1
|
Chen CS, Mueller D, Knep E, Ebitz RB, Grissom NM. Dopamine and Norepinephrine Differentially Mediate the Exploration-Exploitation Tradeoff. J Neurosci 2024; 44:e1194232024. [PMID: 39214707 PMCID: PMC11529815 DOI: 10.1523/jneurosci.1194-23.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 08/18/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024] Open
Abstract
Dopamine (DA) and norepinephrine (NE) have been repeatedly implicated in neuropsychiatric vulnerability, in part via their roles in mediating the decision-making processes. Although two neuromodulators share a synthesis pathway and are coactivated under states of arousal, they engage in distinct circuits and modulatory roles. However, the specific role of each neuromodulator in decision-making, in particular the exploration-exploitation tradeoff, remains unclear. Revealing how each neuromodulator contributes to exploration-exploitation tradeoff is important in guiding mechanistic hypotheses emerging from computational psychiatric approaches. To understand the differences and overlaps of the roles of these two catecholamine systems in regulating exploration, a direct comparison using the same dynamic decision-making task is needed. Here, we ran male and female mice in a restless two-armed bandit task, which encourages both exploration and exploitation. We systemically administered a nonselective DA antagonist (flupenthixol), a nonselective DA agonist (apomorphine), a NE beta-receptor antagonist (propranolol), and a NE beta-receptor agonist (isoproterenol) and examined changes in exploration within subjects across sessions. We found a bidirectional modulatory effect of dopamine on exploration. Increasing dopamine activity decreased exploration and decreasing dopamine activity increased exploration. The modulatory effect of beta-noradrenergic receptor activity on exploration was mediated by sex. Reinforcement learning model parameters suggested that dopamine modulation affected exploration via decision noise and norepinephrine modulation affected exploration via sensitivity to outcome. Together, these findings suggested that the mechanisms that govern the exploration-exploitation transition are sensitive to changes in both catecholamine functions and revealed differential roles for NE and DA in mediating exploration.
Collapse
Affiliation(s)
- Cathy S Chen
- Department of Psychology, University of Minnesota, Minneapolis, Minnesota 55455
| | - Dana Mueller
- Department of Psychology, University of Minnesota, Minneapolis, Minnesota 55455
| | - Evan Knep
- Department of Psychology, University of Minnesota, Minneapolis, Minnesota 55455
| | - R Becket Ebitz
- Department of Neurosciences, Université de Montréal, Montréal, Quebec H3T 1J4, Canada
| | - Nicola M Grissom
- Department of Psychology, University of Minnesota, Minneapolis, Minnesota 55455
| |
Collapse
|
2
|
Goyal A, Yuen J, Sinicrope S, Winter B, Randall L, Rusheen AE, Blaha CD, Bennet KE, Lee KH, Shin H, Oh Y. Resolution of tonic concentrations of highly similar neurotransmitters using voltammetry and deep learning. Mol Psychiatry 2024; 29:3076-3085. [PMID: 38664492 PMCID: PMC11449650 DOI: 10.1038/s41380-024-02537-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 03/11/2024] [Accepted: 03/15/2024] [Indexed: 06/27/2024]
Abstract
With advances in our understanding regarding the neurochemical underpinnings of neurological and psychiatric diseases, there is an increased demand for advanced computational methods for neurochemical analysis. Despite having a variety of techniques for measuring tonic extracellular concentrations of neurotransmitters, including voltammetry, enzyme-based sensors, amperometry, and in vivo microdialysis, there is currently no means to resolve concentrations of structurally similar neurotransmitters from mixtures in the in vivo environment with high spatiotemporal resolution and limited tissue damage. Since a variety of research and clinical investigations involve brain regions containing electrochemically similar monoamines, such as dopamine and norepinephrine, developing a model to resolve the respective contributions of these neurotransmitters is of vital importance. Here we have developed a deep learning network, DiscrimNet, a convolutional autoencoder capable of accurately predicting individual tonic concentrations of dopamine, norepinephrine, and serotonin from both in vitro mixtures and the in vivo environment in anesthetized rats, measured using voltammetry. The architecture of DiscrimNet is described, and its ability to accurately predict in vitro and unseen in vivo concentrations is shown to vastly outperform a variety of shallow learning algorithms previously used for neurotransmitter discrimination. DiscrimNet is shown to generalize well to data captured from electrodes unseen during model training, eliminating the need to retrain the model for each new electrode. DiscrimNet is also shown to accurately predict the expected changes in dopamine and serotonin after cocaine and oxycodone administration in anesthetized rats in vivo. DiscrimNet therefore offers an exciting new method for real-time resolution of in vivo voltammetric signals into component neurotransmitters.
Collapse
Affiliation(s)
- Abhinav Goyal
- Medical Scientist Training Program, Mayo Clinic, Rochester, MN, 55905, USA
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN, 55905, USA
| | - Jason Yuen
- Department of Neurosurgery, Southmead Hospital, Bristol, BS10 5NB, UK
| | - Stephen Sinicrope
- Department of Neuroscience, University of Chicago, Chicago, IL, 60637, USA
| | - Bailey Winter
- Medical Scientist Training Program, Mayo Clinic, Rochester, MN, 55905, USA
| | - Lindsey Randall
- Medical Scientist Training Program, Mayo Clinic, Rochester, MN, 55905, USA
| | - Aaron E Rusheen
- Department of Neurosurgery, Johns Hopkins University, Baltimore, MD, 21287, USA
| | - Charles D Blaha
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN, 55905, USA
| | - Kevin E Bennet
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN, 55905, USA
- Division of Engineering, Mayo Clinic, Rochester, MN, 55905, USA
| | - Kendall H Lee
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN, 55905, USA
- Department of Biomedical Engineering, Mayo Clinic, Rochester, MN, 55905, USA
| | - Hojin Shin
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN, 55905, USA.
- Department of Biomedical Engineering, Mayo Clinic, Rochester, MN, 55905, USA.
| | - Yoonbae Oh
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN, 55905, USA.
- Department of Biomedical Engineering, Mayo Clinic, Rochester, MN, 55905, USA.
| |
Collapse
|
3
|
Benatti BM, Adiletta A, Sgadò P, Malgaroli A, Ferro M, Lamanna J. Epigenetic Modifications and Neuroplasticity in the Pathogenesis of Depression: A Focus on Early Life Stress. Behav Sci (Basel) 2024; 14:882. [PMID: 39457754 PMCID: PMC11504006 DOI: 10.3390/bs14100882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 09/24/2024] [Accepted: 09/27/2024] [Indexed: 10/28/2024] Open
Abstract
Major depressive disorder (MDD) is a debilitating mental illness, and it is considered to be one of the leading causes of disability globally. The etiology of MDD is multifactorial, involving an interplay between biological, psychological, and social factors. Early life represents a critical period for development. Exposure to adverse childhood experiences is a major contributor to the global burden of disease and disability, doubling the risk of developing MDD later in life. Evidence suggests that stressful events experienced during that timeframe play a major role in the emergence of MDD, leading to epigenetic modifications, which might, in turn, influence brain structure, function, and behavior. Neuroplasticity seems to be a primary pathogenetic mechanism of MDD, and, similarly to epigenetic mechanisms, it is particularly sensitive to stress in the early postnatal period. In this review, we will collect and discuss recent studies supporting the role of epigenetics and neuroplasticity in the pathogenesis of MDD, with a focus on early life stress (ELS). We believe that understanding the epigenetic mechanisms by which ELS affects neuroplasticity offers potential pathways for identifying novel therapeutic targets for MDD, ultimately aiming to improve treatment outcomes for this debilitating disorder.
Collapse
Affiliation(s)
- Bianca Maria Benatti
- Center for Behavioral Neuroscience and Communication (BNC), Vita-Salute San Raffaele University, 20132 Milan, Italy; (B.M.B.); (M.F.)
| | - Alice Adiletta
- Center for Mind/Brain Sciences, University of Trento, 38068 Rovereto, Italy; (A.A.); (P.S.)
| | - Paola Sgadò
- Center for Mind/Brain Sciences, University of Trento, 38068 Rovereto, Italy; (A.A.); (P.S.)
| | - Antonio Malgaroli
- Center for Behavioral Neuroscience and Communication (BNC), Vita-Salute San Raffaele University, 20132 Milan, Italy; (B.M.B.); (M.F.)
- Faculty of Psychology, Vita-Salute San Raffaele University, 20132 Milan, Italy
- Clinical Center Tourette Syndrome, IRCCS Ospedale San Raffaele, 20127 Milan, Italy
| | - Mattia Ferro
- Center for Behavioral Neuroscience and Communication (BNC), Vita-Salute San Raffaele University, 20132 Milan, Italy; (B.M.B.); (M.F.)
- Department of Psychology, Sigmund Freud Private University, 20143 Milan, Italy
| | - Jacopo Lamanna
- Center for Behavioral Neuroscience and Communication (BNC), Vita-Salute San Raffaele University, 20132 Milan, Italy; (B.M.B.); (M.F.)
- Clinical Center Tourette Syndrome, IRCCS Ospedale San Raffaele, 20127 Milan, Italy
| |
Collapse
|
4
|
Ma L, Tao Q, Dang J, Sun J, Niu X, Zhang M, Kang Y, Wang W, Cheng J, Han S, Zhang Y. Altered local intrinsic neural activity and molecular architecture in internet use disorders. Brain Res Bull 2024; 216:111052. [PMID: 39173776 DOI: 10.1016/j.brainresbull.2024.111052] [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: 05/08/2024] [Revised: 07/20/2024] [Accepted: 08/17/2024] [Indexed: 08/24/2024]
Abstract
BACKGROUND Internet gaming disorder (IGD) is mainly characterized by its core dysfunction in higher-order brain cortices involved in inhibitory control, whose neurobiological basis remains unclear. Then, we will investigate local intrinsic neural activity (INA) alterations in IGD, ascertain whether these potential alterations are related to clinical characteristics, and further explore the underlying molecular architecture. METHOD In this study, we performed the fractional amplitude of low-frequency fluctuation (fALFF) and regional homogeneity (ReHo) derived from resting-state functional magnetic resonance imaging (rs-fMRI) to explore the impact of IGD on local INA. Correlation analysis revealed the relationship between ReHo and fALFF in terms of group differences and clinical characteristics. Moreover, correlations between fALFF, ReHo, and PET- and SPECT-driven maps were investigated to elucidate the specific molecular architecture alternations in IGD. Finally, receiver operating characteristic curve (ROC) analysis was used to show the potential abilities of fALFF and ReHo in distinguishing individuals with IGD (IGDs) from healthy controls (HCs). RESULT Compared with HCs, IGDs revealed increased ReHo and fALFF in the prefrontal cortex. Significantly decreased ReHo was observed in the temporal lobe, occipital lobe, and cerebellum. In addition, the ReHo values in the cerebellum_7b_R were positively correlated with internet addiction severity. ROC curve analysis showed that ReHo and fALFF-altered brain regions could effectively distinguish IGDs from HCs. More importantly, cross-modal correlations revealed local INA changes in brain regions associated with the monoamine neurotransmitter system and the less studied cholinergic/GABAergic system. CONCLUSION These results suggest that local functional impairments are shown in the audiovisual and inhibitory control circuits in IGDs. This may be associated with underlying neurotransmitter system alterations. Therefore, this study provides the possibility of GABAergic receptor agonists and cholinergic receptor inhibitors for the treatment of IGD.
