metabolism on adult neurogenesis. Biochem Soc Trans 2023; 51:233-244. [PMID: 36606681 DOI: 10.1042/bst20220762] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Revised: 12/02/2022] [Accepted: 12/05/2022] [Indexed: 01/07/2023]
Abstract
Adult neurogenesis is a multistage process during which newborn neurons are generated through the activation and proliferation of neural stem cells (NSCs) and integrated into existing neural networks. Impaired adult neurogenesis has been observed in various neurological and psychiatric disorders, suggesting its critical role in cognitive function, brain homeostasis, and neural repair. Over the past decades, mounting evidence has identified a strong association between metabolic status and adult neurogenesis. Here, we aim to summarize how amino acids and their neuroactive metabolites affect adult neurogenesis. Furthermore, we discuss the causal link between amino acid metabolism, adult neurogenesis, and neurological diseases. Finally, we propose that systematic elucidation of how amino acid metabolism regulates adult neurogenesis has profound implications not only for understanding the biological underpinnings of brain development and neurological diseases, but also for providing potential therapeutic strategies to intervene in disease progression.
Collapse
|
20
|
Silva-Parra J, Sandu C, Felder-Schmittbuhl MP, Hernández-Kelly LC, Ortega A. Aryl Hydrocarbon Receptor in Glia Cells: A Plausible Glutamatergic Neurotransmission Orchestrator. Neurotox Res 2023; 41:103-117. [PMID: 36607593 DOI: 10.1007/s12640-022-00623-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 11/23/2022] [Accepted: 12/15/2022] [Indexed: 01/07/2023]
Abstract
Glutamate is the major excitatory amino acid in the vertebrate brain. Glutamatergic signaling is involved in most of the central nervous system functions. Its main components, namely receptors, ion channels, and transporters, are tightly regulated at the transcriptional, translational, and post-translational levels through a diverse array of extracellular signals, such as food, light, and neuroactive molecules. An exquisite and well-coordinated glial/neuronal bidirectional communication is required for proper excitatory amino acid signal transactions. Biochemical shuttles such as the glutamate/glutamine and the astrocyte-neuronal lactate represent the fundamental involvement of glial cells in glutamatergic transmission. In fact, the disruption of any of these coordinated biochemical intercellular cascades leads to an excitotoxic insult that underlies some aspects of most of the neurodegenerative diseases characterized thus far. In this contribution, we provide a comprehensive summary of the involvement of the Aryl hydrocarbon receptor, a ligand-dependent transcription factor in the gene expression regulation of glial glutamate transporters. These receptors might serve as potential targets for the development of novel strategies for the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Janisse Silva-Parra
- Departamento de Toxicología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Av. IPN 2508, San Pedro Zacatenco, 07360, CDMX, México
| | - Cristina Sandu
- Centre National de la Recherche Scientifique, Institut des Neurosciences Cellulaires et Intégratives, Université de Strasbourg, Strasbourg, France
| | - Marie-Paule Felder-Schmittbuhl
- Centre National de la Recherche Scientifique, Institut des Neurosciences Cellulaires et Intégratives, Université de Strasbourg, Strasbourg, France
| | - Luisa C Hernández-Kelly
- Departamento de Toxicología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Av. IPN 2508, San Pedro Zacatenco, 07360, CDMX, México
| | - Arturo Ortega
- Departamento de Toxicología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Av. IPN 2508, San Pedro Zacatenco, 07360, CDMX, México.
| |
Collapse
|
21
|
Kossack ME, Manz KE, Martin NR, Pennell KD, Plavicki J. Environmentally relevant uptake, elimination, and metabolic changes following early embryonic exposure to 2,3,7,8-tetrachlorodibenzo-p-dioxin in zebrafish. CHEMOSPHERE 2023; 310:136723. [PMID: 36241106 PMCID: PMC9835613 DOI: 10.1016/j.chemosphere.2022.136723] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 08/16/2022] [Accepted: 09/30/2022] [Indexed: 06/03/2023]
Abstract
Dioxin and dioxin-like compounds are ubiquitous environmental contaminants that induce toxicity by binding to the aryl hydrocarbon receptor (AHR), a ligand activated transcription factor. The zebrafish model has been used to define the developmental toxicity observed following exposure to exogenous AHR ligands such as the potent agonist 2,3,7,8-tetrachlorodibenzo-p-dioxin (dioxin, TCDD). While the model has successfully identified cellular targets of TCDD and molecular mechanisms mediating TCDD-induced phenotypes, fundamental information such as the body burden produced by standard exposure models is still unknown. We performed targeted gas chromatography (GC) high-resolution mass spectrometry (HRMS) in tandem with non-targeted liquid chromatography (LC) HRMS to quantify TCDD uptake, model the elimination dynamics of TCDD, and determine how TCDD exposure affects the zebrafish metabolome. We found that 50 ppt, 10 ppb, and 1 ppb waterborne exposures to TCDD during early embryogenesis produced environmentally relevant body burdens: 38 ± 4.34, 26.6 ± 1.2, and 8.53 ± 0.341 pg/embryo, respectively, at 24 hours post fertilization. TCDD exposure was associated with the dysregulation of metabolic pathways that are associated with the AHR signaling pathway as well as pathways shown to be affected in mammals following TCDD exposure. In addition, we discovered that TCDD exposure affected several metabolic pathways that are critical for brain development and function including glutamate metabolism, chondroitin sulfate biosynthesis, and tyrosine metabolism. Together, these data demonstrate that existing exposure methods produce environmentally relevant body burdens of TCDD in zebrafish and provide insight into the biochemical pathways impacted by toxicant-induced AHR activation.
Collapse
Affiliation(s)
- Michelle E Kossack
- Department of Pathology and Laboratory Medicine, Brown University, 70 Ship St, Providence, RI, 02903, USA
| | - Katherine E Manz
- School of Engineering, Brown University, 184 Hope St, Box D, Providence, RI, 02903, USA
| | - Nathan R Martin
- Department of Pathology and Laboratory Medicine, Brown University, 70 Ship St, Providence, RI, 02903, USA
| | - Kurt D Pennell
- School of Engineering, Brown University, 184 Hope St, Box D, Providence, RI, 02903, USA
| | - Jessica Plavicki
- Department of Pathology and Laboratory Medicine, Brown University, 70 Ship St, Providence, RI, 02903, USA.
| |
Collapse
|
22
|
Hossain MM, Belkadi A, Zhou X, DiCicco-Bloom E. Exposure to deltamethrin at the NOAEL causes ER stress and disruption of hippocampal neurogenesis in adult mice. Neurotoxicology 2022; 93:233-243. [DOI: 10.1016/j.neuro.2022.10.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 09/29/2022] [Accepted: 10/07/2022] [Indexed: 11/15/2022]
|
23
|
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
|
24
|
Salminen A. Aryl hydrocarbon receptor (AhR) reveals evidence of antagonistic pleiotropy in the regulation of the aging process. Cell Mol Life Sci 2022; 79:489. [PMID: 35987825 PMCID: PMC9392714 DOI: 10.1007/s00018-022-04520-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 07/14/2022] [Accepted: 08/08/2022] [Indexed: 11/24/2022]
Abstract
The antagonistic pleiotropy hypothesis is a well-known evolutionary theory to explain the aging process. It proposes that while a particular gene may possess beneficial effects during development, it can exert deleterious properties in the aging process. The aryl hydrocarbon receptor (AhR) has a significant role during embryogenesis, but later in life, it promotes several age-related degenerative processes. For instance, AhR factor (i) controls the pluripotency of stem cells and the stemness of cancer stem cells, (ii) it enhances the differentiation of embryonal stem cells, especially AhR signaling modulates the differentiation of hematopoietic stem cells and progenitor cells, (iii) it also stimulates the differentiation of immunosuppressive Tregs, Bregs, and M2 macrophages, and finally, (iv) AhR signaling participates in the differentiation of many peripheral tissues. On the other hand, AhR signaling is involved in many processes promoting cellular senescence and pathological processes, e.g., osteoporosis, vascular dysfunction, and the age-related remodeling of the immune system. Moreover, it inhibits autophagy and aggravates extracellular matrix degeneration. AhR signaling also stimulates oxidative stress, promotes excessive sphingolipid synthesis, and disturbs energy metabolism by catabolizing NAD+ degradation. The antagonistic pleiotropy of AhR signaling is based on the complex and diverse connections with major signaling pathways in a context-dependent manner. The major regulatory steps include, (i) a specific ligand-dependent activation, (ii) modulation of both genetic and non-genetic responses, (iii) a competition and crosstalk with several transcription factors, such as ARNT, HIF-1α, E2F1, and NF-κB, and (iv) the epigenetic regulation of target genes with binding partners. Thus, not only mTOR signaling but also the AhR factor demonstrates antagonistic pleiotropy in the regulation of the aging process.
Collapse
Affiliation(s)
- Antero Salminen
- Department of Neurology, Institute of Clinical Medicine, University of Eastern Finland, P.O. Box 1627, 70211, Kuopio, Finland.
| |
Collapse
|
25
|
Salminen A. Mutual antagonism between aryl hydrocarbon receptor and hypoxia-inducible factor-1α (AhR/HIF-1α) signaling: Impact on the aging process. Cell Signal 2022; 99:110445. [PMID: 35988806 DOI: 10.1016/j.cellsig.2022.110445] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 07/27/2022] [Accepted: 08/16/2022] [Indexed: 11/26/2022]
Abstract
The ambient oxygen level, many environmental toxins, and the rays of ultraviolet light (UV) provide a significant risk for the maintenance of organismal homeostasis. The aryl hydrocarbon receptors (AhR) represent a complex sensor system not only for environmental toxins and UV radiation but also for many endogenous ligands, e.g., L-tryptophan metabolites. The AhR signaling system is evolutionarily conserved and AhR homologs existed as many as 600 million years ago. The ancient atmosphere demanded the evolution of an oxygen-sensing system, i.e., hypoxia-inducible transcription factors (HIF) and their prolyl hydroxylase regulators (PHD). Given that both signaling systems have important roles in embryogenesis, it seems that they have been involved in the evolution of multicellular organisms. The evolutionary origin of the aging process is unknown although it is most likely associated with the evolution of multicellularity. Intriguingly, there is compelling evidence that while HIF-1α signaling extends the lifespan, that of AhR promotes many age-related degenerative processes, e.g., it increases oxidative stress, inhibits autophagy, promotes cellular senescence, and aggravates extracellular matrix degeneration. In contrast, HIF-1α signaling stimulates autophagy, inhibits cellular senescence, and enhances cell proliferation. Interestingly, there is a clear antagonism between the AhR and HIF-1α signaling pathways. For instance, (i) AhR and HIF-1α factors heterodimerize with the same factor, ARNT/HIF-1β, leading to their competition for DNA-binding, (ii) AhR and HIF-1α signaling exert antagonistic effects on autophagy, and (iii) co-chaperone p23 exhibits specific functions in the signaling of AhR and HIF-1α factors. One might speculate that it is the competition between the AhR and HIF-1α signaling pathways that is a driving force in the aging process.
Collapse
Affiliation(s)
- Antero Salminen
- Department of Neurology, Institute of Clinical Medicine, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland.
| |
Collapse
|
26
|
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
|
27
|
Fehsel K, Schwanke K, Kappel BA, Fahimi E, Meisenzahl-Lechner E, Esser C, Hemmrich K, Haarmann-Stemmann T, Kojda G, Lange-Asschenfeldt C. Activation of the aryl hydrocarbon receptor by clozapine induces preadipocyte differentiation and contributes to endothelial dysfunction. J Psychopharmacol 2022; 36:191-201. [PMID: 34979820 PMCID: PMC8847763 DOI: 10.1177/02698811211055811] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
BACKGROUND The superior therapeutic benefit of clozapine is often associated with metabolic disruptions as obesity, insulin resistance, tachycardia, higher blood pressure, and even hypertension. AIMS These adverse vascular/ metabolic events under clozapine are similar to those caused by polycyclic aromatic hydrocarbons (PAHs), and clozapine shows structural similarity to well-known ligands of the aryl hydrocarbon receptor (AhR). Therefore, we speculated that the side effects caused by clozapine might rely on AhR signaling. METHODS We examined clozapine-induced AhR activation by luciferase reporter assays in hepatoma HepG2 cells and we proved upregulation of the prototypical AhR target gene Cyp1A1 by realtime-PCR (RT-PCR) analysis and enzyme activity. Next we studied the physiological role of AhR in clozapine's effects on human preadipocyte differentiation and on vasodilatation by myography in wild-type and AhR-/- mice. RESULTS In contrast to other antipsychotic drugs (APDs), clozapine triggered AhR activation and Cyp1A1 expression in HepG2 cells and adipocytes. Clozapine induced adipogenesis via AhR signaling. After PGF2α-induced constriction of mouse aortic rings, clozapine strongly reduced the maximal vasorelaxation under acetylcholine in rings from wild-type mice, but only slightly in rings from AhR-/- mice. The reduction was also prevented by pretreatment with the AhR antagonist CH-223191. CONCLUSION Identification of clozapine as a ligand for the AhR opens new perspectives to explain common clozapine therapy-associated adverse effects at the molecular level.
Collapse
Affiliation(s)
- K Fehsel
- Neurobiochemical Research Unit, Department of Psychiatry, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany,K Fehsel, Neurobiochemical Research Unit, Department of Psychiatry, Medical Faculty, Heinrich Heine University Düsseldorf, Bergische Landstrasse 2, 40629 Düsseldorf, Germany.
| | - K Schwanke
- Leibniz Research Institute for Environmental Medicine (IUF), Düsseldorf, Germany
| | - BA Kappel
- Department of Internal Medicine I, University Hospital Aachen, RWTH Aachen University, Aachen, Germany
| | - E Fahimi
- Institute for Pharmacology and Clinical Pharmacology, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - E Meisenzahl-Lechner
- Neurobiochemical Research Unit, Department of Psychiatry, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - C Esser
- Leibniz Research Institute for Environmental Medicine (IUF), Düsseldorf, Germany
| | - K Hemmrich
- Department of Plastic Surgery and Hand Surgery, Burn Center, University Hospital of the Aachen University of Technology, Aachen, Germany
| | - T Haarmann-Stemmann
- Leibniz Research Institute for Environmental Medicine (IUF), Düsseldorf, Germany
| | - G Kojda
- Institute for Pharmacology and Clinical Pharmacology, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - C Lange-Asschenfeldt
- Neurobiochemical Research Unit, Department of Psychiatry, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| |
Collapse
|
28
|
Rajendran R, Ragavan RP, Al-Sehemi AG, Uddin MS, Aleya L, Mathew B. Current understandings and perspectives of petroleum hydrocarbons in Alzheimer's disease and Parkinson's disease: a global concern. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2022; 29:10928-10949. [PMID: 35000177 DOI: 10.1007/s11356-021-17931-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Accepted: 11/30/2021] [Indexed: 06/14/2023]
Abstract
Over the last few decades, the global prevalence of neurodevelopmental and neurodegenerative illnesses has risen rapidly. Although the aetiology remains unclear, evidence is mounting that exposure to persistent hydrocarbon pollutants is a substantial risk factor, predisposing a person to neurological diseases later in life. Epidemiological studies correlate environmental hydrocarbon exposure to brain disorders including neuropathies, cognitive, motor and sensory impairments; neurodevelopmental disorders like autism spectrum disorder (ASD); and neurodegenerative disorders like Alzheimer's disease (AD) and Parkinson's disease (PD). Particulate matter, benzene, toluene, ethylbenzene, xylenes, polycyclic aromatic hydrocarbons and endocrine-disrupting chemicals have all been linked to neurodevelopmental problems in all class of people. There is mounting evidence that supports the prevalence of petroleum hydrocarbon becoming neurotoxic and being involved in the pathogenesis of AD and PD. More study is needed to fully comprehend the scope of these problems in the context of unconventional oil and natural gas. This review summarises in vitro, animal and epidemiological research on the genesis of neurodegenerative disorders, highlighting evidence that supports inexorable role of hazardous hydrocarbon exposure in the pathophysiology of AD and PD. In this review, we offer a summary of the existing evidence gathered through a Medline literature search of systematic reviews and meta-analyses of the most important epidemiological studies published so far.
Collapse
Affiliation(s)
- Rajalakshmi Rajendran
- Department of Pharmacy Practice, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, Kochi, 682041, Kerala, India
| | - Roshni Pushpa Ragavan
- Research Center for Advanced Materials Science, King Khalid University, Abha, 61413, Saudi Arabia.
| | - Abdullah G Al-Sehemi
- Research Center for Advanced Materials Science, King Khalid University, Abha, 61413, Saudi Arabia
- Department of Chemistry, King Khalid University, Abha, 61413, Saudi Arabia
| | - Md Sahab Uddin
- Department of Pharmacy, Southeast University, Dhaka, Bangladesh
- Pharmakon Neuroscience Research Network, Dhaka, Bangladesh
| | - Lotfi Aleya
- Laboratoire Chrono-Environment, CNRS6249, Universite de Bourgogne Franche-Comte, Besancon, France
| | - Bijo Mathew
- Department of Pharmaceutical Chemistry, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Sciences Campus, Kochi, 682 041, India.
| |
Collapse
|
29
|
Hu JR, Myint L, Levey AS, Coresh J, Inker LA, Grams ME, Guallar E, Hansen KD, Rhee EP, Shafi T. A metabolomics approach identified toxins associated with uremic symptoms in advanced chronic kidney disease. Kidney Int 2022; 101:369-378. [PMID: 34843755 PMCID: PMC8792216 DOI: 10.1016/j.kint.2021.10.035] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 09/27/2021] [Accepted: 10/01/2021] [Indexed: 02/03/2023]
Abstract
Uremic symptoms are common in patients with advanced chronic kidney disease, but the toxins that cause these symptoms are unknown. To evaluate this, we performed a cross-sectional study of the 12 month post-randomization follow-up visit of Modification of Diet in Renal Disease (MDRD) participants reporting uremic symptoms who also had available stored serum. We quantified 1,163 metabolites by liquid chromatography-tandem mass spectrometry. For each uremic symptom, we calculated a score as the severity multiplied by the number of days the symptom was experienced. We analyzed the associations of the individual symptom scores with metabolites using linear models with empirical Bayesian inference, adjusted for multiple comparisons. Among 695 participants, the mean measured glomerular filtration rate (mGFR) was 28 mL/min/1.73 m2. Uremic symptoms were more common in the subgroup of 214 patients with an mGFR under 20 mL/min/1.73 m2 (mGFR under 20 subgroup) than in the full group. For all metabolites with significant associations, the direction of the association was concordant in the full group and the subgroup. For gastrointestinal symptoms (bad taste, loss of appetite, nausea, and vomiting), eleven metabolites were associated with symptoms. For neurologic symptoms (decreased alertness, falling asleep during the day, forgetfulness, lack of pep and energy, and tiring easily/weakness), seven metabolites were associated with symptoms. Associations were consistent across sensitivity analyses. Thus, our proof-of-principle study demonstrates the potential for metabolomics to understand metabolic pathways associated with uremic symptoms. Larger, prospective studies with external validation are needed.
Collapse
Affiliation(s)
- Jiun-Ruey Hu
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT
| | - Leslie Myint
- Department of Mathematics, Statistics, and Computer Science, Macalester College, St. Paul, MN
| | | | - Josef Coresh
- Welch Center for Prevention, Epidemiology, and Clinical Research, Johns Hopkins University School of Medicine, Baltimore, MD
| | | | - Morgan E. Grams
- Welch Center for Prevention, Epidemiology, and Clinical Research, Johns Hopkins University School of Medicine, Baltimore, MD,Division of Nephrology, Johns Hopkins Medical Institutions, Baltimore, MD
| | - Eliseo Guallar
- Welch Center for Prevention, Epidemiology, and Clinical Research, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Kasper D. Hansen
- Department of Biostatistics, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD
| | - Eugene P. Rhee
- Division of Nephrology, Massachussetts General Hospital, Boston, MA
| | - Tariq Shafi
- Division of Nephrology, University of Mississippi Medical Center, Jackson, MS
| |
Collapse
|
30
|
Keshavarzi M, Moradbeygi F, Mobini K, Ghaffarian Bahraman A, Mohammadi P, Ghaedi A, Mohammadi-Bardbori A. The interplay of aryl hydrocarbon receptor/WNT/CTNNB1/Notch signaling pathways regulate amyloid beta precursor mRNA/protein expression and effected the learning and memory of mice. Toxicol Res (Camb) 2021; 11:147-161. [PMID: 35237419 PMCID: PMC8882790 DOI: 10.1093/toxres/tfab120] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 11/22/2021] [Accepted: 12/01/2021] [Indexed: 12/31/2022] Open
Abstract
The amyloid beta precursor protein (APP) plays a pathophysiological role in the development of Alzheimer's disease as well as a physiological role in neuronal growth and synaptogenesis. The aryl hydrocarbon receptor (AhR)/WNT/Catenin Beta 1 (CTNNB1)/Notch signaling pathways stamp in many functions, including development and growth of neurons. However, the regulatory role of AhR-/WNT-/CTNNB1-/Notch-induced APP expression and its influence on hippocampal-dependent learning and memory deficits is not clear. Male BALB/C mice received 6-formylindolo[3,2-b]carbazole (an AhR agonist), CH223191(an AhR antagonist), DAPT (an inhibitor of Notch signaling), and XAV-939 (a WNT pathway inhibitor) at a single dose of 100 μg/kg, 1, 5 , and 5 mg/kg of body weight, respectively, via intraperitoneal injection alone or in combination. Gene expression analyses and protein assay were performed on the 7th and 29th days. To assess the hippocampal-dependent memory, all six mice also underwent contextual fear conditioning on the 28th day after treatments. Our results showed that endogenous ligand of AhR has a regulatory effect on APP gene. Also, the interaction of AhR/WNT/CTNNB1 has a positive regulatory effect, but Notch has a negative regulatory effect on the mRNA and protein expression of APP, which have a correlation with mice's learning skills and memory.