Collapse
Affiliation(s)
- Longyao Ma
- Department of Magnetic Resonance Imaging, the First Affiliated Hospital of Zhengzhou University, China; Zhengzhou Key Laboratory of Brain Function And Cognitive Magnetic Resonance Imaging, China
| | - Qiuying Tao
- Department of Magnetic Resonance Imaging, the First Affiliated Hospital of Zhengzhou University, China; Zhengzhou Key Laboratory of Brain Function And Cognitive Magnetic Resonance Imaging, China
| | - Jinghan Dang
- Department of Magnetic Resonance Imaging, the First Affiliated Hospital of Zhengzhou University, China; Zhengzhou Key Laboratory of Brain Function And Cognitive Magnetic Resonance Imaging, China
| | - Jieping Sun
- Department of Magnetic Resonance Imaging, the First Affiliated Hospital of Zhengzhou University, China; Zhengzhou Key Laboratory of Brain Function And Cognitive Magnetic Resonance Imaging, China
| | - Xiaoyu Niu
- Department of Magnetic Resonance Imaging, the First Affiliated Hospital of Zhengzhou University, China; Zhengzhou Key Laboratory of Brain Function And Cognitive Magnetic Resonance Imaging, China
| | - Mengzhe Zhang
- Department of Magnetic Resonance Imaging, the First Affiliated Hospital of Zhengzhou University, China; Zhengzhou Key Laboratory of Brain Function And Cognitive Magnetic Resonance Imaging, China
| | - Yimeng Kang
- Department of Magnetic Resonance Imaging, the First Affiliated Hospital of Zhengzhou University, China; Zhengzhou Key Laboratory of Brain Function And Cognitive Magnetic Resonance Imaging, China
| | - Weijian Wang
- Department of Magnetic Resonance Imaging, the First Affiliated Hospital of Zhengzhou University, China; Zhengzhou Key Laboratory of Brain Function And Cognitive Magnetic Resonance Imaging, China
| | - Jingliang Cheng
- Department of Magnetic Resonance Imaging, the First Affiliated Hospital of Zhengzhou University, China; Zhengzhou Key Laboratory of Brain Function And Cognitive Magnetic Resonance Imaging, China
| | - Shaoqiang Han
- Department of Magnetic Resonance Imaging, the First Affiliated Hospital of Zhengzhou University, China; Zhengzhou Key Laboratory of Brain Function And Cognitive Magnetic Resonance Imaging, China
| | - Yong Zhang
- Department of Magnetic Resonance Imaging, the First Affiliated Hospital of Zhengzhou University, China; Zhengzhou Key Laboratory of Brain Function And Cognitive Magnetic Resonance Imaging, China.
| |
Collapse
|
5
|
Allen SJ, Morishita H. Local and long-range input balance: A framework for investigating frontal cognitive circuit maturation in health and disease. SCIENCE ADVANCES 2024; 10:eadh3920. [PMID: 39292771 PMCID: PMC11409946 DOI: 10.1126/sciadv.adh3920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 08/12/2024] [Indexed: 09/20/2024]
Abstract
Frontal cortical circuits undergo prolonged maturation across childhood and adolescence; however, it remains unknown what specific changes are occurring at the circuit level to establish adult cognitive function. With the recent advent of circuit dissection techniques, it is now feasible to examine circuit-specific changes in connectivity, activity, and function in animal models. Here, we propose that the balance of local and long-range inputs onto frontal cognitive circuits is an understudied metric of circuit maturation. This review highlights research on a frontal-sensory attention circuit that undergoes refinement of local/long-range connectivity, regulated by circuit activity and neuromodulatory signaling, and evaluates how this process may occur generally in the frontal cortex to support adult cognitive behavior. Notably, this balance can be bidirectionally disrupted through various mechanisms relevant to psychiatric disorders. Pharmacological or environmental interventions to normalize or reset the local and long-range balance could hold great therapeutic promise to prevent or rescue cognitive deficits.
Collapse
Affiliation(s)
- Samuel J. Allen
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
- Department of Ophthalmology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
- Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
| | - Hirofumi Morishita
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
- Department of Ophthalmology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
- Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
| |
Collapse
|
6
|
Pedersen CN, Yang F, Ita S, Xu Y, Akunuri R, Trampari S, Neumann CMT, Desdorf LM, Schiøtt B, Salvino JM, Mortensen OV, Nissen P, Shahsavar A. Cryo-EM structure of the dopamine transporter with a novel atypical non-competitive inhibitor bound to the orthosteric site. J Neurochem 2024; 168:2043-2055. [PMID: 39010681 PMCID: PMC11449642 DOI: 10.1111/jnc.16179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Revised: 06/28/2024] [Accepted: 07/01/2024] [Indexed: 07/17/2024]
Abstract
The regulation of dopamine (DA) removal from the synaptic cleft is a crucial process in neurotransmission and is facilitated by the sodium- and chloride-coupled dopamine transporter DAT. Psychostimulant drugs, cocaine, and amphetamine, both block the uptake of DA, while amphetamine also triggers the release of DA. As a result, they prolong or even amplify neurotransmitter signaling. Atypical inhibitors of DAT lack cocaine-like rewarding effects and offer a promising strategy for the treatment of drug use disorders. Here, we present the 3.2 Å resolution cryo-electron microscopy structure of the Drosophila melanogaster dopamine transporter (dDAT) in complex with the atypical non-competitive inhibitor AC-4-248. The inhibitor partially binds at the central binding site, extending into the extracellular vestibule, and locks the transporter in an outward open conformation. Our findings propose mechanisms for the non-competitive inhibition of DAT and attenuation of cocaine potency by AC-4-248 and provide a basis for the rational design of more efficacious atypical inhibitors.
Collapse
Affiliation(s)
- Clara Nautrup Pedersen
- DANDRITE, Nordic EMBL Partnership for Molecular Medicine, Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | - Fuyu Yang
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Samantha Ita
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Yibin Xu
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA, USA
| | | | - Sofia Trampari
- DANDRITE, Nordic EMBL Partnership for Molecular Medicine, Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | - Caroline Marie Teresa Neumann
- DANDRITE, Nordic EMBL Partnership for Molecular Medicine, Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | | | - Birgit Schiøtt
- Department of Chemistry, Aarhus University, Aarhus, Denmark
| | | | - Ole Valente Mortensen
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Poul Nissen
- DANDRITE, Nordic EMBL Partnership for Molecular Medicine, Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | - Azadeh Shahsavar
- DANDRITE, Nordic EMBL Partnership for Molecular Medicine, Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
7
|
Müller TE, Dos Santos MM, Ferreira SA, Claro MT, de Macedo GT, Fontana BD, Barbosa NV. Negative impacts of social isolation on behavior and neuronal functions are recovered after short-term social reintroduction in zebrafish. Prog Neuropsychopharmacol Biol Psychiatry 2024; 134:111038. [PMID: 38810717 DOI: 10.1016/j.pnpbp.2024.111038] [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: 11/10/2023] [Revised: 05/16/2024] [Accepted: 05/22/2024] [Indexed: 05/31/2024]
Abstract
Recently, social isolation measures were crucial to prevent the spread of the coronavirus pandemic. However, the lack of social interactions affected the population mental health and may have long-term consequences on behavior and brain functions. Here, we evaluated the behavioral, physiological, and molecular effects of a social isolation (SI) in adult zebrafish, and whether the animals recover such changes after their reintroduction to the social environment. Fish were submitted to 12 days of SI, and then reintroduced to social context (SR). Behavioral analyses to evaluate locomotion, anxiety-like and social-related behaviors were performed after SI protocol, and 3 and 6 days after SR. Cortisol and transcript levels from genes involved in neuronal homeostasis (c-fos, egr, bdnf), and serotonergic (5-HT) and dopaminergic (DA) neurotransmission (thp, th) were also measured. SI altered social behaviors in zebrafish such as aggression, social preference, and shoaling. Fish submitted to SI also presented changes in the transcript levels of genes related to neural activity, and 5-HT/DA signaling. Interestingly, most of the behavioral and molecular changes induced by SI were not found again 6 days after SR. Thus, we highlight that SR of zebrafish to their conspecifics played a positive role in social behaviors and in the expression of genes involved in different neuronal signaling pathways that were altered after 12 days of SI. This study brings unprecedented data on the effects of SR in the recovery from SI neurobehavioral alterations, and reinforces the role of zebrafish as a translational model for understanding the neurobiological mechanisms adjacent to SI and resocialization.
Collapse
Affiliation(s)
- Talise E Müller
- Laboratory of Toxicological Biochemistry, Department of Biochemistry and Molecular Biology, Center of Natural and Exact Sciences, Federal University of Santa Maria. 1000 Roraima Avenue, Santa Maria, RS 97105-900, Brazil.; Graduate Program in Biological Sciences: Toxicological Biochemistry, Federal University of Santa Maria. 1000 Roraima Avenue, Santa Maria, RS 97105-900, Brazil..
| | - Matheus M Dos Santos
- Laboratory of Toxicological Biochemistry, Department of Biochemistry and Molecular Biology, Center of Natural and Exact Sciences, Federal University of Santa Maria. 1000 Roraima Avenue, Santa Maria, RS 97105-900, Brazil.; Graduate Program in Biological Sciences: Toxicological Biochemistry, Federal University of Santa Maria. 1000 Roraima Avenue, Santa Maria, RS 97105-900, Brazil
| | - Sabrina A Ferreira
- Laboratory of Toxicological Biochemistry, Department of Biochemistry and Molecular Biology, Center of Natural and Exact Sciences, Federal University of Santa Maria. 1000 Roraima Avenue, Santa Maria, RS 97105-900, Brazil.; Graduate Program in Biological Sciences: Toxicological Biochemistry, Federal University of Santa Maria. 1000 Roraima Avenue, Santa Maria, RS 97105-900, Brazil
| | - Mariana T Claro
- Laboratory of Toxicological Biochemistry, Department of Biochemistry and Molecular Biology, Center of Natural and Exact Sciences, Federal University of Santa Maria. 1000 Roraima Avenue, Santa Maria, RS 97105-900, Brazil.; Graduate Program in Biological Sciences: Toxicological Biochemistry, Federal University of Santa Maria. 1000 Roraima Avenue, Santa Maria, RS 97105-900, Brazil
| | - Gabriel T de Macedo
- Laboratory of Toxicological Biochemistry, Department of Biochemistry and Molecular Biology, Center of Natural and Exact Sciences, Federal University of Santa Maria. 1000 Roraima Avenue, Santa Maria, RS 97105-900, Brazil.; Graduate Program in Biological Sciences: Toxicological Biochemistry, Federal University of Santa Maria. 1000 Roraima Avenue, Santa Maria, RS 97105-900, Brazil
| | - Barbara D Fontana
- Department of Biological Sciences, Wayne State University, Detroit, MI 48202, USA
| | - Nilda V Barbosa
- Laboratory of Toxicological Biochemistry, Department of Biochemistry and Molecular Biology, Center of Natural and Exact Sciences, Federal University of Santa Maria. 1000 Roraima Avenue, Santa Maria, RS 97105-900, Brazil.; Graduate Program in Biological Sciences: Toxicological Biochemistry, Federal University of Santa Maria. 1000 Roraima Avenue, Santa Maria, RS 97105-900, Brazil..
| |
Collapse
|
8
|
Yang H, Chen Y, Tao Q, Shi W, Tian Y, Wei Y, Li S, Zhang Y, Han S, Cheng J. Integrative molecular and structural neuroimaging analyses of the interaction between depression and age of onset: A multimodal magnetic resonance imaging study. Prog Neuropsychopharmacol Biol Psychiatry 2024; 134:111052. [PMID: 38871019 DOI: 10.1016/j.pnpbp.2024.111052] [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: 04/08/2024] [Revised: 05/30/2024] [Accepted: 06/10/2024] [Indexed: 06/15/2024]
Abstract
Depression is a neurodevelopmental disorder that exhibits progressive gray matter volume (GMV) atrophy. Research indicates that brain development is influential in depression-induced GMV alterations. However, the interaction between depression and age of onset is not well understood by the underlying molecular and neuropathological mechanisms. Thus, 152 first-episode depression individuals and matched 130 healthy controls (HCs) were recruited to undergo T1-weighted high-resolution magnetic resonance imaging for this study. By two-way ANOVA, age and diagnosis were used as factors when analyzing the interaction of GMV in the participants. Then, spatial correlations between neurotransmitter maps and factor-related volume maps are established. Results illustrate a pronounced antagonistic interaction between depression and age of onset in the right insula, superior temporal gyrus, anterior cingulate gyrus, and orbitofrontal gyrus. Depression-caused reductions in GMV are mainly distributed in thalamic-limbic-cortical regions, regardless of age. For the main effect of age, adults exhibit brain atrophy in frontal, cerebellum, parietal, and temporal lobe structures. Cross-modal correlations showed that GMV changes in the interactive regions were linked with the serotonergic system and dopaminergic systems. Summarily, our results reveal the interaction between depression and age of onset in neurobiological mechanisms, which provide hints for future treatment of different ages of depression.
Collapse
Affiliation(s)
- Huiting Yang
- Department of Magnetic Resonance Imaging, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China; Zhengzhou Key Laboratory of brain function and cognitive magnetic resonance imaging, Zhengzhou, China; Henan Engineering Technology Research Center for detection and application of brain function, Zhengzhou, China; Henan Engineering Research Center of medical imaging intelligent diagnosis and treatment, Zhengzhou, China; Henan key laboratory of imaging intelligence research, Zhengzhou, China; Henan Engineering Research Center of Brain Function Development and Application, Zhengzhou, China
| | - Yuan Chen
- Department of Magnetic Resonance Imaging, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China; Zhengzhou Key Laboratory of brain function and cognitive magnetic resonance imaging, Zhengzhou, China; Henan Engineering Technology Research Center for detection and application of brain function, Zhengzhou, China; Henan Engineering Research Center of medical imaging intelligent diagnosis and treatment, Zhengzhou, China; Henan key laboratory of imaging intelligence research, Zhengzhou, China; Henan Engineering Research Center of Brain Function Development and Application, Zhengzhou, China
| | - Qiuying Tao
- Department of Magnetic Resonance Imaging, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China; Zhengzhou Key Laboratory of brain function and cognitive magnetic resonance imaging, Zhengzhou, China; Henan Engineering Technology Research Center for detection and application of brain function, Zhengzhou, China; Henan Engineering Research Center of medical imaging intelligent diagnosis and treatment, Zhengzhou, China; Henan key laboratory of imaging intelligence research, Zhengzhou, China; Henan Engineering Research Center of Brain Function Development and Application, Zhengzhou, China
| | - Wenqing Shi
- Department of Magnetic Resonance Imaging, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China; Zhengzhou Key Laboratory of brain function and cognitive magnetic resonance imaging, Zhengzhou, China; Henan Engineering Technology Research Center for detection and application of brain function, Zhengzhou, China; Henan Engineering Research Center of medical imaging intelligent diagnosis and treatment, Zhengzhou, China; Henan key laboratory of imaging intelligence research, Zhengzhou, China; Henan Engineering Research Center of Brain Function Development and Application, Zhengzhou, China
| | - Ya Tian
- Department of Magnetic Resonance Imaging, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China; Zhengzhou Key Laboratory of brain function and cognitive magnetic resonance imaging, Zhengzhou, China; Henan Engineering Technology Research Center for detection and application of brain function, Zhengzhou, China; Henan Engineering Research Center of medical imaging intelligent diagnosis and treatment, Zhengzhou, China; Henan key laboratory of imaging intelligence research, Zhengzhou, China; Henan Engineering Research Center of Brain Function Development and Application, Zhengzhou, China
| | - Yarui Wei
- Department of Magnetic Resonance Imaging, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China; Zhengzhou Key Laboratory of brain function and cognitive magnetic resonance imaging, Zhengzhou, China; Henan Engineering Technology Research Center for detection and application of brain function, Zhengzhou, China; Henan Engineering Research Center of medical imaging intelligent diagnosis and treatment, Zhengzhou, China; Henan key laboratory of imaging intelligence research, Zhengzhou, China; Henan Engineering Research Center of Brain Function Development and Application, Zhengzhou, China
| | - Shuying Li
- Department of Psychiatry, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yong Zhang
- Department of Magnetic Resonance Imaging, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China; Zhengzhou Key Laboratory of brain function and cognitive magnetic resonance imaging, Zhengzhou, China; Henan Engineering Technology Research Center for detection and application of brain function, Zhengzhou, China; Henan Engineering Research Center of medical imaging intelligent diagnosis and treatment, Zhengzhou, China; Henan key laboratory of imaging intelligence research, Zhengzhou, China; Henan Engineering Research Center of Brain Function Development and Application, Zhengzhou, China.
| | - Shaoqiang Han
- Department of Magnetic Resonance Imaging, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China; Zhengzhou Key Laboratory of brain function and cognitive magnetic resonance imaging, Zhengzhou, China; Henan Engineering Technology Research Center for detection and application of brain function, Zhengzhou, China; Henan Engineering Research Center of medical imaging intelligent diagnosis and treatment, Zhengzhou, China; Henan key laboratory of imaging intelligence research, Zhengzhou, China; Henan Engineering Research Center of Brain Function Development and Application, Zhengzhou, China.
| | - Jingliang Cheng
- Department of Magnetic Resonance Imaging, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China; Zhengzhou Key Laboratory of brain function and cognitive magnetic resonance imaging, Zhengzhou, China; Henan Engineering Technology Research Center for detection and application of brain function, Zhengzhou, China; Henan Engineering Research Center of medical imaging intelligent diagnosis and treatment, Zhengzhou, China; Henan key laboratory of imaging intelligence research, Zhengzhou, China; Henan Engineering Research Center of Brain Function Development and Application, Zhengzhou, China.
| |
Collapse
|
9
|
Zhang K, He L, Li Z, Ding R, Han X, Chen B, Cao G, Ye JH, Li T, Fu R. Bridging Neurobiological Insights and Clinical Biomarkers in Postpartum Depression: A Narrative Review. Int J Mol Sci 2024; 25:8835. [PMID: 39201521 PMCID: PMC11354679 DOI: 10.3390/ijms25168835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Revised: 08/10/2024] [Accepted: 08/13/2024] [Indexed: 09/02/2024] Open
Abstract
Postpartum depression (PPD) affects 174 million women worldwide and is characterized by profound sadness, anxiety, irritability, and debilitating fatigue, which disrupt maternal caregiving and the mother-infant relationship. Limited pharmacological interventions are currently available. Our understanding of the neurobiological pathophysiology of PPD remains incomplete, potentially hindering the development of novel treatment strategies. Recent hypotheses suggest that PPD is driven by a complex interplay of hormonal changes, neurotransmitter imbalances, inflammation, genetic factors, psychosocial stressors, and hypothalamic-pituitary-adrenal (HPA) axis dysregulation. This narrative review examines recent clinical studies on PPD within the past 15 years, emphasizing advancements in neuroimaging findings and blood biomarker detection. Additionally, we summarize recent laboratory work using animal models to mimic PPD, focusing on hormone withdrawal, HPA axis dysfunction, and perinatal stress theories. We also revisit neurobiological results from several brain regions associated with negative emotions, such as the amygdala, prefrontal cortex, hippocampus, and striatum. These insights aim to improve our understanding of PPD's neurobiological mechanisms, guiding future research for better early detection, prevention, and personalized treatment strategies for women affected by PPD and their families.
Collapse
Affiliation(s)
- Keyi Zhang
- Department of Anatomy, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen 518107, China; (K.Z.); (L.H.); (Z.L.); (R.D.); (X.H.); (B.C.); (G.C.)
| | - Lingxuan He
- Department of Anatomy, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen 518107, China; (K.Z.); (L.H.); (Z.L.); (R.D.); (X.H.); (B.C.); (G.C.)
| | - Zhuoen Li
- Department of Anatomy, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen 518107, China; (K.Z.); (L.H.); (Z.L.); (R.D.); (X.H.); (B.C.); (G.C.)
| | - Ruxuan Ding
- Department of Anatomy, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen 518107, China; (K.Z.); (L.H.); (Z.L.); (R.D.); (X.H.); (B.C.); (G.C.)
| | - Xiaojiao Han
- Department of Anatomy, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen 518107, China; (K.Z.); (L.H.); (Z.L.); (R.D.); (X.H.); (B.C.); (G.C.)
| | - Bingqing Chen
- Department of Anatomy, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen 518107, China; (K.Z.); (L.H.); (Z.L.); (R.D.); (X.H.); (B.C.); (G.C.)
| | - Guoxin Cao
- Department of Anatomy, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen 518107, China; (K.Z.); (L.H.); (Z.L.); (R.D.); (X.H.); (B.C.); (G.C.)
| | - Jiang-Hong Ye
- Department of Anesthesiology, Pharmacology, Physiology & Neuroscience, Rutgers, The State University of New Jersey, New Jersey Medical School, Newark, NJ 07103, USA;
| | - Tian Li
- Department of Gynecology and Obstetrics, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen 518107, China
| | - Rao Fu
- Department of Anatomy, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen 518107, China; (K.Z.); (L.H.); (Z.L.); (R.D.); (X.H.); (B.C.); (G.C.)
| |
Collapse
|
10
|
Jamal T, Yan X, Lantyer ADS, Ter Horst JG, Celikel T. Experience-dependent regulation of dopaminergic signaling in the somatosensory cortex. Prog Neurobiol 2024; 239:102630. [PMID: 38834131 DOI: 10.1016/j.pneurobio.2024.102630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 05/04/2024] [Accepted: 05/10/2024] [Indexed: 06/06/2024]
Abstract
Dopamine critically influences reward processing, sensory perception, and motor control. Yet, the modulation of dopaminergic signaling by sensory experiences is not fully delineated. Here, by manipulating sensory experience using bilateral single-row whisker deprivation, we demonstrated that gene transcription in the dopaminergic signaling pathway (DSP) undergoes experience-dependent plasticity in both granular and supragranular layers of the primary somatosensory (barrel) cortex (S1). Sensory experience and deprivation compete for the regulation of DSP transcription across neighboring cortical columns, and sensory deprivation-induced changes in DSP are topographically constrained. These changes in DSP extend beyond cortical map plasticity and influence neuronal information processing. Pharmacological regulation of D2 receptors, a key component of DSP, revealed that D2 receptor activation suppresses excitatory neuronal excitability, hyperpolarizes the action potential threshold, and reduces the instantaneous firing rate. These findings suggest that the dopaminergic drive originating from midbrain dopaminergic neurons, targeting the sensory cortex, is subject to experience-dependent regulation and might create a regulatory feedback loop for modulating sensory processing. Finally, using topological gene network analysis and mutual information, we identify the molecular hubs of experience-dependent plasticity of DSP. These findings provide new insights into the mechanisms by which sensory experience shapes dopaminergic signaling in the brain and might help unravel the sensory deficits observed after dopamine depletion.
Collapse
Affiliation(s)
- Tousif Jamal
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, the Netherlands
| | - Xuan Yan
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, the Netherlands
| | | | - Judith G Ter Horst
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, the Netherlands
| | - Tansu Celikel
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, the Netherlands; School of Psychology, Georgia Institute of Technology, Atlanta, GA, USA.
| |
Collapse
|
11
|
Aziz HC, Mangieri RA. Sex differences in membrane properties and cellular excitability of dopamine D1 receptor-expressing neurons within the shell of the nucleus accumbens of pre- and mid-adolescent mice. Biol Sex Differ 2024; 15:54. [PMID: 39003495 PMCID: PMC11245857 DOI: 10.1186/s13293-024-00631-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Accepted: 07/02/2024] [Indexed: 07/15/2024] Open
Abstract
BACKGROUND The transition from childhood to adulthood, or adolescence, a developmental stage, is characterized by psychosocial and biological changes. The nucleus accumbens (NAc), a striatal brain region composed of the core (NAcC) and shell (NAcSh), has been linked to risk-taking behavior and implicated in reward seeking and evaluation. Most neurons in the NAc are medium spiny neurons (MSNs) that express dopamine D1 receptors (D1R +) and/or dopamine D2 receptors (D2R +). Changes in dopaminergic and glutamatergic systems occur during adolescence and converge in the NAc. While there are previous investigations into sex differences in membrane excitability and synaptic glutamate transmission in both subdivisions of the NAc, to our knowledge, none have specified NAcSh D1R + MSNs from mice during pre- and mid-adolescence. METHODS Sagittal brain slices containing the NAc were prepared from B6.Cg-Tg(Drd1a-tdTomato)6Calak/J mice of both sexes from postnatal days 21-25 and 35-47, representing pre- and mid-adolescence, respectively. Whole-cell electrophysiology recordings were collected from NAcSh D1R + MSNs in the form of membrane-voltage responses to current injections, to assess membrane properties and action potential waveform characteristics, and spontaneous excitatory postsynaptic currents (sEPSCs) to assess glutamatergic synaptic activity. RESULTS Relative to pre-adolescent males, pre-adolescent female NAcSh D1R + MSNs exhibited a less hyperpolarized resting membrane potential, increased input resistance, and smaller action potential afterhyperpolarization amplitudes. During mid-adolescence, decreased input resistance and a shorter action potential duration in females were the only sex differences observed. CONCLUSIONS Taken together, our results indicate that NAcSh D1R + MSNs in mice exhibit sex differences in membrane properties and AP waveform during pre-adolescence that are overall indicative of increased cellular excitability in females and are suggestive of possible sex differences in glycine receptors, inwardly-rectifying potassium channels, and large conductance voltage-gated potassium channels. These differences do not appear to persist into mid-adolescence, when sex was observed to affect input resistance oppositely to that of pre-adolescence and AP waveform in a manner suggestive of differences in voltage-gated potassium channels.
Collapse
Affiliation(s)
- Heather C Aziz
- Division of Pharmacology and Toxicology, College of Pharmacy, The University of Texas at Austin, 2409 University Avenue, Austin, TX, 78712, USA.
| | - Regina A Mangieri
- Division of Pharmacology and Toxicology, College of Pharmacy, The University of Texas at Austin, 2409 University Avenue, Austin, TX, 78712, USA
| |
Collapse
|
12
|
Obukhova AL, Khabarova MY, Semenova MN, Starunov VV, Voronezhskaya EE, Ivashkin EG. Spontaneous intersibling polymorphism in the development of dopaminergic neuroendocrine cells in sea urchin larvae: impacts on the expansion of marine benthic species. Front Neurosci 2024; 18:1348999. [PMID: 38660226 PMCID: PMC11039814 DOI: 10.3389/fnins.2024.1348999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Accepted: 03/27/2024] [Indexed: 04/26/2024] Open
Abstract
Introduction The plasticity of the nervous system plays a crucial role in shaping adaptive neural circuits and corresponding animal behaviors. Understanding the mechanisms underlying neural plasticity during development and its implications for animal adaptation constitutes an intriguing area of research. Sea urchin larvae offer a fascinating subject for investigation due to their remarkable evolutionary and ecological diversity, as well as their diverse developmental forms and behavioral patterns. Materials and methods We conducted immunochemical and histochemical analyses of serotonin-containing (5-HT-neurons) and dopamine-containing (DA-positive) neurons to study their developmental dynamics in two sea urchin species: Mesocentrotus nudus and Paracentrotus lividus. Our approach involved detailed visualization of 5-HT- and DA-positive neurons at gastrula-pluteus stages, coupled with behavioral assays to assess larval upward and downward swimming in the water column, with a focus on correlating cell numbers with larval swimming ability. Results The study reveals a heterochronic polymorphism in the appearance of post-oral DA-positive neuroendocrine cells and confirms the stable differentiation pattern of apical 5-HT neurons in larvae of both species. Notably, larvae of the same age exhibit a two- to four-fold difference in DA neurons. An increased number of DA neurons and application of dopamine positively correlate with larval downward swimming, whereas 5-HT-neurons and serotonin application induce upward swimming. The ratio of 5-HT/DA neurons determines the stage-dependent vertical distribution of larvae within the water column. Consequently, larvae from the same generation with a higher number of DA-positive neurons tend to remain at the bottom compared to those with fewer DA-positive neurons. Discussion The proportion of 5-HT and DA neurons within larvae of the same age underlies the different potentials of individuals for upward and downward swimming. A proposed model illustrates how coordination in humoral regulation, based on heterochrony in DA-positive neuroendocrine cell differentiation, influences larval behavior, mitigates competition between siblings, and ensures optimal population expansion. The study explores the evolutionary and ecological implications of these neuroendocrine adaptations in marine species.