Collapse
Affiliation(s)
- Majid Keshavarzi
- Department of Pharmacology and Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz 7146864685, Iran,Department of Environmental Health, Faculty of Health, Sabzevar University of Medical Sciences, Sabzevar 7146864685, Iran
| | - Fatemeh Moradbeygi
- Department of Pharmacology and Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz 7146864685, Iran
| | - Keivan Mobini
- Department of Pharmacology and Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz 7146864685, Iran
| | - Ali Ghaffarian Bahraman
- Department of Pharmacology and Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz 7146864685, Iran,Occupational Environment Research Center, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Parisa Mohammadi
- Department of Environmental Health, Faculty of Health, Sabzevar University of Medical Sciences, Sabzevar 7146864685, Iran
| | - Afsaneh Ghaedi
- Department of Pharmacology and Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz 7146864685, Iran
| | - Afshin Mohammadi-Bardbori
- Correspondence address. Department of Pharmacology and Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz 7146864685, Iran. Tel.: +98(71)32425374; Fax: +98(71)32424326; E-mail:
| |
Collapse
|
31
|
Nisa FY, Rahman MA, Hossen MA, Khan MF, Khan MAN, Majid M, Sultana F, Haque MA. Role of neurotoxicants in the pathogenesis of Alzheimer's disease: a mechanistic insight. Ann Med 2021; 53:1476-1501. [PMID: 34433343 PMCID: PMC8405119 DOI: 10.1080/07853890.2021.1966088] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 08/04/2021] [Indexed: 12/13/2022] Open
Abstract
Alzheimer's disease (AD) is the most conspicuous chronic neurodegenerative syndrome, which has become a significant challenge for the global healthcare system. Multiple studies have corroborated a clear association of neurotoxicants with AD pathogenicity, such as Amyloid beta (Aβ) proteins and neurofibrillary tangles (NFTs), signalling pathway modifications, cellular stress, cognitive dysfunctions, neuronal apoptosis, neuroinflammation, epigenetic modification, and so on. This review, therefore, aimed to address several essential mechanisms and signalling cascades, including Wnt (wingless and int.) signalling pathway, autophagy, mammalian target of rapamycin (mTOR), protein kinase C (PKC) signalling cascades, cellular redox status, energy metabolism, glutamatergic neurotransmissions, immune cell stimulations (e.g. microglia, astrocytes) as well as an amyloid precursor protein (APP), presenilin-1 (PSEN1), presenilin-2 (PSEN2) and other AD-related gene expressions that have been pretentious and modulated by the various neurotoxicants. This review concluded that neurotoxicants play a momentous role in developing AD through modulating various signalling cascades. Nevertheless, comprehension of this risk agent-induced neurotoxicity is far too little. More in-depth epidemiological and systematic investigations are needed to understand the potential mechanisms better to address these neurotoxicants and improve approaches to their risk exposure that aid in AD pathogenesis.Key messagesInevitable cascade mechanisms of how Alzheimer's Disease-related (AD-related) gene expressions are modulated by neurotoxicants have been discussed.Involvement of the neurotoxicants-induced pathways caused an extended risk of AD is explicited.Integration of cell culture, animals and population-based analysis on the clinical severity of AD is addressed.
Collapse
Affiliation(s)
- Fatema Yasmin Nisa
- Department of Biochemistry and Molecular Biology, Faculty of Biological Sciences, University of Chittagong, Chittagong, Bangladesh
| | - Md. Atiar Rahman
- Department of Biochemistry and Molecular Biology, Faculty of Biological Sciences, University of Chittagong, Chittagong, Bangladesh
| | - Md. Amjad Hossen
- Department of Pharmacy, International Islamic University Chittagong, Chittagong, Bangladesh
| | - Mohammad Forhad Khan
- Department of Pharmacy, International Islamic University Chittagong, Chittagong, Bangladesh
| | - Md. Asif Nadim Khan
- Department of Biochemistry and Molecular Biology, Faculty of Biological Sciences, University of Chittagong, Chittagong, Bangladesh
| | - Mumtahina Majid
- Department of Biochemistry and Molecular Biology, Faculty of Biological Sciences, University of Chittagong, Chittagong, Bangladesh
| | - Farjana Sultana
- Department of Biochemistry and Molecular Biology, Faculty of Biological Sciences, University of Chittagong, Chittagong, Bangladesh
| | - Md. Areeful Haque
- Department of Pharmacy, International Islamic University Chittagong, Chittagong, Bangladesh
- Faculty of Pharmacy, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| |
Collapse
|
32
|
Traboulsi H, de Souza AR, Allard B, Haidar Z, Sorin M, Moarbes V, Fixman ED, Martin JG, Eidelman DH, Baglole CJ. Differential Regulation of the Asthmatic Phenotype by the Aryl Hydrocarbon Receptor. Front Physiol 2021; 12:720196. [PMID: 34744763 PMCID: PMC8566992 DOI: 10.3389/fphys.2021.720196] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Accepted: 09/28/2021] [Indexed: 11/26/2022] Open
Abstract
The aryl hydrocarbon receptor (AhR) is a ligand-activated transcription factor that regulates the metabolism of xenobiotics. There is growing evidence that the AhR is implicated in physiological processes such proliferation, differentiation, and immune responses. Recently, a role of the AhR in regulating allergic asthma has been suggested, but whether the AhR also regulates other type of asthma, particularly occupational/irritant-induced asthma, remains unknown. Using AhR-deficient (Ahr−/−) mice, we compared the function of the AhR in the response to ovalbumin (OVA; allergic asthma) vs. chlorine (Cl2; irritant-induced asthma) exposure. Lung inflammation and airway hyperresponsiveness were assessed 24h after exposure to Cl2 or OVA challenge in Ahr−/− and heterozygous (Ahr+/−) mice. After OVA challenge, absence of AhR was associated with significantly enhanced eosinophilia and lymphocyte influx into the airways of Ahr−/− mice. There were also increased levels of interleukin-4 (IL-4) and IL-5 in the airways. However, OVA-induced airway hyperresponsiveness was not affected. In the irritant-induced asthma model caused by exposure to Cl2, the AhR did not regulate the inflammatory response. However, absence of AhR reduced Cl2-induced airway hyperresponsiveness. Collectively, these results support a differential role for the AhR in regulating asthma outcomes in response to diverse etiological agents.
Collapse
Affiliation(s)
- Hussein Traboulsi
- Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre, Montreal, QC, Canada.,Translational Research in Respiratory Diseases Program at the Research Institute of the McGill University Health Centre, Montreal, QC, Canada.,Department of Medicine, McGill University, Montreal, QC, Canada
| | - Angela Rico de Souza
- Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre, Montreal, QC, Canada.,Translational Research in Respiratory Diseases Program at the Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Benoit Allard
- Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Zahraa Haidar
- Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre, Montreal, QC, Canada.,Translational Research in Respiratory Diseases Program at the Research Institute of the McGill University Health Centre, Montreal, QC, Canada.,Department of Medicine, McGill University, Montreal, QC, Canada
| | - Mark Sorin
- Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre, Montreal, QC, Canada.,Translational Research in Respiratory Diseases Program at the Research Institute of the McGill University Health Centre, Montreal, QC, Canada.,Department of Medicine, McGill University, Montreal, QC, Canada
| | - Vanessa Moarbes
- Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre, Montreal, QC, Canada.,Translational Research in Respiratory Diseases Program at the Research Institute of the McGill University Health Centre, Montreal, QC, Canada.,Department of Medicine, McGill University, Montreal, QC, Canada
| | - Elizabeth D Fixman
- Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre, Montreal, QC, Canada.,Translational Research in Respiratory Diseases Program at the Research Institute of the McGill University Health Centre, Montreal, QC, Canada.,Department of Medicine, McGill University, Montreal, QC, Canada
| | - James G Martin
- Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre, Montreal, QC, Canada.,Translational Research in Respiratory Diseases Program at the Research Institute of the McGill University Health Centre, Montreal, QC, Canada.,Department of Medicine, McGill University, Montreal, QC, Canada
| | - David H Eidelman
- Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre, Montreal, QC, Canada.,Translational Research in Respiratory Diseases Program at the Research Institute of the McGill University Health Centre, Montreal, QC, Canada.,Department of Medicine, McGill University, Montreal, QC, Canada
| | - Carolyn J Baglole
- Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre, Montreal, QC, Canada.,Translational Research in Respiratory Diseases Program at the Research Institute of the McGill University Health Centre, Montreal, QC, Canada.,Department of Medicine, McGill University, Montreal, QC, Canada.,Department of Pathology, McGill University, Montreal, QC, Canada.,Department of Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada
| |
Collapse
|
33
|
Behavioural adaptations after antibiotic treatment in male mice are reversed by activation of the aryl hydrocarbon receptor. Brain Behav Immun 2021; 98:317-329. [PMID: 34461234 DOI: 10.1016/j.bbi.2021.08.228] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 07/15/2021] [Accepted: 08/21/2021] [Indexed: 01/08/2023] Open
Abstract
The intestinal microbiota plays an important role in regulating brain functions and behaviour. Microbiota-dependent changes in host physiology have been suggested to be key contributors to psychiatric conditions. However, specific host pathways modulated by the microbiota involved in behavioural control are lacking. Here, we assessed the role of the aryl hydrocarbon receptor (Ahr) in modulating microbiota-related alterations in behaviour in male and female mice after antibiotic (Abx) treatment. Mice of both sexes were treated with Abx to induce bacterial depletion. Mice were then tested in a battery of behavioural tests, including the elevated plus maze and open field tests (anxiety-like behaviour), 3 chamber test (social preference), and the tail suspension and forced swim tests (despair behaviour). Behavioural measurements in the tail suspension test were also performed after microbiota reconstitution and after administration of an Ahr agonist, β-naphthoflavone. Gene expression analyses were performed in the brain, liver, and colon by qPCR. Abx-induced bacterial depletion did not alter anxiety-like behaviour, locomotion, or social preference in either sex. A sex-dependent effect was observed in despair behaviour. Male mice had a reduction in despair behaviour after Abx treatment in both the tail suspension and forced swim tests. A similar alteration in despair behaviour was observed in Ahr knockout mice. Despair behaviour was normalized by either microbiota recolonization or Ahr activation in Abx-treated mice. Ahr activation by β-naphthoflavone was confirmed by increased expression of the Ahr-target genes Cyp1a1, Cyp1b1, and Ahrr. Our results demonstrate a role for Ahr in mediating the behaviours that are regulated by the crosstalk between the intestinal microbiota and the host. Ahr represents a novel potential modulator of behavioural conditions influenced by the intestinal microbiota.
Collapse
|
34
|
The Role of AhR in the Hallmarks of Brain Aging: Friend and Foe. Cells 2021; 10:cells10102729. [PMID: 34685709 PMCID: PMC8534784 DOI: 10.3390/cells10102729] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 10/05/2021] [Accepted: 10/10/2021] [Indexed: 12/24/2022] Open
Abstract
In recent years, aryl hydrocarbon receptor (AhR), a ligand-activated transcription factor, has been considered to be involved in aging phenotypes across several species. This receptor is a highly conserved biosensor that is activated by numerous exogenous and endogenous molecules, including microbiota metabolites, to mediate several physiological and toxicological functions. Brain aging hallmarks, which include glial cell activation and inflammation, increased oxidative stress, mitochondrial dysfunction, and cellular senescence, increase the vulnerability of humans to various neurodegenerative diseases. Interestingly, many studies have implicated AhR signaling pathways in the aging process and longevity across several species. This review provides an overview of the impact of AhR pathways on various aging hallmarks in the brain and the implications for AhR signaling as a mechanism in regulating aging-related diseases of the brain. We also explore how the nature of AhR ligands determines the outcomes of several signaling pathways in brain aging processes.
Collapse
|
35
|
Zhou Y, Zhao WJ, Quan W, Qiao CM, Cui C, Hong H, Shi Y, Niu GY, Zhao LP, Shen YQ. Dynamic changes of activated AHR in microglia and astrocytes in the substantia nigra-striatum system in an MPTP-induced Parkinson's disease mouse model. Brain Res Bull 2021; 176:174-183. [PMID: 34478811 DOI: 10.1016/j.brainresbull.2021.08.013] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 08/10/2021] [Accepted: 08/29/2021] [Indexed: 01/19/2023]
Abstract
Aryl Hydrocarbon Receptor (AHR) is a ligand-activated transcription factor expressed in various brain regions. However, little is known about the role of AHR during neuroinflammation in the 1-methyl-4-phenyl-1,2,3,6-tetrathydropyridine (MPTP)-induced Parkinson's disease (PD) mouse model. Here, mice were sacrificed at day 4, day 6 and day 8 respectively after MPTP or saline treatment. Behavioral tests, Tyrosine hydroxylase (TH) expression, glial reaction, and AHR expression and activation were then assayed. As expected, mice treated with MPTP showed apparent behavioral dysfunctions and significantly reduced TH content. Immunofluorescence (IF) labeling showed an increased trend of phosphorylated AHR activation in the Substantia Nigra pars compacta (SNpc) and striatum after MPTP treatment. Western blot analysis demonstrated that MPTP treatment induced a significantly increased level of AHR at each time point tested, with the highest level observed at day 6 in the striatum. To determine exactly the AHR activation in relation to changes of glial cell reactivity, double IF labeling was performed for either IBA1 (microglia marker) and p-AHR, or GFAP (astrocyte marker) and p-AHR. The results demonstrated that MPTP treatment not only increases the number of p-AHR-positive IBA1-expressing cells in the striatum and the SNpc, but also increases that of p-AHR-positive GFAP-expressing cells in the striatum. Intriguingly, the increase of the number of cells co-expressing both p-AHR and IBA1 was highest at day 4 in response to MPTP in the striatum and at day 8 in the SNpc. The number of cells co-expressing both p-AHR and GFAP was increased at days 4, 6 and 8 in the striatum. In conclusion, our study suggests that AHR activation may facilitate PD diagnosis and serve as a target for the treatment of PD.
Collapse
Affiliation(s)
- Yu Zhou
- Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China; Department of Neurodegeneration and Injury, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China
| | - Wei-Jiang Zhao
- Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China; Department of Neurodegeneration and Injury, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China.
| | - Wei Quan
- Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China; Department of Neurodegeneration and Injury, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China
| | - Chen-Meng Qiao
- Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China; Department of Neurodegeneration and Injury, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China
| | - Chun Cui
- Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China; Department of Neurodegeneration and Injury, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China
| | - Hui Hong
- Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China; Department of Neurodegeneration and Injury, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China
| | - Yun Shi
- Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China; Department of Neurodegeneration and Injury, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China
| | - Gu-Yu Niu
- Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China; Department of Neurodegeneration and Injury, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China
| | - Li-Ping Zhao
- Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China; Department of Neurodegeneration and Injury, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China
| | - Yan-Qin Shen
- Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China; Department of Neurodegeneration and Injury, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China.
| |
Collapse
|
36
|
Hossain MM, Belkadi A, Al-Haddad S, Richardson JR. Deltamethrin Exposure Inhibits Adult Hippocampal Neurogenesis and Causes Deficits in Learning and Memory in Mice. Toxicol Sci 2021; 178:347-357. [PMID: 32976580 DOI: 10.1093/toxsci/kfaa144] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Deficits in learning and memory are often associated with disruption of hippocampal neurogenesis, which is regulated by numerous processes, including precursor cell proliferation, survival, migration, and differentiation to mature neurons. Recent studies demonstrate that adult born neurons in the dentate gyrus (DG) in the hippocampus can functionally integrate into the existing neuronal circuitry and contribute to hippocampal-dependent learning and memory. Here, we demonstrate that relatively short-term deltamethrin exposure (3 mg/kg every 3 days for 1 month) inhibits adult hippocampal neurogenesis and causes deficits in learning and memory in mice. Hippocampal-dependent cognitive functions were evaluated using 2 independent hippocampal-dependent behavioral tests, the novel object recognition task and Morris water maze. We found that deltamethrin-treated mice exhibited profound deficits in novel object recognition and learning and memory in water maze. Deltamethrin exposure significantly decreased bromodeoxyuridine (BrdU)-positive cells (39%) and Ki67+ cells (47%) in the DG of the hippocampus, indicating decreased cellular proliferation. In addition, deltamethrin-treated mice exhibited a 44% decrease in nestin-expressing neural progenitor cells and a 38% reduction in the expression of doublecortin (DCX), an early neuronal differentiation marker. Furthermore, deltamethrin-exposed mice exhibited a 25% reduction in total number of granule cells in the DG. These findings indicate that relatively short-term exposure to deltamethrin causes significant deficits in hippocampal neurogenesis that is associated with impaired learning and memory.
Collapse
Affiliation(s)
- Muhammad M Hossain
- Department of Environmental Health Sciences, Robert Stempel College of Public Health & Social work, Florida International University, Miami, Florida 33199.,Department of Pharmaceutical Sciences, Center for Neurodegenerative Disease and Aging, Northeast Ohio Medical University, Rootstown, Ohio 44272
| | - Abdelmadjid Belkadi
- Department of Environmental Health Sciences, Robert Stempel College of Public Health & Social work, Florida International University, Miami, Florida 33199.,Department of Pharmaceutical Sciences, Center for Neurodegenerative Disease and Aging, Northeast Ohio Medical University, Rootstown, Ohio 44272
| | - Sara Al-Haddad
- Department of Pharmaceutical Sciences, Center for Neurodegenerative Disease and Aging, Northeast Ohio Medical University, Rootstown, Ohio 44272
| | - Jason R Richardson
- Department of Environmental Health Sciences, Robert Stempel College of Public Health & Social work, Florida International University, Miami, Florida 33199.,Department of Pharmaceutical Sciences, Center for Neurodegenerative Disease and Aging, Northeast Ohio Medical University, Rootstown, Ohio 44272
| |
Collapse
|
37
|
Wei GZ, Martin KA, Xing PY, Agrawal R, Whiley L, Wood TK, Hejndorf S, Ng YZ, Low JZY, Rossant J, Nechanitzky R, Holmes E, Nicholson JK, Tan EK, Matthews PM, Pettersson S. Tryptophan-metabolizing gut microbes regulate adult neurogenesis via the aryl hydrocarbon receptor. Proc Natl Acad Sci U S A 2021; 118:e2021091118. [PMID: 34210797 PMCID: PMC8271728 DOI: 10.1073/pnas.2021091118] [Citation(s) in RCA: 95] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
While modulatory effects of gut microbes on neurological phenotypes have been reported, the mechanisms remain largely unknown. Here, we demonstrate that indole, a tryptophan metabolite produced by tryptophanase-expressing gut microbes, elicits neurogenic effects in the adult mouse hippocampus. Neurogenesis is reduced in germ-free (GF) mice and in GF mice monocolonized with a single-gene tnaA knockout (KO) mutant Escherichia coli unable to produce indole. External administration of systemic indole increases adult neurogenesis in the dentate gyrus in these mouse models and in specific pathogen-free (SPF) control mice. Indole-treated mice display elevated synaptic markers postsynaptic density protein 95 and synaptophysin, suggesting synaptic maturation effects in vivo. By contrast, neurogenesis is not induced by indole in aryl hydrocarbon receptor KO (AhR-/-) mice or in ex vivo neurospheres derived from them. Neural progenitor cells exposed to indole exit the cell cycle, terminally differentiate, and mature into neurons that display longer and more branched neurites. These effects are not observed with kynurenine, another AhR ligand. The indole-AhR-mediated signaling pathway elevated the expression of β-catenin, Neurog2, and VEGF-α genes, thus identifying a molecular pathway connecting gut microbiota composition and their metabolic function to neurogenesis in the adult hippocampus. Our data have implications for the understanding of mechanisms of brain aging and for potential next-generation therapeutic opportunities.