Collapse
Affiliation(s)
- Alexandra L. Obukhova
- Koltsov Institute of Developmental Biology, Russian Academy Sciences, Moscow, Russia
| | - Marina Yu. Khabarova
- Koltsov Institute of Developmental Biology, Russian Academy Sciences, Moscow, Russia
| | - Marina N. Semenova
- Koltsov Institute of Developmental Biology, Russian Academy Sciences, Moscow, Russia
| | - Viktor V. Starunov
- Department of Invertebrate Zoology, St-Petersburg State University, Saint Petersburg, Russia
- Zoological Institute, Russian Academy Sciences, Saint Petersburg, Russia
| | | | - Evgeny G. Ivashkin
- Koltsov Institute of Developmental Biology, Russian Academy Sciences, Moscow, Russia
- Severtsov Institute of Ecology and Evolution, Russian Academy of Sciences, Moscow, Russia
| |
Collapse
|
13
|
Homberg JR, Brivio P, Greven CU, Calabrese F. Individuals being high in their sensitivity to the environment: Are sensitive period changes in play? Neurosci Biobehav Rev 2024; 159:105605. [PMID: 38417743 DOI: 10.1016/j.neubiorev.2024.105605] [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: 10/11/2023] [Revised: 02/13/2024] [Accepted: 02/25/2024] [Indexed: 03/01/2024]
Abstract
All individuals on planet earth are sensitive to the environment, but some more than others. These individual differences in sensitivity to environments are seen across many animal species including humans, and can influence personalities as well as vulnerability and resilience to mental disorders. Yet, little is known about the underlying brain mechanisms. Key genes that contribute to individual differences in environmental sensitivity are the serotonin transporter, dopamine D4 receptor and brain-derived neurotrophic factor genes. By synthesizing neurodevelopmental findings of these genetic factors, and discussing them through the lens of mechanisms related to sensitive periods, which are phases of heightened neuronal plasticity during which a certain network is being finetuned by experiences, we propose that these genetic factors delay but extend postnatal sensitive periods. This may explain why sensitive individuals show behavioral features that are characteristic of a young brain state at the level of sensory information processing, such as reduced filtering or blockade of irrelevant information, resulting in a sensory processing system that 'keeps all options open'.
Collapse
Affiliation(s)
- Judith R Homberg
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition and Behavior, Radboud University Medical Center, Nijmegen, the Netherlands.
| | - Paola Brivio
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università degli Studi di Milano, Milan, Italy
| | - Corina U Greven
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition and Behavior, Radboud University Medical Center, Nijmegen, the Netherlands; Karakter Child and Adolescent Psychiatry University Center, Nijmegen, the Netherlands; King's College London, Institute of Psychiatry, Psychology and Neuroscience, Social, Genetic and Developmental Psychiatry Center, London, United Kingdom
| | - Francesca Calabrese
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università degli Studi di Milano, Milan, Italy
| |
Collapse
|
14
|
Lu X, Song Y, Wang J, Cai Y, Peng S, Lin J, Lai B, Sun J, Liu T, Chen G, Xing L. Developmental dopaminergic signaling modulates neural circuit formation and contributes to autism spectrum disorder (ASD)-related phenotypes. THE AMERICAN JOURNAL OF PATHOLOGY 2024:S0002-9440(24)00086-5. [PMID: 38492733 DOI: 10.1016/j.ajpath.2024.02.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Revised: 12/25/2023] [Accepted: 02/06/2024] [Indexed: 03/18/2024]
Abstract
Autism spectrum disorder (ASD) is a prevalent neurodevelopmental disorder with a complex etiology. Recent evidence suggests that dopamine plays a crucial role in neural development. However, it remains unclear whether and how disrupted dopaminergic signaling during development contributes to ASD. In this study, human brain RNA-seq transcriptome analysis revealed a significant correlation between changes in dopaminergic signaling pathways and neural developmental signaling in ASD patients. In the zebrafish model, disrupted developmental dopaminergic signaling led to neural circuit abnormalities and behavior reminiscent of autism. Dopaminergic signaling may impact neuronal specification by potentially modulating integrins. These findings shed light on the mechanisms underlying the link between disrupted developmental dopamine signaling and ASD, and they point to the possibility of targeting dopaminergic signaling in early development for ASD treatment.
Collapse
Affiliation(s)
- Xiaojuan Lu
- Key Laboratory of Neuroregeneration of Jiangsu and the Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong 226001, China
| | - Yixing Song
- Key Laboratory of Neuroregeneration of Jiangsu and the Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong 226001, China
| | - Jiaqi Wang
- Medical School of Nantong University, Nantong, Jiangsu Province, 226001, China
| | - Yunyun Cai
- Medical School of Nantong University, Nantong, Jiangsu Province, 226001, China
| | - Siwan Peng
- Key Laboratory of Neuroregeneration of Jiangsu and the Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong 226001, China
| | - Jiaqi Lin
- Medical School of Nantong University, Nantong, Jiangsu Province, 226001, China
| | - Biqin Lai
- Key Laboratory for Stem Cells and Tissue Engineering (Sun Yat-sen University), Ministry of Education, Co-innovation Center of Neuroregeneration, Jiangsu Province, 226001, China
| | - Junjie Sun
- Key Laboratory of Neuroregeneration of Jiangsu and the Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong 226001, China
| | - Tianqing Liu
- NICM Health Research Institute, Western Sydney University, Westmead 2145, Australia
| | - Gang Chen
- Key Laboratory of Neuroregeneration of Jiangsu and the Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong 226001, China; Medical School of Nantong University, Nantong, Jiangsu Province, 226001, China.
| | - Lingyan Xing
- Key Laboratory of Neuroregeneration of Jiangsu and the Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong 226001, China.
| |
Collapse
|
15
|
Ramya R, Venkatesh CR, Shyamala BV. olf413 an octopamine biogenesis pathway gene is required for axon growth and pathfinding during embryonic nervous system development in Drosophila melanogaster. BMC Res Notes 2024; 17:46. [PMID: 38326892 PMCID: PMC10848397 DOI: 10.1186/s13104-024-06700-3] [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: 04/12/2023] [Accepted: 01/23/2024] [Indexed: 02/09/2024] Open
Abstract
OBJECTIVE Neurotransmitters have been extensively studied as neural communication molecules. Genetic associations discovered, and indirect intervention studies in Humans and mammals have led to a general proposition that neurotransmitters have a role in structuring of neuronal network during development. olf413 is a Drosophila gene annotated as coding for dopamine beta-monooxygenase enzyme with a predicted function in octopaminergic pathway. The biological function of this gene is very little worked out. In this study we investigate the requirement of olf413 gene function for octopamine biogenesis and developmental patterning of embryonic nervous system. RESULT In our study we have used the newly characterized neuronal specific allele olf413SG1.1, and the gene disruption strain olf413MI02014 to dissect out the function of olf413. olf413 has an enhancer activity as depicted by reporter GFP expression, in the embryonic ventral nerve cord, peripheral nervous system and the somatic muscle bundles. Homozygous loss of function mutants show reduced levels of octopamine, and this finding supports the proposed function of the gene in octopamine biogenesis. Further, loss of function of olf413 causes embryonic lethality. FasII staining of these embryos reveal a range of phenotypes in the central and peripheral motor nerves, featuring axonal growth, pathfinding, branching and misrouting defects. Our findings are important as they implicate a key functional requirement of this gene in precise axonal patterning events, a novel developmental role imparted for an octopamine biosynthesis pathway gene in structuring of embryonic nervous system.
Collapse
Affiliation(s)
- Ravindrakumar Ramya
- Developmental Genetics Laboratory, Department of Studies in Zoology, University of Mysore, Mysuru, 570006, India
| | | | | |
Collapse
|
16
|
Fenske SJ, Liu J, Chen H, Diniz MA, Stephens RL, Cornea E, Gilmore JH, Gao W. Sex differences in brain-behavior relationships in the first two years of life. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.31.578147. [PMID: 38352542 PMCID: PMC10862872 DOI: 10.1101/2024.01.31.578147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/24/2024]
Abstract
Background Evidence for sex differences in cognition in childhood is established, but less is known about the underlying neural mechanisms for these differences. Recent findings suggest the existence of brain-behavior relationship heterogeneities during infancy; however, it remains unclear whether sex underlies these heterogeneities during this critical period when sex-related behavioral differences arise. Methods A sample of 316 infants was included with resting-state functional magnetic resonance imaging scans at neonate (3 weeks), 1, and 2 years of age. We used multiple linear regression to test interactions between sex and resting-state functional connectivity on behavioral scores of working memory, inhibitory self-control, intelligence, and anxiety collected at 4 years of age. Results We found six age-specific, intra-hemispheric connections showing significant and robust sex differences in functional connectivity-behavior relationships. All connections are either with the prefrontal cortex or the temporal pole, which has direct anatomical pathways to the prefrontal cortex. Sex differences in functional connectivity only emerge when associated with behavior, and not in functional connectivity alone. Furthermore, at neonate and 2 years of age, these age-specific connections displayed greater connectivity in males and lower connectivity in females in association with better behavioral scores. Conclusions Taken together, we critically capture robust and conserved brain mechanisms that are distinct to sex and are defined by their relationship to behavioral outcomes. Our results establish brain-behavior mechanisms as an important feature in the search for sex differences during development.
Collapse
Affiliation(s)
- Sonja J Fenske
- Biomedical Imaging Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048
- Department of Biomedical Sciences and Imaging, Cedars-Sinai Medical Center, Los Angeles, CA 90048
| | - Janelle Liu
- Biomedical Imaging Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048
- Department of Biomedical Sciences and Imaging, Cedars-Sinai Medical Center, Los Angeles, CA 90048
| | - Haitao Chen
- Biomedical Imaging Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048
- Department of Biomedical Sciences and Imaging, Cedars-Sinai Medical Center, Los Angeles, CA 90048
- David Geffen School of Medicine, University of California, Los Angeles, CA 90025
| | - Marcio A Diniz
- The Biostatistics and Bioinformatics Research Center, Cedars-Sinai Medical Center, Los Angeles, CA 90048
| | - Rebecca L Stephens
- Department of Psychiatry, University of North Carolina Chapel Hill, Chapel Hill, 27599
| | - Emil Cornea
- Department of Psychiatry, University of North Carolina Chapel Hill, Chapel Hill, 27599
| | - John H Gilmore
- Department of Psychiatry, University of North Carolina Chapel Hill, Chapel Hill, 27599
| | - Wei Gao
- Biomedical Imaging Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048
- Department of Biomedical Sciences and Imaging, Cedars-Sinai Medical Center, Los Angeles, CA 90048
- David Geffen School of Medicine, University of California, Los Angeles, CA 90025
| |
Collapse
|
17
|
Mayer FP, Stewart A, Blakely RD. Leaky lessons learned: Efflux prone dopamine transporter variant reveals sex and circuit specific contributions of D2 receptor signalling to neuropsychiatric disease. Basic Clin Pharmacol Toxicol 2024; 134:206-218. [PMID: 37987120 DOI: 10.1111/bcpt.13964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 11/14/2023] [Accepted: 11/17/2023] [Indexed: 11/22/2023]
Abstract
Aberrant dopamine (DA) signalling has been implicated in various neuropsychiatric disorders, including attention-deficit/hyperactivity disorder (ADHD), autism spectrum disorder (ASD), schizophrenia, bipolar disorder (BPD) and addiction. The availability of extracellular DA is sculpted by the exocytotic release of vesicular DA and subsequent transporter-mediated clearance, rendering the presynaptic DA transporter (DAT) a crucial regulator of DA neurotransmission. D2-type DA autoreceptors (D2ARs) regulate multiple aspects of DA homeostasis, including (i) DA synthesis, (ii) vesicular release, (iii) DA neuron firing and (iv) the surface expression of DAT and DAT-mediated DA clearance. The DAT Val559 variant, identified in boys with ADHD or ASD, as well as in a girl with BPD, supports anomalous DA efflux (ADE), which we have shown drives tonic activation of D2ARs. Through ex vivo and in vivo studies of the DAT Val559 variant using transgenic knock-in mice, we have uncovered a circuit and sex-specific capacity of D2ARs to regulate DAT, which consequently disrupts DA signalling and behaviour differently in males and females. Our studies reveal the ability of the construct-valid DAT Val559 model to elucidate endogenous mechanisms that support DA signalling, findings that may be of translational and/or therapeutic importance.