Collapse
Affiliation(s)
- George Zhang Wei
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 636921
- National Neuroscience Institute, Singapore 169857
| | - Katherine A Martin
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 636921
- National Neuroscience Institute, Singapore 169857
| | - Peter Yuli Xing
- The Singapore Centre for Environmental Life Sciences Engineering, School of Biological Sciences, Nanyang Technological University, Singapore 637551
- Interdisciplinary Graduate School, Nanyang Technological University, Singapore 637335
| | - Ruchi Agrawal
- The Singapore Centre for Environmental Life Sciences Engineering, School of Biological Sciences, Nanyang Technological University, Singapore 637551
| | - Luke Whiley
- Australian National Phenome Centre, Health Futures Institute, Murdoch University, Perth WA 6150, Australia
- Perron Institute for Neurological and Translational Science, Nedlands WA 6009, Australia
| | - Thomas K Wood
- Department of Chemical Engineering, Pennsylvania State University, University Park, PA 16802
| | - Sophia Hejndorf
- Department of Neurobiology, Care and Society, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Yong Zhi Ng
- The School of Biological Sciences, Nanyang Technological University, Singapore 637551
| | - Jeremy Zhi Yan Low
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 636921
| | - Janet Rossant
- Program in Developmental and Stem Cell Biology, Peter Gilgan Centre for Research and Learning, Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Robert Nechanitzky
- Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, ON M5G 2C1, Canada
| | - Elaine Holmes
- Australian National Phenome Centre, Health Futures Institute, Murdoch University, Perth WA 6150, Australia
- Section for Nutrition Research, Imperial College London, London SW7 2AZ, United Kingdom
| | - Jeremy K Nicholson
- Australian National Phenome Centre, Health Futures Institute, Murdoch University, Perth WA 6150, Australia
- Institute of Global Health Innovation, Imperial College London, London SW7 2NA, United Kingdom
| | - Eng-King Tan
- National Neuroscience Institute, Singapore 169857
| | - Paul M Matthews
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 636921
- UK Dementia Research Institute, Imperial College London, London SW7 2AZ, United Kingdom
- Department of Surgery and Cancer, Imperial College London, London SW7 2AZ, United Kingdom
- Department of Brain Sciences, Imperial College London, London W12 0NN, United Kingdom
| | - Sven Pettersson
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 636921;
- National Neuroscience Institute, Singapore 169857
- Department of Neurobiology, Care and Society, Karolinska Institutet, 171 77 Stockholm, Sweden
- Faculty of Medical Sciences, Sunway University, 47500 Kuala Lumpur, Malaysia
| |
Collapse
|
38
|
Kimura E, Kohda M, Maekawa F, Fujii-Kuriyama Y, Tohyama C. Neurons expressing the aryl hydrocarbon receptor in the locus coeruleus and island of Calleja major are novel targets of dioxin in the mouse brain. Histochem Cell Biol 2021; 156:147-163. [PMID: 33963922 PMCID: PMC8397641 DOI: 10.1007/s00418-021-01990-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/21/2021] [Indexed: 12/14/2022]
Abstract
The aryl hydrocarbon receptor (AhR) acts as a receptor that responds to ligands, including dioxin. The AhR-ligand complex translocates from the cytoplasm into the nucleus to induce gene expression. Because dioxin exposure impairs cognitive and neurobehavioral functions, AhR-expressing neurons need to be identified for elucidation of the dioxin neurotoxicity mechanism. Immunohistochemistry was performed to detect AhR-expressing neurons in the mouse brain and confirm the specificity of the anti-AhR antibody using Ahr-/- mice. Intracellular distribution of AhR and expression level of AhR-target genes, Cyp1a1, Cyp1b1, and Ahr repressor (Ahrr), were analyzed by immunohistochemistry and quantitative RT-PCR, respectively, using mice exposed to 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD). The mouse brains were shown to harbor AhR in neurons of the locus coeruleus (LC) and island of Calleja major (ICjM) during developmental period in Ahr+/+ mice but not in Ahr-/- mice. A significant increase in nuclear AhR of ICjM neurons but not LC neurons was found in 14-day-old mice compared to 5- and 7-day-old mice. AhR was significantly translocated into the nucleus in LC and ICjM neurons of TCDD-exposed adult mice. Additionally, the expression levels of Cyp1a1, Cyp1b1, and Ahrr genes in the brain, a surrogate of TCDD in the tissue, were significantly increased by dioxin exposure, suggesting that dioxin-activated AhR induces gene expression in LC and ICjM neurons. This histochemical study shows the ligand-induced nuclear translocation of AhR at the single-neuron level in vivo. Thus, the neurotoxicological significance of the dioxin-activated AhR in the LC and ICjM warrants further studies.
Collapse
Affiliation(s)
- Eiki Kimura
- Laboratory of Environmental Health Sciences, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan. .,Center for Health and Environmental Risk Research, National Institute for Environmental Studies, 16-2 Onogawa, Tsukuba, 305-8506, Japan. .,Research Fellow, Japan Society for the Promotion of Science (JSPS), 5-3-1 Kojimachi, Chiyoda-ku, Tokyo, 102-0083, Japan.
| | - Masanobu Kohda
- Laboratory of Environmental Health Sciences, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan.,Center for Health and Environmental Risk Research, National Institute for Environmental Studies, 16-2 Onogawa, Tsukuba, 305-8506, Japan
| | - Fumihiko Maekawa
- Center for Health and Environmental Risk Research, National Institute for Environmental Studies, 16-2 Onogawa, Tsukuba, 305-8506, Japan
| | - Yoshiaki Fujii-Kuriyama
- Medical Research Institute, Molecular Epidemiology, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan
| | - Chiharu Tohyama
- Laboratory of Environmental Health Sciences, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan. .,Center for Health and Environmental Risk Research, National Institute for Environmental Studies, 16-2 Onogawa, Tsukuba, 305-8506, Japan. .,Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, 305-8575, Japan.
| |
Collapse
|
39
|
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
|
40
|
Zaragoza-Ojeda M, Apatiga-Vega E, Arenas-Huertero F. Role of aryl hydrocarbon receptor in central nervous system tumors: Biological and therapeutic implications. Oncol Lett 2021; 21:460. [PMID: 33907570 PMCID: PMC8063300 DOI: 10.3892/ol.2021.12721] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Accepted: 01/25/2021] [Indexed: 12/12/2022] Open
Abstract
Aryl hydrocarbon receptor (AHR) is a ligand-activated transcription factor, whose canonical pathway mainly regulates the genes involved in xenobiotic metabolism. However, it can also regulate several responses in a non-canonical manner, such as proliferation, differentiation, cell death and cell adhesion. AhR plays an important role in central nervous system tumors, as it can regulate several cellular responses via different pathways. The polymorphisms of the AHR gene have been associated with the development of gliomas. In addition, the metabolism of tumor cells promotes tumor growth, particularly in tryptophan synthesis, where some metabolites, such as kynurenine, can activate the AhR pathway, triggering cell proliferation in astrocytomas, medulloblastomas and glioblastomas. Furthermore, as part of the changes in neuroblastomas, AHR is able to downregulate the expression of proto-oncogene c-Myc, induce differentiation in tumor cells, and cause cell cycle arrest and apoptosis. Collectively, these data suggested that the modulation of the AhR pathway may downregulate tumor growth, providing a novel strategy for applications for the treatment of certain tumors through the control of the AhR pathway.
Collapse
Affiliation(s)
- Montserrat Zaragoza-Ojeda
- Laboratorio de Investigación en Patología Experimental, Hospital Infantil de México Federico Gómez, Mexico City 06720, México.,Posgrado en Ciencias Biológicas, Facultad de Medicina, Universidad Nacional Autónoma de México (UNAM), Mexico City 04510, México
| | - Elisa Apatiga-Vega
- Laboratorio de Investigación en Patología Experimental, Hospital Infantil de México Federico Gómez, Mexico City 06720, México
| | - Francisco Arenas-Huertero
- Laboratorio de Investigación en Patología Experimental, Hospital Infantil de México Federico Gómez, Mexico City 06720, México
| |
Collapse
|
41
|
Tryptophan Metabolism and Gut-Brain Homeostasis. Int J Mol Sci 2021; 22:ijms22062973. [PMID: 33804088 PMCID: PMC8000752 DOI: 10.3390/ijms22062973] [Citation(s) in RCA: 237] [Impact Index Per Article: 59.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 03/10/2021] [Accepted: 03/11/2021] [Indexed: 02/06/2023] Open
Abstract
Tryptophan is an essential amino acid critical for protein synthesis in humans that has emerged as a key player in the microbiota-gut-brain axis. It is the only precursor for the neurotransmitter serotonin, which is vital for the processing of emotional regulation, hunger, sleep, and pain, as well as colonic motility and secretory activity in the gut. Tryptophan catabolites from the kynurenine degradation pathway also modulate neural activity and are active in the systemic inflammatory cascade. Additionally, tryptophan and its metabolites support the development of the central and enteric nervous systems. Accordingly, dysregulation of tryptophan metabolites plays a central role in the pathogenesis of many neurologic and psychiatric disorders. Gut microbes influence tryptophan metabolism directly and indirectly, with corresponding changes in behavior and cognition. The gut microbiome has thus garnered much attention as a therapeutic target for both neurologic and psychiatric disorders where tryptophan and its metabolites play a prominent role. In this review, we will touch upon some of these features and their involvement in health and disease.
Collapse
|
42
|
Davidsen N, Lauvås AJ, Myhre O, Ropstad E, Carpi D, Gyves EMD, Berntsen HF, Dirven H, Paulsen RE, Bal-Price A, Pistollato F. Exposure to human relevant mixtures of halogenated persistent organic pollutants (POPs) alters neurodevelopmental processes in human neural stem cells undergoing differentiation. Reprod Toxicol 2021; 100:17-34. [PMID: 33333158 PMCID: PMC7992035 DOI: 10.1016/j.reprotox.2020.12.013] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Revised: 12/03/2020] [Accepted: 12/11/2020] [Indexed: 12/11/2022]
Abstract
Halogenated persistent organic pollutants (POPs) like perfluorinated alkylated substances (PFASs), brominated flame retardants (BFRs), organochlorine pesticides and polychlorinated biphenyls (PCBs) are known to cause cancer, immunotoxicity, neurotoxicity and interfere with reproduction and development. Concerns have been raised about the impact of POPs upon brain development and possibly neurodevelopmental disorders. The developing brain is a particularly vulnerable organ due to dynamic and complex neurodevelopmental processes occurring early in life. However, very few studies have reported on the effects of POP mixtures at human relevant exposures, and their impact on key neurodevelopmental processes using human in vitro test systems. Aiming to reduce this knowledge gap, we exposed mixed neuronal/glial cultures differentiated from neural stem cells (NSCs) derived from human induced pluripotent stem cells (hiPSCs) to reconstructed mixtures of 29 different POPs using concentrations comparable to Scandinavian human blood levels. Effects of the POP mixtures on neuronal proliferation, differentiation and synaptogenesis were evaluated using in vitro assays anchored to common key events identified in the existing developmental neurotoxicity (DNT) adverse outcome pathways (AOPs). The present study showed that mixtures of POPs (in particular brominated and chlorinated compounds) at human relevant concentrations increased proliferation of NSCs and decreased synapse number. Based on a mathematical modelling, synaptogenesis and neurite outgrowth seem to be the most sensitive DNT in vitro endpoints. Our results indicate that prenatal exposure to POPs may affect human brain development, potentially contributing to recently observed learning and memory deficits in children.
Collapse
Affiliation(s)
- Nichlas Davidsen
- Department of Environmental Health, Section for Toxicology and Risk Assessment, Norwegian Institute of Public Health, Oslo, Norway
| | - Anna Jacobsen Lauvås
- Department of Environmental Health, Section for Toxicology and Risk Assessment, Norwegian Institute of Public Health, Oslo, Norway
| | - Oddvar Myhre
- Department of Environmental Health, Section for Toxicology and Risk Assessment, Norwegian Institute of Public Health, Oslo, Norway
| | - Erik Ropstad
- Department of Production Animal Clinical Sciences, Faculty of Veterinary Medicine, Norwegian University of Life Sciences, Oslo, Norway
| | - Donatella Carpi
- European Commission, Joint Research Centre (JRC), Ispra, Italy
| | | | - Hanne Friis Berntsen
- Department of Production Animal Clinical Sciences, Faculty of Veterinary Medicine, Norwegian University of Life Sciences, Oslo, Norway; National Institute of Occupational Health, Oslo, Norway
| | - Hubert Dirven
- Department of Environmental Health, Section for Toxicology and Risk Assessment, Norwegian Institute of Public Health, Oslo, Norway
| | - Ragnhild E Paulsen
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, Norway
| | - Anna Bal-Price
- European Commission, Joint Research Centre (JRC), Ispra, Italy
| | | |
Collapse
|
43
|
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: 48] [Impact Index Per Article: 12.0] [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
|
44
|
Lai WT, Deng WF, Xu SX, Zhao J, Xu D, Liu YH, Guo YY, Wang MB, He FS, Ye SW, Yang QF, Liu TB, Zhang YL, Wang S, Li MZ, Yang YJ, Xie XH, Rong H. Shotgun metagenomics reveals both taxonomic and tryptophan pathway differences of gut microbiota in major depressive disorder patients. Psychol Med 2021; 51:90-101. [PMID: 31685046 DOI: 10.1017/s0033291719003027] [Citation(s) in RCA: 81] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
BACKGROUND The microbiota-gut-brain axis, especially the microbial tryptophan (Trp) biosynthesis and metabolism pathway (MiTBamp), may play a critical role in the pathogenesis of major depressive disorder (MDD). However, studies on the MiTBamp in MDD are lacking. The aim of the present study was to analyze the gut microbiota composition and the MiTBamp in MDD patients. METHODS We performed shotgun metagenomic sequencing of stool samples from 26 MDD patients and 29 healthy controls (HCs). In addition to the microbiota community and the MiTBamp analyses, we also built a classification based on the Random Forests (RF) and Boruta algorithm to identify the gut microbiota as biomarkers for MDD. RESULTS The Bacteroidetes abundance was strongly reduced whereas that of Actinobacteria was significantly increased in the MDD patients compared with the abundance in the HCs. Most noteworthy, the MDD patients had increased levels of Bifidobacterium, which is commonly used as a probiotic. Four Kyoto Encyclopedia of Genes and Genomes (KEGG) orthologies (KOs) (K01817, K11358, K01626, K01667) abundances in the MiTBamp were significantly lower in the MDD group. Furthermore, we found a negative correlation between the K01626 abundance and the HAMD scores in the MDD group. Finally, RF classification at the genus level can achieve an area under the receiver operating characteristic curve of 0.890. CONCLUSIONS The present findings enabled a better understanding of the changes in gut microbiota and the related Trp pathway in MDD. Alterations of the gut microbiota may have the potential as biomarkers for distinguishing MDD patients form HCs.
Collapse
Affiliation(s)
- Wen-Tao Lai
- Department of Psychiatry, Shenzhen Kangning Hospital, Shenzhen Mental Health Center, Shenzhen, Guangdong, China
| | - Wen-Feng Deng
- Laboratory of Brain Stimulation and Biological Psychiatry, Brain Function and Psychosomatic Medicine Institute, Second People's Hospital of Huizhou, Huizhou, Guangdong, China
| | - Shu-Xian Xu
- Laboratory of Brain Stimulation and Biological Psychiatry, Brain Function and Psychosomatic Medicine Institute, Second People's Hospital of Huizhou, Huizhou, Guangdong, China
| | - Jie Zhao
- Department of Psychiatry, Shenzhen Kangning Hospital, Shenzhen Mental Health Center, Shenzhen, Guangdong, China
| | - Dan Xu
- Department of Psychiatry, Shenzhen Kangning Hospital, Shenzhen Mental Health Center, Shenzhen, Guangdong, China
| | - Yang-Hui Liu
- Department of Psychiatry, Shenzhen Kangning Hospital, Shenzhen Mental Health Center, Shenzhen, Guangdong, China
| | - Yuan-Yuan Guo
- Department of Psychiatry, Shenzhen Kangning Hospital, Shenzhen Mental Health Center, Shenzhen, Guangdong, China
| | - Ming-Bang Wang
- Xiamen Branch, Shanghai Key Laboratory of Birth Defects, Division of Neonatology, Children's Hospital of Fudan University, National Center for Children's Health, Shanghai, China
| | | | - Shu-Wei Ye
- Department of Psychiatry, Shenzhen Kangning Hospital, Shenzhen Mental Health Center, Shenzhen, Guangdong, China
| | - Qi-Fan Yang
- Laboratory of Brain Stimulation and Biological Psychiatry, Brain Function and Psychosomatic Medicine Institute, Second People's Hospital of Huizhou, Huizhou, Guangdong, China
| | - Tie-Bang Liu
- Department of Psychiatry, Shenzhen Kangning Hospital, Shenzhen Mental Health Center, Shenzhen, Guangdong, China
| | - Ying-Li Zhang
- Department of Depression, Shenzhen Kangning Hospital, Shenzhen Mental Health Center, Shenzhen, Guangdong, China
| | - Sheng Wang
- Department of Psychiatry, Shenzhen Kangning Hospital, Shenzhen Mental Health Center, Shenzhen, Guangdong, China
| | - Min-Zhi Li
- Department of Psychiatry, Shenzhen Kangning Hospital, Shenzhen Mental Health Center, Shenzhen, Guangdong, China
| | - Ying-Jia Yang
- Department of Depression, Shenzhen Kangning Hospital, Shenzhen Mental Health Center, Shenzhen, Guangdong, China
| | - Xin-Hui Xie
- Department of Psychiatry, Shenzhen Kangning Hospital, Shenzhen Mental Health Center, Shenzhen, Guangdong, China
- Laboratory of Brain Stimulation and Biological Psychiatry, Brain Function and Psychosomatic Medicine Institute, Second People's Hospital of Huizhou, Huizhou, Guangdong, China
- Center of Acute Psychiatry Service, Second People's Hospital of Huizhou, Huizhou, Guangdong, China
| | - Han Rong
- Department of Psychiatry, Shenzhen Kangning Hospital, Shenzhen Mental Health Center, Shenzhen, Guangdong, China
- Affiliated Shenzhen Clinical College of Psychiatry, Jining Medical University, Jining, Shandong, China
| |
Collapse
|
45
|
Mir RH, Sawhney G, Pottoo FH, Mohi-Ud-Din R, Madishetti S, Jachak SM, Ahmed Z, Masoodi MH. Role of environmental pollutants in Alzheimer's disease: a review. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2020; 27:44724-44742. [PMID: 32715424 DOI: 10.1007/s11356-020-09964-x] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Accepted: 06/30/2020] [Indexed: 06/11/2023]
Abstract
Neurodegenerative disorders are commonly erratic influenced by various factors including lifestyle, environmental, and genetic factors. In recent observations, it has been hypothesized that exposure to various environmental factors enhances the risk of Alzheimer's disease (AD). The exact etiology of Alzheimer's disease is still unclear; however, the contribution of environmental factors in the pathology of AD is widely acknowledged. Based on the available literature, the review aims to culminate in the prospective correlation between the various environmental factors and AD. The prolonged exposure to the various well-known environmental factors including heavy metals, air pollutants (particulate matter), pesticides, nanoparticles containing metals, industrial chemicals results in accelerating the progression of AD. Common mechanisms have been documented in the field of environmental contaminants for enhancing amyloid-β (Aβ) peptide along with tau phosphorylation, resulting in the initiation of senile plaques and neurofibrillary tangles, which results in the death of neurons. This review offers a compilation of available data to support the long-suspected correlation between environmental risk factors and AD pathology. Graphical abstract .
Collapse
Affiliation(s)
- Reyaz Hassan Mir
- Pharmaceutical Chemistry Division, Department of Pharmaceutical Sciences, University of Kashmir, Hazratbal, Srinagar, Kashmir, 190006, India.
| | - Gifty Sawhney
- Inflammation Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Canal Road, Jammu-Tawi, Jammu, 180001, India
| | - Faheem Hyder Pottoo
- Department of Pharmacology, College of Clinical Pharmacy, Imam Abdulrahman Bin Faisal University, P.O.BOX 1982, Dammam, 31441, Saudi Arabia
| | - Roohi Mohi-Ud-Din
- Pharmacognosy Division, Department of Pharmaceutical Sciences, University of Kashmir, Hazratbal, Srinagar, Kashmir, 190006, India
| | - Sreedhar Madishetti
- Inflammation Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Canal Road, Jammu-Tawi, Jammu, 180001, India
| | - Sanjay M Jachak
- Department of Natural Products, National Institute of Pharmaceutical Education and Research (NIPER), Sector 67, SAS Nagar, Mohali, Punjab, 160062, India
| | - Zabeer Ahmed
- Inflammation Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Canal Road, Jammu-Tawi, Jammu, 180001, India
| | - Mubashir Hussain Masoodi
- Pharmaceutical Chemistry Division, Department of Pharmaceutical Sciences, University of Kashmir, Hazratbal, Srinagar, Kashmir, 190006, India.
| |
Collapse
|
46
|
Guerrina N, Aloufi N, Shi F, Prasade K, Mehrotra C, Traboulsi H, Matthews J, Eidelman DH, Hamid Q, Baglole CJ. The aryl hydrocarbon receptor reduces LC3II expression and controls endoplasmic reticulum stress. Am J Physiol Lung Cell Mol Physiol 2020; 320:L339-L355. [PMID: 33236922 DOI: 10.1152/ajplung.00122.2020] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
The aryl hydrocarbon receptor (AhR) is a ligand-activated transcription factor whose physiological function is poorly understood. The AhR is highly expressed in barrier organs such as the skin, intestine, and lung. The lungs are continuously exposed to environmental pollutants such as cigarette smoke (CS) that can induce cell death mechanisms such as apoptosis, autophagy, and endoplasmic reticulum (ER) stress. CS also contains toxicants that are AhR ligands. We have previously shown that the AhR protects against apoptosis, but whether the AhR also protects against autophagy or ER stress is not known. Using cigarette smoke extract (CSE) as our in vitro surrogate of environmental tobacco exposure, we first assessed the conversion of LC3I to LC3II, a classic feature of both autophagic and ER stress-mediated cell death pathways. LC3II was elevated in CSE-exposed lung structural cells [mouse lung fibroblasts (MLFs), MLE12 and A549 cells] when AhR was absent. However, this heightened LC3II expression could not be explained by increased expression of key autophagy genes (Gabarapl1, Becn1, Map1lc3b), upregulation of upstream autophagic machinery (Atg5-12, Atg3), or impaired autophagic flux, suggesting that LC3II may be autophagy independent. This was further supported by the absence of autophagosomes in Ahr-/- lung cells. However, Ahr-/- lung cells had widespread ER dilation, elevated expression of the ER stress markers CHOP and GADD34, and an accumulation of ubiquitinated proteins. These findings collectively illustrate a novel role for the AhR in attenuating ER stress by a mechanism that may be autophagy independent.