Collapse
Affiliation(s)
- Felix P Mayer
- Department of Biomedical Science, Florida Atlantic University, Jupiter, Florida, USA
- Stiles-Nicholson Brain Institute, Florida Atlantic University, Jupiter, Florida, USA
| | - Adele Stewart
- Department of Biomedical Science, Florida Atlantic University, Jupiter, Florida, USA
- Stiles-Nicholson Brain Institute, Florida Atlantic University, Jupiter, Florida, USA
| | - Randy D Blakely
- Department of Biomedical Science, Florida Atlantic University, Jupiter, Florida, USA
- Stiles-Nicholson Brain Institute, Florida Atlantic University, Jupiter, Florida, USA
| |
Collapse
|
18
|
Nikolic B, Trnski-Levak S, Kosic K, Drlje M, Banovac I, Hranilovic D, Jovanov-Milosevic N. Lasting mesothalamic dopamine imbalance and altered exploratory behavior in rats after a mild neonatal hypoxic event. Front Integr Neurosci 2024; 17:1304338. [PMID: 38304737 PMCID: PMC10832065 DOI: 10.3389/fnint.2023.1304338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Accepted: 12/22/2023] [Indexed: 02/03/2024] Open
Abstract
Introduction Adversities during the perinatal period can decrease oxygen supply to the fetal brain, leading to various hypoxic brain injuries, which can compromise the regularity of brain development in different aspects. To examine the catecholaminergic contribution to the link between an early-life hypoxic insult and adolescent behavioral aberrations, we used a previously established rat model of perinatal hypoxia but altered the hypobaric to normobaric conditions. Methods Exploratory and social behavior and learning abilities were tested in 70 rats of both sexes at adolescent age. Inherent vertical locomotion, sensory-motor functions and spatial learning abilities were explored in a subset of animals to clarify the background of altered exploratory behavior. Finally, the concentrations of dopamine (DA) and noradrenaline in midbrain and pons, and the relative expression of genes for DA receptors D1 and D2, and their down-stream targets (DA- and cAMP-regulated phosphoprotein, Mr 32 kDa, the regulatory subunit of protein kinase A, and inhibitor-5 of protein phosphatase 1) in the hippocampus and thalamus were investigated in 31 rats. Results A lesser extent of alterations in exploratory and cognitive aspects of behavior in the present study suggests that normobaric conditions mitigate the hypoxic injury compared to the one obtained under hypobaric conditions. Increased exploratory rearing was the most prominent consequence, with impaired spatial learning in the background. In affected rats, increased midbrain/pons DA content, as well as mRNA levels for DA receptors and their down-stream elements in the thalamus, but not the hippocampus, were found. Conclusion We can conclude that a mild hypoxic event induced long-lasting disbalances in mesothalamic DA signaling, contributing to the observed behavioral alterations. The thalamus was thereby indicated as another structure, besides the well-established striatum, involved in mediating hypoxic effects on behavior through DA signaling.
Collapse
Affiliation(s)
- Barbara Nikolic
- Department of Biology, University of Zagreb Faculty of Science, Zagreb, Croatia
| | - Sara Trnski-Levak
- Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Kristina Kosic
- Department of Biology, University of Zagreb Faculty of Science, Zagreb, Croatia
| | - Matea Drlje
- Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Ivan Banovac
- Department of Biology, University of Zagreb School of Medicine, Zagreb, Croatia
- Department for Anatomy and Clinical Anatomy, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Dubravka Hranilovic
- Department of Biology, University of Zagreb Faculty of Science, Zagreb, Croatia
| | - Natasa Jovanov-Milosevic
- Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia
- Department of Biology, University of Zagreb School of Medicine, Zagreb, Croatia
| |
Collapse
|
19
|
Bénac N, Ezequiel Saraceno G, Butler C, Kuga N, Nishimura Y, Yokoi T, Su P, Sasaki T, Petit-Pedrol M, Galland R, Studer V, Liu F, Ikegaya Y, Sibarita JB, Groc L. Non-canonical interplay between glutamatergic NMDA and dopamine receptors shapes synaptogenesis. Nat Commun 2024; 15:27. [PMID: 38167277 PMCID: PMC10762086 DOI: 10.1038/s41467-023-44301-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Accepted: 12/07/2023] [Indexed: 01/05/2024] Open
Abstract
Direct interactions between receptors at the neuronal surface have long been proposed to tune signaling cascades and neuronal communication in health and disease. Yet, the lack of direct investigation methods to measure, in live neurons, the interaction between different membrane receptors at the single molecule level has raised unanswered questions on the biophysical properties and biological roles of such receptor interactome. Using a multidimensional spectral single molecule-localization microscopy (MS-SMLM) approach, we monitored the interaction between two membrane receptors, i.e. glutamatergic NMDA (NMDAR) and G protein-coupled dopamine D1 (D1R) receptors. The transient interaction was randomly observed along the dendritic tree of hippocampal neurons. It was higher early in development, promoting the formation of NMDAR-D1R complexes in an mGluR5- and CK1-dependent manner, favoring NMDAR clusters and synaptogenesis in a dopamine receptor signaling-independent manner. Preventing the interaction in the neonate, and not adult, brain alters in vivo spontaneous neuronal network activity pattern in male mice. Thus, a weak and transient interaction between NMDAR and D1R plays a structural and functional role in the developing brain.
Collapse
Affiliation(s)
- Nathan Bénac
- Univ. Bordeaux, CNRS, IINS, UMR 5297, F-33000, Bordeaux, France
| | | | - Corey Butler
- Univ. Bordeaux, CNRS, IINS, UMR 5297, F-33000, Bordeaux, France
| | - Nahoko Kuga
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo Bunkyo-ku, Tokyo, 113-0033, Japan
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3 Aramaki-aoba, Sendai, Miyagi, 980-8578, Japan
| | - Yuya Nishimura
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Taiki Yokoi
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3 Aramaki-aoba, Sendai, Miyagi, 980-8578, Japan
| | - Ping Su
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, University of Toronto, Toronto, Canada
| | - Takuya Sasaki
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo Bunkyo-ku, Tokyo, 113-0033, Japan
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3 Aramaki-aoba, Sendai, Miyagi, 980-8578, Japan
| | | | - Rémi Galland
- Univ. Bordeaux, CNRS, IINS, UMR 5297, F-33000, Bordeaux, France
| | - Vincent Studer
- Univ. Bordeaux, CNRS, IINS, UMR 5297, F-33000, Bordeaux, France
| | - Fang Liu
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, University of Toronto, Toronto, Canada
| | - Yuji Ikegaya
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo Bunkyo-ku, Tokyo, 113-0033, Japan
- Center for Information and Neural Networks, Suita City, Osaka, 565-0871, Japan
- Institute for AI and Beyond, The University of Tokyo, Tokyo, 113-0033, Japan
| | | | - Laurent Groc
- Univ. Bordeaux, CNRS, IINS, UMR 5297, F-33000, Bordeaux, France.
| |
Collapse
|
20
|
Vaidya B, Padhy DS, Joshi HC, Sharma SS, Singh JN. Ion Channels and Metal Ions in Parkinson's Disease: Historical Perspective to the Current Scenario. Methods Mol Biol 2024; 2761:529-557. [PMID: 38427260 DOI: 10.1007/978-1-0716-3662-6_36] [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: 03/02/2024]
Abstract
Parkinson's disease (PD) is a neurodegenerative condition linked to the deterioration of motor and cognitive performance. It produces degeneration of the dopaminergic neurons along the nigrostriatal pathway in the central nervous system (CNS), which leads to symptoms such as bradykinesias, tremors, rigidity, and postural instability. There are several medications currently approved for the therapy of PD, but a permanent cure for it remains elusive. With the aging population set to increase, a number of PD cases are expected to shoot up in the coming times. Hence, there is a need to look for new molecular targets that could be investigated both preclinically and clinically for PD treatment. Among these, several ion channels and metal ions are being studied for their effects on PD pathology and the functioning of dopaminergic neurons. Ion channels such as N-methyl-D-aspartate (NMDA), γ-aminobutyric acid A (GABAA), voltage-gated calcium channels, potassium channels, HCN channels, Hv1 proton channels, and voltage-gated sodium channels and metal ions such as mercury, zinc, copper, iron, manganese, calcium, and lead showed prominent involvement in PD. Pharmacological agents have been used to target these ion channels and metal ions to prevent or treat PD. Hence, in the present review, we summarize the pathophysiological events linked to PD with an emphasis on the role of ions and ion channels in PD pathology, and pharmacological agents targeting these ion channels have also been listed.
Collapse
Affiliation(s)
- Bhupesh Vaidya
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Punjab, India
| | - Dibya S Padhy
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Punjab, India
| | - Hem C Joshi
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Punjab, India
| | - Shyam S Sharma
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Punjab, India.
| | - Jitendra Narain Singh
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Punjab, India.
| |
Collapse
|
21
|
Dubois MA, Pelletier CA, Mérette C, Jomphe V, Turgeon R, Bélanger RE, Grondin S, Hébert M. Evaluation of electroretinography (ERG) parameters as a biomarker for ADHD. Prog Neuropsychopharmacol Biol Psychiatry 2023; 127:110807. [PMID: 37290571 DOI: 10.1016/j.pnpbp.2023.110807] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 05/19/2023] [Accepted: 06/01/2023] [Indexed: 06/10/2023]
Abstract
BACKGROUND The retina is recognized as an accessible part of the brain due to their common embryonic origin. The electroretinogram (ERG) has proven to be a valuable tool for detecting schizophrenia and bipolarity. We therefore investigated its ability to detect ADHD. METHODS The cone and rod luminance response functions of the ERG were recorded in 26 ADHD subjects (17 women and 9 men) and 25 controls (16 women and 9 men). RESULTS No significant differences were found between the mixed groups, but sexual dysmorphia was observed in the significant results. In males, a significant prolonged cone a-wave latency was observed in the ADHD group. In females, we observed a significant decrease in the cone a- and b-wave amplitudes and a trend for a prolonged cone b-wave latency as well as a higher scotopic mixed rod-cone a-wave in the ADHD group. CONCLUSION The data obtained in this study show the potential of the ERG to detect ADHD, warranting further large-scale studies.
Collapse
Affiliation(s)
- Marc-André Dubois
- CERVO Brain Research Centre, Centre Intégré Universitaire de Santé et des Services Sociaux de la Capitale Nationale, Quebec, QC, Canada; School of Psychology, Faculty of Social Sciences, Université Laval, Quebec, QC, Canada
| | - Charles-Antoine Pelletier
- CERVO Brain Research Centre, Centre Intégré Universitaire de Santé et des Services Sociaux de la Capitale Nationale, Quebec, QC, Canada
| | - Chantal Mérette
- CERVO Brain Research Centre, Centre Intégré Universitaire de Santé et des Services Sociaux de la Capitale Nationale, Quebec, QC, Canada; Department of Psychiatry and Neurosciences, Faculty of Medicine, Université Laval, Quebec, QC, Canada
| | - Valérie Jomphe
- CERVO Brain Research Centre, Centre Intégré Universitaire de Santé et des Services Sociaux de la Capitale Nationale, Quebec, QC, Canada
| | - Rose Turgeon
- CERVO Brain Research Centre, Centre Intégré Universitaire de Santé et des Services Sociaux de la Capitale Nationale, Quebec, QC, Canada
| | - Richard E Bélanger
- CHU de Québec Research Centre, Quebec, QC, Canada; Department of Pediatrics, Faculty of Medicine, Université Laval, Quebec, QC, Canada
| | - Simon Grondin
- School of Psychology, Faculty of Social Sciences, Université Laval, Quebec, QC, Canada
| | - Marc Hébert
- CERVO Brain Research Centre, Centre Intégré Universitaire de Santé et des Services Sociaux de la Capitale Nationale, Quebec, QC, Canada; Department of Ophthalmology and Otorhinolaryngology - Head and Neck Surgery, Faculty of Medicine, Université Laval, Quebec, QC, Canada.
| |
Collapse
|
22
|
Talavera-Barber MM, Morehead E, Ziegler K, Hockett C, Elliott AJ. Prenatal cannabinoid exposure and early language development. Front Pediatr 2023; 11:1290707. [PMID: 38078314 PMCID: PMC10702953 DOI: 10.3389/fped.2023.1290707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 10/31/2023] [Indexed: 02/12/2024] Open
Abstract
Introduction The effect of prenatal cannabis exposure (PCE) on childhood neurodevelopment remains poorly understood. There is a paucity of studies describing the neurodevelopment impact of PCE in infancy. The Mullen Scale of Early Learning (MSEL) is a cognitive screening tool that can be used from birth to 68 months and includes language and motor domains. Here we aim to explore the association between PCE during pregnancy and neurodevelopmental outcomes at 12 months of age. Methods Participants were pregnant persons/infant pairs enrolled in The Safe Passage Study, a large prospective cohort study. Inclusion criteria included data available on PCE with associated MSEL scores at 12 months of age. Exposed participants were defined as early exposure (1st trimester only) or late exposure (2nd or 3rd trimester) and were randomly matched with unexposed participants. Multiple linear regression models were performed to test associations between prenatal cannabis exposure and the five Mullen subscales: gross motor, fine motor, expressive language, receptive language, and visual reception. Results Sixty-nine exposed and 138 randomly matched unexposed infants were included in the analyses. Mothers of children with PCE were younger with the mean age 23.7 years for early exposure (n = 51) and 22.8 years for late exposure (n = 18). Maternal characteristics with prenatal cannabis use include a high-school education, American Indian or Alaska Native descent, lower socioeconomic status and co-use of tobacco. There were no gestational age or sex difference among the groups. Expressive (95% CI: 2.54-12.76; p = 0.0036,) and receptive language scores (95% CI: 0.39-8.72; p = 0.0322) were significantly increased between late-exposed infants compared to unexposed infants following adjustment for covariates. Gross motor scores (95% CI: 1.75-13; p = 0.0105) were also significantly increased for early-exposed infants with no difference in visual reception scores. Conclusion Preclinical studies have shown abnormal brain connectivity in offspring exposed to cannabis affecting emotional regulation, hyperactivity, and language development. Results from this study link PCE to altered early language development within the first year of life. Exposed infants demonstrated increased expressive and receptive language scores at 12 months of age, which can translate to better performance in school. However, further research is needed to determine the implications of these results later in childhood.
Collapse
Affiliation(s)
- Maria M. Talavera-Barber
- Avera Research Institute, Sioux Falls, SD, United States
- Department of Pediatrics, University of South Dakota Sanford School of Medicine, Sioux Falls, SD, United States
| | - Evlyn Morehead
- Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL, United States
| | - Katherine Ziegler
- Avera Research Institute, Sioux Falls, SD, United States
- Department of Pediatrics, University of South Dakota Sanford School of Medicine, Sioux Falls, SD, United States
| | - Christine Hockett
- Avera Research Institute, Sioux Falls, SD, United States
- Department of Pediatrics, University of South Dakota Sanford School of Medicine, Sioux Falls, SD, United States
| | - Amy J. Elliott
- Avera Research Institute, Sioux Falls, SD, United States
- Department of Pediatrics, University of South Dakota Sanford School of Medicine, Sioux Falls, SD, United States
| |
Collapse
|
23
|
Rudibaugh TT, Stuppy SR, Keung AJ. Reactive Oxygen Species Mediate Transcriptional Responses to Dopamine and Cocaine in Human Cerebral Organoids. Int J Mol Sci 2023; 24:16474. [PMID: 38003664 PMCID: PMC10671319 DOI: 10.3390/ijms242216474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 11/13/2023] [Accepted: 11/14/2023] [Indexed: 11/26/2023] Open
Abstract
Dopamine signaling in the adult ventral forebrain regulates behavior, stress response, and memory formation and in neurodevelopment regulates neural differentiation and cell migration. Excessive dopamine levels, including those due to cocaine use in utero and in adults, could lead to long-term adverse consequences. The mechanisms underlying both homeostatic and pathological changes remain unclear, in part due to the diverse cellular responses elicited by dopamine and the reliance on animal models that exhibit species-specific differences in dopamine signaling. In this study, we use the human-derived ventral forebrain organoid model of Xiang-Tanaka and characterize their response to cocaine or dopamine. We explore dosing regimens of dopamine or cocaine to simulate acute or chronic exposure. We then use calcium imaging, cAMP imaging, and bulk RNA-sequencing to measure responses to cocaine or dopamine exposure. We observe an upregulation of inflammatory pathways in addition to indicators of oxidative stress following exposure. Using inhibitors of reactive oxygen species (ROS), we then show ROS to be necessary for multiple transcriptional responses of cocaine exposure. These results highlight novel response pathways and validate the potential of cerebral organoids as in vitro human models for studying complex biological processes in the brain.
Collapse
Affiliation(s)
| | | | - Albert J. Keung
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, NC 27606, USA; (T.T.R.); (S.R.S.)
| |
Collapse
|
24
|
Wu L, Zeeshan M, Dang Y, Zhang YT, Liang LX, Huang JW, Zhou JX, Guo LH, Fan YY, Sun MK, Yu T, Wen Y, Lin LZ, Liu RQ, Dong GH, Chu C. Maternal transfer of F-53B inhibited neurobehavior in zebrafish offspring larvae and potential mechanisms: Dopaminergic dysfunction, eye development defects and disrupted calcium homeostasis. THE SCIENCE OF THE TOTAL ENVIRONMENT 2023; 894:164838. [PMID: 37353013 DOI: 10.1016/j.scitotenv.2023.164838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 05/25/2023] [Accepted: 06/10/2023] [Indexed: 06/25/2023]
Abstract
Maternal exposure to environment toxicants is an important risk factor for neurobehavioral health in their offspring. In our study, we investigated the impact of maternal exposure to chlorinated polyfluoroalkyl ether sulfonic acids (Cl-PFESAs, commercial name: F-53B) on behavioral changes and the potential mechanism in the offspring larvae of zebrafish. Adult zebrafish exposed to Cl-PFESAs (0, 0.2, 2, 20 and 200 μg/L) for 21 days were subsequently mated their embryos were cultured for 5 days. Higher concentrations of Cl-PFESAs in zebrafish embryos were observed, along with, reduced swimming speed and distance travelled in the offspring larvae. Molecular docking analysis revealed that Cl-PFESAs can form hydrogen bonds with brain-derived neurotropic factor (BDNF), protein kinase C, alpha, (PKCα), Ca2+-ATPase and Na, K - ATPase. Molecular and biochemical studies evidenced Cl-PFESAs induce dopaminergic dysfunction, eye developmental defects and disrupted Ca2+ homeostasis. Together, our results showed that maternal exposure to Cl-PFESAs lead to behavioral alteration in offspring mediated by disruption in Ca2+ homeostasis, dopaminergic dysfunction and eye developmental defects.
Collapse
Affiliation(s)
- Luyin Wu
- Guangdong Provincial Engineering Technology Research Center of Environmental Pollution and Health Risk Assessment, Department of Occupational and Environmental Health, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Mohammed Zeeshan
- Guangdong Provincial Engineering Technology Research Center of Environmental Pollution and Health Risk Assessment, Department of Occupational and Environmental Health, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Yao Dang
- State Environmental Protection Key Laboratory of Environmental Pollution Health Risk Assessment, South China Institute of Environmental Sciences, Ministry of Ecology and Environment, Guangzhou 510655, China
| | - Yun-Ting Zhang
- Guangdong Provincial Engineering Technology Research Center of Environmental Pollution and Health Risk Assessment, Department of Occupational and Environmental Health, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Li-Xia Liang
- Guangdong Provincial Engineering Technology Research Center of Environmental Pollution and Health Risk Assessment, Department of Occupational and Environmental Health, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Jing-Wen Huang
- Guangdong Provincial Engineering Technology Research Center of Environmental Pollution and Health Risk Assessment, Department of Occupational and Environmental Health, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Jia-Xin Zhou
- Guangdong Provincial Engineering Technology Research Center of Environmental Pollution and Health Risk Assessment, Department of Occupational and Environmental Health, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Li-Hao Guo
- Guangdong Provincial Engineering Technology Research Center of Environmental Pollution and Health Risk Assessment, Department of Occupational and Environmental Health, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Yuan-Yuan Fan
- Guangdong Provincial Engineering Technology Research Center of Environmental Pollution and Health Risk Assessment, Department of Occupational and Environmental Health, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Ming-Kun Sun
- Guangdong Provincial Engineering Technology Research Center of Environmental Pollution and Health Risk Assessment, Department of Occupational and Environmental Health, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Tao Yu
- Guangdong Provincial Engineering Technology Research Center of Environmental Pollution and Health Risk Assessment, Department of Occupational and Environmental Health, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Yue Wen
- Guangdong Provincial Engineering Technology Research Center of Environmental Pollution and Health Risk Assessment, Department of Occupational and Environmental Health, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Li-Zi Lin
- Guangdong Provincial Engineering Technology Research Center of Environmental Pollution and Health Risk Assessment, Department of Occupational and Environmental Health, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Ru-Qing Liu
- Guangdong Provincial Engineering Technology Research Center of Environmental Pollution and Health Risk Assessment, Department of Occupational and Environmental Health, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Guang-Hui Dong
- Guangdong Provincial Engineering Technology Research Center of Environmental Pollution and Health Risk Assessment, Department of Occupational and Environmental Health, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Chu Chu
- Guangdong Provincial Engineering Technology Research Center of Environmental Pollution and Health Risk Assessment, Department of Occupational and Environmental Health, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China.
| |
Collapse
|
25
|
Toth BA, Chang KS, Fechtali S, Burgess CR. Dopamine release in the nucleus accumbens promotes REM sleep and cataplexy. iScience 2023; 26:107613. [PMID: 37664637 PMCID: PMC10470413 DOI: 10.1016/j.isci.2023.107613] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 07/21/2023] [Accepted: 08/09/2023] [Indexed: 09/05/2023] Open
Abstract
Patients with the sleep disorder narcolepsy suffer from excessive daytime sleepiness, disrupted nighttime sleep, and cataplexy-the abrupt loss of postural muscle tone during wakefulness, often triggered by strong emotion. The dopamine (DA) system is implicated in both sleep-wake states and cataplexy, but little is known about the function of DA release in the striatum and sleep disorders. Recording DA release in the ventral striatum revealed orexin-independent changes across sleep-wake states as well as striking increases in DA release in the ventral, but not dorsal, striatum prior to cataplexy onset. Tonic low-frequency stimulation of ventral tegmental efferents in the ventral striatum suppressed both cataplexy and rapid eye movement (REM) sleep, while phasic high-frequency stimulation increased cataplexy propensity and decreased the latency to REM sleep. Together, our findings demonstrate a functional role of DA release in the striatum in regulating cataplexy and REM sleep.
Collapse
Affiliation(s)
- Brandon A. Toth
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI, USA
- Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI, USA
| | - Katie S. Chang
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI, USA
| | - Sarah Fechtali
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - Christian R. Burgess
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI, USA
- Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI, USA
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
26
|
Saha S, Chatterjee M, Dutta N, Sinha S, Mukhopadhyay K. Analysis of neurotransmitters validates the importance of the dopaminergic system in autism spectrum disorder. World J Pediatr 2023; 19:770-781. [PMID: 36847977 DOI: 10.1007/s12519-023-00702-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Accepted: 02/03/2023] [Indexed: 03/01/2023]
Abstract
BACKGROUND The reasons behind the cardinal symptoms of communication deficits and repetitive, stereotyped behaviors that characterize autism spectrum disorder (ASD) remain unknown. The dopamine (DA) system, which regulates motor activity, goal-directed behaviors, and reward function, is believed to play a crucial role in ASD, although the exact mechanism is still unclear. Investigations have shown an association of the dopamine receptor D4 (DRD4) with various neurobehavioral disorders. METHODS We analyzed the association between ASD and four DRD4 genetic polymorphisms, 5' flanking 120-bp duplication (rs4646984), rs1800955 in the promoter, exon 1 12 bp duplication (rs4646983), and exon 3 48 bp repeats. We also examined plasma DA and its metabolite levels, DRD4 mRNA expression, and correlations of the studied polymorphisms with these parameters by case-control comparative analyses. The expression of DA transporter (DAT), which is important in regulating the circulating DA level, was also evaluated. RESULTS A significantly higher occurrence of rs1800955 "T/TT" was observed in the probands. ASD traits were affected by rs1800955 "T" and the higher repeat alleles of the exon 3 48 bp repeats, rs4646983 and rs4646984. ASD probands exhibited lower DA and norepinephrine levels together with higher homovanillic acid levels than the control subjects. DAT and DRD4 mRNA expression were down-regulated in the probands, especially in the presence of DAT rs3836790 "6R" and rs27072 "CC" and DRD4 rs4646984 higher repeat allele and rs1800955 "T". CONCLUSION This pioneering investigation revealed a positive correlation between genetic variants, hypodopaminergic state, and impairment in socio-emotional and communication reciprocity in Indian subjects with ASD, warranting further in-depth analysis.