Collapse
Affiliation(s)
- Necola Guerrina
- Research Institute of the McGill University Health Centre, McGill University, Montreal, Quebec, Canada.,Department of Pathology, McGill University, Montreal, Quebec, Canada
| | - Noof Aloufi
- Research Institute of the McGill University Health Centre, McGill University, Montreal, Quebec, Canada.,Department of Pathology, McGill University, Montreal, Quebec, Canada
| | - Fangyi Shi
- Research Institute of the McGill University Health Centre, McGill University, Montreal, Quebec, Canada.,Department of Pathology, McGill University, Montreal, Quebec, Canada
| | - Kashmira Prasade
- Research Institute of the McGill University Health Centre, McGill University, Montreal, Quebec, Canada
| | - Caitlin Mehrotra
- Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| | - Hussein Traboulsi
- Research Institute of the McGill University Health Centre, McGill University, Montreal, Quebec, Canada.,Department of Medicine, McGill University, Montreal, Quebec, Canada
| | - Jason Matthews
- Department of Nutrition, University of Oslo, Oslo, Norway.,Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada
| | - David H Eidelman
- Research Institute of the McGill University Health Centre, McGill University, Montreal, Quebec, Canada.,Department of Medicine, McGill University, Montreal, Quebec, Canada
| | - Qutayba Hamid
- Research Institute of the McGill University Health Centre, McGill University, Montreal, Quebec, Canada.,Department of Medicine, McGill University, Montreal, Quebec, Canada.,College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
| | - Carolyn J Baglole
- Research Institute of the McGill University Health Centre, McGill University, Montreal, Quebec, Canada.,Department of Pathology, McGill University, Montreal, Quebec, Canada.,Department of Medicine, McGill University, Montreal, Quebec, Canada.,Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
47
|
Wnuk A, Rzemieniec J, Przepiórska K, Wesołowska J, Wójtowicz AK, Kajta M. Autophagy-related neurotoxicity is mediated via AHR and CAR in mouse neurons exposed to DDE. THE SCIENCE OF THE TOTAL ENVIRONMENT 2020; 742:140599. [PMID: 32721735 DOI: 10.1016/j.scitotenv.2020.140599] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 06/25/2020] [Accepted: 06/27/2020] [Indexed: 06/11/2023]
Abstract
DDE (dichlorodiphenyldichloroethylene) is an environmental metabolite of the pesticide DDT, which is still present in the environment, and its insecticidal properties are used to fight malaria and the Zika virus disease. We showed for the first time that the neurotoxic effects of DDE involve autophagy, as demonstrated by elevated levels of Becn1, Map1lc3a/MAP1LC3A, Map1lc3b, and Nup62/NUP62 and an increase in autophagosome formation. The suggestion that the aryl hydrocarbon receptor (AHR) and the constitutive androstane receptor (CAR) are involved in the neurotoxic effect of DDE was supported by increases in the mRNA and protein expression of these receptors, as detected by qPCR, ELISA, immunofluorescence labeling and confocal microscopy. Selective antagonists of the receptors, including alpha-naphthoflavone, CH223191, and CINPA 1, inhibited p,p'-DDE- and o,p'-DDE-induced LDH release and caspase-3 activity, while specific siRNAs (Ahr and Car siRNA) reduced the levels of p,p'-DDE- and o,p'-DDE-induced autophagosome formation. Although the neurotoxic effects of DDE were isomer independent, the mechanisms of p,p'- and o,p'-DDE were isomer specific. Therefore, we identified previously unknown mechanisms of the neurotoxic actions of DDE that, in addition to inducing apoptosis, stimulate autophagy in mouse neocortical cultures and induce AHR and CAR signaling.
Collapse
Affiliation(s)
- Agnieszka Wnuk
- Maj Institute of Pharmacology, Polish Academy of Sciences, Department of Experimental Neuroendocrinology, Laboratory of Molecular Neuroendocrinology, Smetna street 12, 31-343 Krakow, Poland
| | - Joanna Rzemieniec
- Maj Institute of Pharmacology, Polish Academy of Sciences, Department of Experimental Neuroendocrinology, Laboratory of Molecular Neuroendocrinology, Smetna street 12, 31-343 Krakow, Poland
| | - Karolina Przepiórska
- Maj Institute of Pharmacology, Polish Academy of Sciences, Department of Experimental Neuroendocrinology, Laboratory of Molecular Neuroendocrinology, Smetna street 12, 31-343 Krakow, Poland
| | - Julita Wesołowska
- Maj Institute of Pharmacology, Polish Academy of Sciences, Laboratory for In vivo and In Vitro Imaging, Smetna street 12, 31-343 Krakow, Poland
| | - Anna Katarzyna Wójtowicz
- University of Agriculture, Faculty of Animal Sciences, Department of Nutrition, Animal Biotechnology and Fisheries, Adama Mickiewicza 24/28, 30-059 Krakow, Poland
| | - Małgorzata Kajta
- Maj Institute of Pharmacology, Polish Academy of Sciences, Department of Experimental Neuroendocrinology, Laboratory of Molecular Neuroendocrinology, Smetna street 12, 31-343 Krakow, Poland.
| |
Collapse
|
48
|
Skonieczna-Żydecka K, Jakubczyk K, Maciejewska-Markiewicz D, Janda K, Kaźmierczak-Siedlecka K, Kaczmarczyk M, Łoniewski I, Marlicz W. Gut Biofactory-Neurocompetent Metabolites within the Gastrointestinal Tract. A Scoping Review. Nutrients 2020; 12:E3369. [PMID: 33139656 PMCID: PMC7693392 DOI: 10.3390/nu12113369] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 10/27/2020] [Accepted: 10/29/2020] [Indexed: 12/12/2022] Open
Abstract
The gut microbiota have gained much scientific attention recently. Apart from unravelling the taxonomic data, we should understand how the altered microbiota structure corresponds to functions of this complex ecosystem. The metabolites of intestinal microorganisms, especially bacteria, exert pleiotropic effects on the human organism and contribute to the host systemic balance. These molecules play key roles in regulating immune and metabolic processes. A subset of them affect the gut brain axis signaling and balance the mental wellbeing. Neurotransmitters, short chain fatty acids, tryptophan catabolites, bile acids and phosphatidylcholine, choline, serotonin, and L-carnitine metabolites possess high neuroactive potential. A scoping literature search in PubMed/Embase was conducted up until 20 June 2020, using three major search terms "microbiota metabolites" AND "gut brain axis" AND "mental health". This review aimed to enhance our knowledge regarding the gut microbiota functional capacity, and support current and future attempts to create new compounds for future clinical interventions.
Collapse
Affiliation(s)
- Karolina Skonieczna-Żydecka
- Department of Human Nutrition and Metabolomics, Pomeranian Medical University in Szczecin, 71-460 Szczecin, Poland; (K.S.-Ż.); (K.J.); (D.M.-M.); (K.J.)
| | - Karolina Jakubczyk
- Department of Surgical Oncology, Medical University of Gdansk, Smoluchowskiego 17, 80-214 Gdańsk, Poland;
| | - Dominika Maciejewska-Markiewicz
- Department of Human Nutrition and Metabolomics, Pomeranian Medical University in Szczecin, 71-460 Szczecin, Poland; (K.S.-Ż.); (K.J.); (D.M.-M.); (K.J.)
| | - Katarzyna Janda
- Department of Human Nutrition and Metabolomics, Pomeranian Medical University in Szczecin, 71-460 Szczecin, Poland; (K.S.-Ż.); (K.J.); (D.M.-M.); (K.J.)
| | | | - Mariusz Kaczmarczyk
- Department of Clinical and Molecular Biochemistry, Pomeranian Medical University in Szczecin, 70-111 Szczecin, Poland;
| | - Igor Łoniewski
- Department of Human Nutrition and Metabolomics, Pomeranian Medical University in Szczecin, 71-460 Szczecin, Poland; (K.S.-Ż.); (K.J.); (D.M.-M.); (K.J.)
| | - Wojciech Marlicz
- Department of Gastroenterology, Pomeranian Medical University, 71-252 Szczecin, Poland
- The Centre for Digestive Diseases Endoklinika, 70-535 Szczecin, Poland
| |
Collapse
|
49
|
Arnoriaga-Rodríguez M, Mayneris-Perxachs J, Burokas A, Contreras-Rodríguez O, Blasco G, Coll C, Biarnés C, Miranda-Olivos R, Latorre J, Moreno-Navarrete JM, Castells-Nobau A, Sabater M, Palomo-Buitrago ME, Puig J, Pedraza S, Gich J, Pérez-Brocal V, Ricart W, Moya A, Fernández-Real X, Ramió-Torrentà L, Pamplona R, Sol J, Jové M, Portero-Otin M, Maldonado R, Fernández-Real JM. Obesity Impairs Short-Term and Working Memory through Gut Microbial Metabolism of Aromatic Amino Acids. Cell Metab 2020; 32:548-560.e7. [PMID: 33027674 DOI: 10.1016/j.cmet.2020.09.002] [Citation(s) in RCA: 81] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 05/12/2020] [Accepted: 08/31/2020] [Indexed: 02/07/2023]
Abstract
The gut microbiome has been linked to fear extinction learning in animal models. Here, we aimed to explore the gut microbiome and memory domains according to obesity status. A specific microbiome profile associated with short-term memory, working memory, and the volume of the hippocampus and frontal regions of the brain differentially in human subjects with and without obesity. Plasma and fecal levels of aromatic amino acids, their catabolites, and vegetable-derived compounds were longitudinally associated with short-term and working memory. Functionally, microbiota transplantation from human subjects with obesity led to decreased memory scores in mice, aligning this trait from humans with that of recipient mice. RNA sequencing of the medial prefrontal cortex of mice revealed that short-term memory associated with aromatic amino acid pathways, inflammatory genes, and clusters of bacterial species. These results highlight the potential therapeutic value of targeting the gut microbiota for memory impairment, specifically in subjects with obesity.