Collapse
Affiliation(s)
- Sharmistha Saha
- Manovikas Biomedical Research and Diagnostic Centre, Manovikas Kendra, 482 Madudah, Plot I-24, Sector-J, E.M. Bypass, Kolkata, West Bengal, 700107, India
| | - Mahasweta Chatterjee
- Manovikas Biomedical Research and Diagnostic Centre, Manovikas Kendra, 482 Madudah, Plot I-24, Sector-J, E.M. Bypass, Kolkata, West Bengal, 700107, India
| | - Nilanjana Dutta
- Manovikas Biomedical Research and Diagnostic Centre, Manovikas Kendra, 482 Madudah, Plot I-24, Sector-J, E.M. Bypass, Kolkata, West Bengal, 700107, India
| | - Swagata Sinha
- Manovikas Biomedical Research and Diagnostic Centre, Manovikas Kendra, 482 Madudah, Plot I-24, Sector-J, E.M. Bypass, Kolkata, West Bengal, 700107, India
| | - Kanchan Mukhopadhyay
- Manovikas Biomedical Research and Diagnostic Centre, Manovikas Kendra, 482 Madudah, Plot I-24, Sector-J, E.M. Bypass, Kolkata, West Bengal, 700107, India.
| |
Collapse
|
27
|
Taibl KR, Dunlop AL, Barr DB, Li YY, Eick SM, Kannan K, Ryan PB, Schroder M, Rushing B, Fennell T, Chang CJ, Tan Y, Marsit CJ, Jones DP, Liang D. Newborn metabolomic signatures of maternal per- and polyfluoroalkyl substance exposure and reduced length of gestation. Nat Commun 2023; 14:3120. [PMID: 37253729 PMCID: PMC10229585 DOI: 10.1038/s41467-023-38710-3] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Accepted: 05/10/2023] [Indexed: 06/01/2023] Open
Abstract
Marginalized populations experience disproportionate rates of preterm birth and early term birth. Exposure to per- and polyfluoroalkyl substances (PFAS) has been reported to reduce length of gestation, but the underlying mechanisms are unknown. In the present study, we characterized the molecular signatures of prenatal PFAS exposure and gestational age at birth outcomes in the newborn dried blood spot metabolome among 267 African American dyads in Atlanta, Georgia between 2016 and 2020. Pregnant people with higher serum perfluorooctanoic acid and perfluorohexane sulfonic acid concentrations had increased odds of an early birth. After false discovery rate correction, the effect of prenatal PFAS exposure on reduced length of gestation was associated with 8 metabolomic pathways and 52 metabolites in newborn dried blood spots, which suggested perturbed tissue neogenesis, neuroendocrine function, and redox homeostasis. These mechanisms explain how prenatal PFAS exposure gives rise to the leading cause of infant death in the United States.
Collapse
Grants
- R01 NR014800 NINR NIH HHS
- U2C ES026542 NIEHS NIH HHS
- P50 ES026071 NIEHS NIH HHS
- R01 MD009064 NIMHD NIH HHS
- UH3 OD023318 NIH HHS
- R01 MD009746 NIMHD NIH HHS
- R21 ES032117 NIEHS NIH HHS
- U2C ES026560 NIEHS NIH HHS
- P30 ES019776 NIEHS NIH HHS
- R24 ES029490 NIEHS NIH HHS
- U24 ES029490 NIEHS NIH HHS
- UG3 OD023318 NIH HHS
- U.S. Department of Health & Human Services | NIH | National Institute of Environmental Health Sciences (NIEHS)
- U.S. Department of Health & Human Services | NIH | National Institute of Nursing Research (NINR)
- U.S. Department of Health & Human Services | NIH | National Institute on Minority Health and Health Disparities (NIMHD)
- Research reported in this publication was supported by the Environmental Influences on Child Health Outcomes (ECHO) program, Office of the Director, National Institutes of Health, under Award Numbers 5U2COD023375-05/A03-3824, the National Institute of Health (NIH) research grants [R21ES032117, R01NR014800, R01MD009064, R24ES029490, R01MD009746], NIH Center Grants [P50ES02607, P30ES019776, UH3OD023318, U2CES026560, U2CES026542], and Environmental Protection Agency (USEPA) center grant [83615301].
Collapse
Affiliation(s)
- Kaitlin R Taibl
- Gangarosa Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Anne L Dunlop
- Department of Gynecology and Obstetrics, School of Medicine, Emory University, Atlanta, GA, USA.
| | - Dana Boyd Barr
- Gangarosa Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Yuan-Yuan Li
- Metabolomics and Exposome Laboratory, Nutrition Research Institute, University of North Carolina, Chapel Hill, NC, USA
| | - Stephanie M Eick
- Gangarosa Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Kurunthachalam Kannan
- Department of Pediatrics, New York University School of Medicine, New York, NY, USA
- Department of Environmental Medicine, New York University School of Medicine, New York, NY, USA
| | - P Barry Ryan
- Gangarosa Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Madison Schroder
- Metabolomics and Exposome Laboratory, Nutrition Research Institute, University of North Carolina, Chapel Hill, NC, USA
| | - Blake Rushing
- Metabolomics and Exposome Laboratory, Nutrition Research Institute, University of North Carolina, Chapel Hill, NC, USA
| | - Timothy Fennell
- Analytical Chemistry and Pharmaceuticals, RTI International, Research Triangle Park, Durham, NC, USA
| | - Che-Jung Chang
- Gangarosa Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Youran Tan
- Gangarosa Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Carmen J Marsit
- Gangarosa Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Dean P Jones
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, School of Medicine, Emory University, Atlanta, GA, USA
| | - Donghai Liang
- Gangarosa Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA, USA.
| |
Collapse
|
28
|
Tristán‐Noguero A, Fernández‐Carasa I, Calatayud C, Bermejo‐Casadesús C, Pons‐Espinal M, Colini Baldeschi A, Campa L, Artigas F, Bortolozzi A, Domingo‐Jiménez R, Ibáñez S, Pineda M, Artuch R, Raya Á, García‐Cazorla À, Consiglio A. iPSC-based modeling of THD recapitulates disease phenotypes and reveals neuronal malformation. EMBO Mol Med 2023; 15:e15847. [PMID: 36740977 PMCID: PMC9994475 DOI: 10.15252/emmm.202215847] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 12/28/2022] [Accepted: 01/10/2023] [Indexed: 02/07/2023] Open
Abstract
Tyrosine hydroxylase deficiency (THD) is a rare genetic disorder leading to dopaminergic depletion and early-onset Parkinsonism. Affected children present with either a severe form that does not respond to L-Dopa treatment (THD-B) or a milder L-Dopa responsive form (THD-A). We generated induced pluripotent stem cells (iPSCs) from THD patients that were differentiated into dopaminergic neurons (DAn) and compared with control-DAn from healthy individuals and gene-corrected isogenic controls. Consistent with patients, THD iPSC-DAn displayed lower levels of DA metabolites and reduced TH expression, when compared to controls. Moreover, THD iPSC-DAn showed abnormal morphology, including reduced total neurite length and neurite arborization defects, which were not evident in DAn differentiated from control-iPSC. Treatment of THD-iPSC-DAn with L-Dopa rescued the neuronal defects and disease phenotype only in THDA-DAn. Interestingly, L-Dopa treatment at the stage of neuronal precursors could prevent the alterations in THDB-iPSC-DAn, thus suggesting the existence of a critical developmental window in THD. Our iPSC-based model recapitulates THD disease phenotypes and response to treatment, representing a promising tool for investigating pathogenic mechanisms, drug screening, and personalized management.
Collapse
Affiliation(s)
- Alba Tristán‐Noguero
- Neurometabolic Unit and Synaptic Metabolism Lab, Neurology DepartmentInstitut Pediàtric de Recerca, Hospital Sant Joan de DéuBarcelonaSpain
| | - Irene Fernández‐Carasa
- Department of Pathology and Experimental TherapeuticsBellvitge University Hospital‐IDIBELL, Hospitalet de LlobregatBarcelonaSpain
- Institute of Biomedicine of the University of Barcelona (IBUB)BarcelonaSpain
| | - Carles Calatayud
- Department of Pathology and Experimental TherapeuticsBellvitge University Hospital‐IDIBELL, Hospitalet de LlobregatBarcelonaSpain
- Institute of Biomedicine of the University of Barcelona (IBUB)BarcelonaSpain
- Regenerative Medicine ProgramBellvitge Biomedical Research Institute (IDIBELL)BarcelonaSpain
- Program for Translation of Regenerative Medicine in Catalonia (P‐[CMRC])Hospital Duran i Reynals, Hospitalet de LlobregatBarcelonaSpain
| | - Cristina Bermejo‐Casadesús
- Neurometabolic Unit and Synaptic Metabolism Lab, Neurology DepartmentInstitut Pediàtric de Recerca, Hospital Sant Joan de DéuBarcelonaSpain
| | - Meritxell Pons‐Espinal
- Department of Pathology and Experimental TherapeuticsBellvitge University Hospital‐IDIBELL, Hospitalet de LlobregatBarcelonaSpain
- Institute of Biomedicine of the University of Barcelona (IBUB)BarcelonaSpain
| | - Arianna Colini Baldeschi
- Department of Pathology and Experimental TherapeuticsBellvitge University Hospital‐IDIBELL, Hospitalet de LlobregatBarcelonaSpain
- Institute of Biomedicine of the University of Barcelona (IBUB)BarcelonaSpain
| | - Leticia Campa
- Institut d'Investigacions Biomèdiques de Barcelona (IIBB), Spanish National Research Council (CSIC)BarcelonaSpain
- Institut d'Investigacions August Pi i Sunyer (IDIBAPS)BarcelonaSpain
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), ISCIIIMadridSpain
| | - Francesc Artigas
- Institut d'Investigacions Biomèdiques de Barcelona (IIBB), Spanish National Research Council (CSIC)BarcelonaSpain
- Institut d'Investigacions August Pi i Sunyer (IDIBAPS)BarcelonaSpain
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), ISCIIIMadridSpain
| | - Analia Bortolozzi
- Institut d'Investigacions Biomèdiques de Barcelona (IIBB), Spanish National Research Council (CSIC)BarcelonaSpain
- Institut d'Investigacions August Pi i Sunyer (IDIBAPS)BarcelonaSpain
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), ISCIIIMadridSpain
| | - Rosario Domingo‐Jiménez
- Department of Pediatric NeurologyHospital Virgen de la ArrixacaMurciaSpain
- Instituto Murciano de Investigación Biosanitaria Virgen de la Arrixaca (IMIB)MurciaSpain
- Centro de Investigación Biomédica En Red Enfermedades Raras (CIBERER)MadridSpain
| | - Salvador Ibáñez
- Department of Pediatric NeurologyHospital Virgen de la ArrixacaMurciaSpain
- Instituto Murciano de Investigación Biosanitaria Virgen de la Arrixaca (IMIB)MurciaSpain
| | - Mercè Pineda
- Fundació Sant Joan de Déu (FSJD), Hospital Sant Joan de Déu (HSJD)BarcelonaSpain
| | - Rafael Artuch
- Centro de Investigación Biomédica En Red Enfermedades Raras (CIBERER)MadridSpain
- Metabolic Unit, Departments of Neurology, Nutrition Biochemistry and GeneticsInstitut Pediàtric de Recerca, Hospital San Joan de DéuBarcelonaSpain
| | - Ángel Raya
- Regenerative Medicine ProgramBellvitge Biomedical Research Institute (IDIBELL)BarcelonaSpain
- Program for Translation of Regenerative Medicine in Catalonia (P‐[CMRC])Hospital Duran i Reynals, Hospitalet de LlobregatBarcelonaSpain
- Centre for Networked Biomedical Research on Bioengineering, Biomaterials and Nanomedicine (CIBER‐BBN)MadridSpain
- Institució Catalana de Recerca i Estudis Avançats (ICREA)BarcelonaSpain
| | - Àngels García‐Cazorla
- Neurometabolic Unit and Synaptic Metabolism Lab, Neurology DepartmentInstitut Pediàtric de Recerca, Hospital Sant Joan de DéuBarcelonaSpain
- Centro de Investigación Biomédica En Red Enfermedades Raras (CIBERER)MadridSpain
| | - Antonella Consiglio
- Department of Pathology and Experimental TherapeuticsBellvitge University Hospital‐IDIBELL, Hospitalet de LlobregatBarcelonaSpain
- Institute of Biomedicine of the University of Barcelona (IBUB)BarcelonaSpain
- Department of Molecular and Translational MedicineUniversity of BresciaBresciaItaly
| |
Collapse
|
29
|
Mintz Hemed N, Melosh NA. An integrated perspective for the diagnosis and therapy of neurodevelopmental disorders - From an engineering point of view. Adv Drug Deliv Rev 2023; 194:114723. [PMID: 36746077 DOI: 10.1016/j.addr.2023.114723] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 12/14/2022] [Accepted: 01/29/2023] [Indexed: 02/05/2023]
Abstract
Neurodevelopmental disorders (NDDs) are complex conditions with largely unknown pathophysiology. While many NDD symptoms are familiar, the cause of these disorders remains unclear and may involve a combination of genetic, biological, psychosocial, and environmental risk factors. Current diagnosis relies heavily on behaviorally defined criteria, which may be biased by the clinical team's professional and cultural expectations, thus a push for new biological-based biomarkers for NDDs diagnosis is underway. Emerging new research technologies offer an unprecedented view into the electrical, chemical, and physiological activity in the brain and with further development in humans may provide clinically relevant diagnoses. These could also be extended to new treatment options, which can start to address the underlying physiological issues. When combined with current speech, language, occupational therapy, and pharmacological treatment these could greatly improve patient outcomes. The current review will discuss the latest technologies that are being used or may be used for NDDs diagnosis and treatment. The aim is to provide an inspiring and forward-looking view for future research in the field.