Collapse
Affiliation(s)
- María Arnoriaga-Rodríguez
- Department of Diabetes, Endocrinology and Nutrition, Dr. Josep Trueta University Hospital, Girona, Spain; Nutrition, Eumetabolism and Health Group, Girona Biomedical Research Institute (IdibGi), Girona, Spain; Biomedical Research Networking Center for Physiopathology of Obesity and Nutrition (CIBEROBN), Madrid, Spain; Department of Medical Sciences, Faculty of Medicine, Girona University, Girona, Spain
| | - Jordi Mayneris-Perxachs
- Department of Diabetes, Endocrinology and Nutrition, Dr. Josep Trueta University Hospital, Girona, Spain; Nutrition, Eumetabolism and Health Group, Girona Biomedical Research Institute (IdibGi), Girona, Spain; Biomedical Research Networking Center for Physiopathology of Obesity and Nutrition (CIBEROBN), Madrid, Spain
| | - Aurelijus Burokas
- Laboratory of Neuropharmacology, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | - Oren Contreras-Rodríguez
- Nutrition, Eumetabolism and Health Group, Girona Biomedical Research Institute (IdibGi), Girona, Spain; Psychiatry Department, Bellvitge University Hospital, Bellvitge Biomedical Research Institute (IDIBELL) and CIBERSAM, Barcelona, Spain
| | - Gerard Blasco
- Institute of Diagnostic Imaging (IDI)-Research Unit (IDIR), Parc Sanitari Pere Virgili, Barcelona, Spain; Medical Imaging, Girona Biomedical Research Institute (IdibGi), Girona, Spain
| | - Clàudia Coll
- Neuroimmunology and Multiple Sclerosis Unit, Department of Neurology, Dr. Josep Trueta University Hospital, Girona, Spain
| | - Carles Biarnés
- Institute of Diagnostic Imaging (IDI)-Research Unit (IDIR), Parc Sanitari Pere Virgili, Barcelona, Spain
| | - Romina Miranda-Olivos
- Nutrition, Eumetabolism and Health Group, Girona Biomedical Research Institute (IdibGi), Girona, Spain; Psychiatry Department, Bellvitge University Hospital, Bellvitge Biomedical Research Institute (IDIBELL) and CIBERSAM, Barcelona, Spain
| | - Jèssica Latorre
- Department of Diabetes, Endocrinology and Nutrition, Dr. Josep Trueta University Hospital, Girona, Spain; Nutrition, Eumetabolism and Health Group, Girona Biomedical Research Institute (IdibGi), Girona, Spain; Biomedical Research Networking Center for Physiopathology of Obesity and Nutrition (CIBEROBN), Madrid, Spain
| | - José-Maria Moreno-Navarrete
- Department of Diabetes, Endocrinology and Nutrition, Dr. Josep Trueta University Hospital, Girona, Spain; Nutrition, Eumetabolism and Health Group, Girona Biomedical Research Institute (IdibGi), Girona, Spain; Biomedical Research Networking Center for Physiopathology of Obesity and Nutrition (CIBEROBN), Madrid, Spain; Department of Medical Sciences, Faculty of Medicine, Girona University, Girona, Spain
| | - Anna Castells-Nobau
- Department of Diabetes, Endocrinology and Nutrition, Dr. Josep Trueta University Hospital, Girona, Spain; Nutrition, Eumetabolism and Health Group, Girona Biomedical Research Institute (IdibGi), Girona, Spain; Biomedical Research Networking Center for Physiopathology of Obesity and Nutrition (CIBEROBN), Madrid, Spain
| | - Mònica Sabater
- Department of Diabetes, Endocrinology and Nutrition, Dr. Josep Trueta University Hospital, Girona, Spain; Nutrition, Eumetabolism and Health Group, Girona Biomedical Research Institute (IdibGi), Girona, Spain; Biomedical Research Networking Center for Physiopathology of Obesity and Nutrition (CIBEROBN), Madrid, Spain
| | - María Encarnación Palomo-Buitrago
- Department of Diabetes, Endocrinology and Nutrition, Dr. Josep Trueta University Hospital, Girona, Spain; Nutrition, Eumetabolism and Health Group, Girona Biomedical Research Institute (IdibGi), Girona, Spain
| | - Josep Puig
- Department of Medical Sciences, Faculty of Medicine, Girona University, Girona, Spain; Institute of Diagnostic Imaging (IDI)-Research Unit (IDIR), Parc Sanitari Pere Virgili, Barcelona, Spain; Medical Imaging, Girona Biomedical Research Institute (IdibGi), Girona, Spain
| | - Salvador Pedraza
- Department of Medical Sciences, Faculty of Medicine, Girona University, Girona, Spain; Medical Imaging, Girona Biomedical Research Institute (IdibGi), Girona, Spain; Department of Radiology, Dr. Josep Trueta University Hospital, Girona, Spain
| | - Jordi Gich
- Department of Medical Sciences, Faculty of Medicine, Girona University, Girona, Spain; Girona Neurodegeneration and Neuroinflammation Group, Girona Biomedical Research Institute (IdibGi), Girona, Spain
| | - Vicente Pérez-Brocal
- Department of Genomics and Health, Foundation for the Promotion of Health and Biomedical Research of Valencia Region (FISABIO-Public Health), Valencia, Spain; Biomedical Research Networking Center for Epidemiology and Public Health (CIBERESP), Madrid, Spain
| | - Wifredo Ricart
- Department of Diabetes, Endocrinology and Nutrition, Dr. Josep Trueta University Hospital, Girona, Spain; Nutrition, Eumetabolism and Health Group, Girona Biomedical Research Institute (IdibGi), Girona, Spain; Biomedical Research Networking Center for Physiopathology of Obesity and Nutrition (CIBEROBN), Madrid, Spain; Department of Medical Sciences, Faculty of Medicine, Girona University, Girona, Spain
| | - Andrés Moya
- Department of Genomics and Health, Foundation for the Promotion of Health and Biomedical Research of Valencia Region (FISABIO-Public Health), Valencia, Spain; Biomedical Research Networking Center for Epidemiology and Public Health (CIBERESP), Madrid, Spain; Institute for Integrative Systems Biology (I2SysBio), University of Valencia and Spanish National Research Council (CSIC), Valencia, Spain
| | - Xavier Fernández-Real
- Institute of Mathematics, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Lluís Ramió-Torrentà
- Department of Medical Sciences, Faculty of Medicine, Girona University, Girona, Spain; Neuroimmunology and Multiple Sclerosis Unit, Department of Neurology, Dr. Josep Trueta University Hospital, Girona, Spain; Girona Neurodegeneration and Neuroinflammation Group, Girona Biomedical Research Institute (IdibGi), Girona, Spain
| | - Reinald Pamplona
- Metabolic Pathophysiology Research Group, Lleida Biomedical Research Institute (IRBLleida)-Universitat de Lleida, Lleida, Spain
| | - Joaquim Sol
- Metabolic Pathophysiology Research Group, Lleida Biomedical Research Institute (IRBLleida)-Universitat de Lleida, Lleida, Spain
| | - Mariona Jové
- Metabolic Pathophysiology Research Group, Lleida Biomedical Research Institute (IRBLleida)-Universitat de Lleida, Lleida, Spain
| | - Manuel Portero-Otin
- Metabolic Pathophysiology Research Group, Lleida Biomedical Research Institute (IRBLleida)-Universitat de Lleida, Lleida, Spain
| | - Rafael Maldonado
- Laboratory of Neuropharmacology, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain; Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain.
| | - José Manuel Fernández-Real
- Department of Diabetes, Endocrinology and Nutrition, Dr. Josep Trueta University Hospital, Girona, Spain; Nutrition, Eumetabolism and Health Group, Girona Biomedical Research Institute (IdibGi), Girona, Spain; Biomedical Research Networking Center for Physiopathology of Obesity and Nutrition (CIBEROBN), Madrid, Spain; Department of Medical Sciences, Faculty of Medicine, Girona University, Girona, Spain.
| |
Collapse
|
50
|
Liu F, Cheng X, Zhong S, Liu C, Jolkkonen J, Zhang X, Liang Y, Liu Z, Zhao C. Communications Between Peripheral and the Brain-Resident Immune System in Neuronal Regeneration After Stroke. Front Immunol 2020; 11:1931. [PMID: 33042113 PMCID: PMC7530165 DOI: 10.3389/fimmu.2020.01931] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Accepted: 07/17/2020] [Indexed: 12/14/2022] Open
Abstract
Cerebral ischemia may cause irreversible neural network damage and result in functional deficits. Targeting neuronal repair after stroke potentiates the formation of new connections, which can be translated into a better functional outcome. Innate and adaptive immune responses in the brain and the periphery triggered by ischemic damage participate in regulating neural repair after a stroke. Immune cells in the blood circulation and gut lymphatic tissues that have been shaped by immune components including gut microbiota and metabolites can infiltrate the ischemic brain and, once there, influence neuronal regeneration either directly or by modulating the properties of brain-resident immune cells. Immune-related signalings and metabolites from the gut microbiota can also directly alter the phenotypes of resident immune cells to promote neuronal regeneration. In this review, we discuss several potential mechanisms through which peripheral and brain-resident immune components can cooperate to promote first the resolution of neuroinflammation and subsequently to improved neural regeneration and a better functional recovery. We propose that new insights into discovery of regulators targeting pro-regenerative process in this complex neuro-immune network may lead to novel strategies for neuronal regeneration.
Collapse
Affiliation(s)
- Fangxi Liu
- Neurology, The First Hospital of China Medical University, Shenyang, China
| | - Xi Cheng
- Neurology, The First Hospital of China Medical University, Shenyang, China
| | - Shanshan Zhong
- Neurology, The First Hospital of China Medical University, Shenyang, China
| | - Chang Liu
- Neurology, The First Hospital of China Medical University, Shenyang, China
| | - Jukka Jolkkonen
- A.I. Virtanen Institute and Institute of Clinical Medicine/Neurology, University of Eastern Finland, Kuopio, Finland
| | - Xiuchun Zhang
- Neurology, The First Hospital of China Medical University, Shenyang, China
| | - Yifan Liang
- Neurology, The First Hospital of China Medical University, Shenyang, China
| | - Zhouyang Liu
- Neurology, The First Hospital of China Medical University, Shenyang, China
| | - Chuansheng Zhao
- Neurology, The First Hospital of China Medical University, Shenyang, China.,Stroke Center, The First Hospital of China Medical University, Shenyang, China
| |
Collapse
|