Collapse
Affiliation(s)
- Nofar Mintz Hemed
- Department of Materials Science and Engineering, Stanford University, Stanford, CA 94305, USA.
| | - Nicholas A Melosh
- Department of Materials Science and Engineering, Stanford University, Stanford, CA 94305, USA
| |
Collapse
|
30
|
de Leeuw VC, van Oostrom CTM, Zwart EP, Heusinkveld HJ, Hessel EVS. Prolonged Differentiation of Neuron-Astrocyte Co-Cultures Results in Emergence of Dopaminergic Neurons. Int J Mol Sci 2023; 24:ijms24043608. [PMID: 36835019 PMCID: PMC9959280 DOI: 10.3390/ijms24043608] [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: 12/21/2022] [Revised: 02/03/2023] [Accepted: 02/07/2023] [Indexed: 02/15/2023] Open
Abstract
Dopamine is present in a subgroup of neurons that are vital for normal brain functioning. Disruption of the dopaminergic system, e.g., by chemical compounds, contributes to the development of Parkinson's disease and potentially some neurodevelopmental disorders. Current test guidelines for chemical safety assessment do not include specific endpoints for dopamine disruption. Therefore, there is a need for the human-relevant assessment of (developmental) neurotoxicity related to dopamine disruption. The aim of this study was to determine the biological domain related to dopaminergic neurons of a human stem cell-based in vitro test, the human neural progenitor test (hNPT). Neural progenitor cells were differentiated in a neuron-astrocyte co-culture for 70 days, and dopamine-related gene and protein expression was investigated. Expression of genes specific for dopaminergic differentiation and functioning, such as LMX1B, NURR1, TH, SLC6A3, and KCNJ6, were increasing by day 14. From day 42, a network of neurons expressing the catecholamine marker TH and the dopaminergic markers VMAT2 and DAT was present. These results confirm stable gene and protein expression of dopaminergic markers in hNPT. Further characterization and chemical testing are needed to investigate if the model might be relevant in a testing strategy to test the neurotoxicity of the dopaminergic system.
Collapse
|
31
|
Frau R, Melis M. Sex-specific susceptibility to psychotic-like states provoked by prenatal THC exposure: Reversal by pregnenolone. J Neuroendocrinol 2023; 35:e13240. [PMID: 36810840 DOI: 10.1111/jne.13240] [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: 08/01/2022] [Revised: 01/23/2023] [Accepted: 02/02/2023] [Indexed: 02/10/2023]
Abstract
Sociocultural attitudes towards cannabis legalization contribute to the common misconception that it is a relatively safe drug and its use during pregnancy poses no risk to the fetus. However, longitudinal studies demonstrate that maternal cannabis exposure results in adverse outcomes in the offspring, with a heightened risk for developing psychopathology. One of the most reported psychiatric outcomes is the proneness to psychotic-like experiences during childhood. How exposure to cannabis during gestation increases psychosis susceptibility in children and adolescents remains elusive. Preclinical research has indicated that in utero exposure to the major psychoactive component of cannabis, delta-9-tetrahydrocannabinol (THC), deranges brain developmental trajectories towards vulnerable psychotic-like endophenotypes later in life. Here, we present how prenatal THC exposure (PCE) deregulates mesolimbic dopamine development predisposing the offspring to schizophrenia-relevant phenotypes, exclusively when exposed to environmental challenges, such as stress or THC. Detrimental effects of PCE are sex-specific because female offspring do not display psychotic-like outcomes upon exposure to these challenges. Moreover, we present how pregnenolone, a neurosteroid that showed beneficial properties on the effects elicited by cannabis intoxication, normalizes mesolimbic dopamine function and rescues psychotic-like phenotypes. We, therefore, suggest this neurosteroid as a safe "disease-modifying" aid to prevent the onset of psychoses in vulnerable individuals. Our findings corroborate clinical evidence and highlight the relevance of early diagnostic screening and preventative strategies for young individuals at risk for mental diseases, such as male PCE offspring.
Collapse
Affiliation(s)
- Roberto Frau
- Department of Biomedical Sciences, Division of Neuroscience and Clinical Pharmacology, University of Cagliari, Monserrato, Italy
- The Guy Everett Laboratory for Neuroscience, University of Cagliari, Cagliari, Italy
| | - Miriam Melis
- Department of Biomedical Sciences, Division of Neuroscience and Clinical Pharmacology, University of Cagliari, Monserrato, Italy
| |
Collapse
|
32
|
Horackova H, Vachalova V, Abad C, Karahoda R, Staud F. Perfused rat term placenta as a preclinical model to investigate placental dopamine and norepinephrine transport. Clin Sci (Lond) 2023; 137:149-161. [PMID: 36598165 DOI: 10.1042/cs20220726] [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: 10/27/2022] [Revised: 12/22/2022] [Accepted: 01/04/2023] [Indexed: 01/05/2023]
Abstract
The placenta represents a non-neuronal organ capable of transporting and metabolizing monoamines. Since these bioactive molecules participate in numerous processes essential for placental and fetal physiology, any imbalance in their levels during pregnancy may affect brain development, projecting a higher risk of behavioral disorders in childhood or adulthood. Notably, the monoamine system in the placenta is a target of various psychoactive drugs and can be disrupted in several pregnancy pathologies. As research in pregnant women poses significant ethical restrictions, animal models are widely employed to study monoamine homeostasis as a mechanism involved in fetal programming. However, detailed knowledge of monoamine transport in the rat placenta is still lacking. Moreover, relatability to the human placental monoamine system is not examined. The present study provides insights into the transplacental monoamine dynamics between maternal and fetal circulation. We show that norepinephrine maternal-to-fetal transport is <4% due to high metabolism within the trophoblast. In contrast, dopamine maternal-to-fetal transport exceeds 25%, likely through passive transport across the membrane. In addition, we show high clearance of norepinephrine and dopamine from the fetal circulation mediated by the organic cation transporter 3 (OCT3). Altogether, we present transcriptional and functional evidence that the in situ rat placenta perfusion represents a suitable model for (patho)physiological investigation of dopamine and norepinephrine homeostasis in the fetoplacental unit. With the rapid advancements in drug discovery and environmental toxicity, the use of rat placenta as a preclinical model could facilitate screening of possible xenobiotic effects on monoamine homeostasis in the placenta.
Collapse
Affiliation(s)
- Hana Horackova
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Kralove, Charles University, Czech Republic
| | - Veronika Vachalova
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Kralove, Charles University, Czech Republic
| | - Cilia Abad
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Kralove, Charles University, Czech Republic
| | - Rona Karahoda
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Kralove, Charles University, Czech Republic
| | - Frantisek Staud
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Kralove, Charles University, Czech Republic
| |
Collapse
|
33
|
Prakash N. Developmental pathways linked to the vulnerability of adult midbrain dopaminergic neurons to neurodegeneration. Front Mol Neurosci 2022; 15:1071731. [PMID: 36618829 PMCID: PMC9815185 DOI: 10.3389/fnmol.2022.1071731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Accepted: 12/05/2022] [Indexed: 12/24/2022] Open
Abstract
The degeneration of dopaminergic and other neurons in the aging brain is considered a process starting well beyond the infantile and juvenile period. In contrast to other dopamine-associated neuropsychiatric disorders, such as schizophrenia and drug addiction, typically diagnosed during adolescence or young adulthood and, thus, thought to be rooted in the developing brain, Parkinson's Disease (PD) is rarely viewed as such. However, evidences have accumulated suggesting that several factors might contribute to an increased vulnerability to death of the dopaminergic neurons at an already very early (developmental) phase in life. Despite the remarkable ability of the brain to compensate such dopamine deficits, the early loss or dysfunction of these neurons might predispose an individual to suffer from PD because the critical threshold of dopamine function will be reached much earlier in life, even if the time-course and strength of naturally occurring and age-dependent dopaminergic cell death is not markedly altered in this individual. Several signaling and transcriptional pathways required for the proper embryonic development of the midbrain dopaminergic neurons, which are the most affected in PD, either continue to be active in the adult mammalian midbrain or are reactivated at the transition to adulthood and under neurotoxic conditions. The persistent activity of these pathways often has neuroprotective functions in adult midbrain dopaminergic neurons, whereas the reactivation of silenced pathways under pathological conditions can promote the survival and even regeneration of these neurons in the lesioned or aging brain. This article summarizes our current knowledge about signaling and transcription factors involved in midbrain dopaminergic neuron development, whose reduced gene dosage or signaling activity are implicated in a lower survival rate of these neurons in the postnatal or aging brain. It also discusses the evidences supporting the neuroprotection of the midbrain dopaminergic system after the external supply or ectopic expression of some of these secreted and nuclear factors in the adult and aging brain. Altogether, the timely monitoring and/or correction of these signaling and transcriptional pathways might be a promising approach to a much earlier diagnosis and/or prevention of PD.
Collapse
|
34
|
5-HT-dependent synaptic plasticity of the prefrontal cortex in postnatal development. Sci Rep 2022; 12:21015. [PMID: 36470912 PMCID: PMC9723183 DOI: 10.1038/s41598-022-23767-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 11/04/2022] [Indexed: 12/12/2022] Open
Abstract
Important functions of the prefrontal cortex (PFC) are established during early life, when neurons exhibit enhanced synaptic plasticity and synaptogenesis. This developmental stage drives the organization of cortical connectivity, responsible for establishing behavioral patterns. Serotonin (5-HT) emerges among the most significant factors that modulate brain activity during postnatal development. In the PFC, activated 5-HT receptors modify neuronal excitability and interact with intracellular signaling involved in synaptic modifications, thus suggesting that 5-HT might participate in early postnatal plasticity. To test this hypothesis, we employed intracellular electrophysiological recordings of PFC layer 5 neurons to study the modulatory effects of 5-HT on plasticity induced by theta-burst stimulation (TBS) in two postnatal periods of rats. Our results indicate that 5-HT is essential for TBS to result in synaptic changes during the third postnatal week, but not later. TBS coupled with 5-HT2A or 5-HT1A and 5-HT7 receptors stimulation leads to long-term depression (LTD). On the other hand, TBS and synergic activation of 5-HT1A, 5-HT2A, and 5-HT7 receptors lead to long-term potentiation (LTP). Finally, we also show that 5-HT dependent synaptic plasticity of the PFC is impaired in animals that are exposed to early-life chronic stress.
Collapse
|
35
|
Abrantes M, Rodrigues D, Domingues T, Nemala SS, Monteiro P, Borme J, Alpuim P, Jacinto L. Ultrasensitive dopamine detection with graphene aptasensor multitransistor arrays. J Nanobiotechnology 2022; 20:495. [DOI: 10.1186/s12951-022-01695-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 11/01/2022] [Indexed: 11/26/2022] Open
Abstract
AbstractDetecting physiological levels of neurotransmitters in biological samples can advance our understanding of brain disorders and lead to improved diagnostics and therapeutics. However, neurotransmitter sensors for real-world applications must reliably detect low concentrations of target analytes from small volume working samples. Herein, a platform for robust and ultrasensitive detection of dopamine, an essential neurotransmitter that underlies several brain disorders, based on graphene multitransistor arrays (gMTAs) functionalized with a selective DNA aptamer is presented. High-yield scalable methodologies optimized at the wafer level were employed to integrate multiple graphene transistors on small-size chips (4.5 × 4.5 mm). The multiple sensor array configuration permits independent and simultaneous replicate measurements of the same sample that produce robust average data, reducing sources of measurement variability. This procedure allowed sensitive and reproducible dopamine detection in ultra-low concentrations from small volume samples across physiological buffers and high ionic strength complex biological samples. The obtained limit-of-detection was 1 aM (10–18) with dynamic detection ranges spanning 10 orders of magnitude up to 100 µM (10–8), and a 22 mV/decade peak sensitivity in artificial cerebral spinal fluid. Dopamine detection in dopamine-depleted brain homogenates spiked with dopamine was also possible with a LOD of 1 aM, overcoming sensitivity losses typically observed in ion-sensitive sensors in complex biological samples. Furthermore, we show that our gMTAs platform can detect minimal changes in dopamine concentrations in small working volume samples (2 µL) of cerebral spinal fluid samples obtained from a mouse model of Parkinson’s Disease. The platform presented in this work can lead the way to graphene-based neurotransmitter sensors suitable for real-world academic and pre-clinical pharmaceutical research as well as clinical diagnosis.
Collapse
|
36
|
Alsanie WF, Abdelrahman S, Alhomrani M, Gaber A, Alosimi EA, Habeeballah H, Alkhatabi HA, Felimban RI, Hauser CAE, Tayeb HH, Alamri AS, Alamri A, Raafat BM, Alswat KA, Althobaiti YS, Asiri YA. The Influence of Prenatal Exposure to Quetiapine Fumarate on the Development of Dopaminergic Neurons in the Ventral Midbrain of Mouse Embryos. Int J Mol Sci 2022; 23:ijms232012352. [PMID: 36293205 PMCID: PMC9603924 DOI: 10.3390/ijms232012352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 10/07/2022] [Accepted: 10/12/2022] [Indexed: 11/06/2022] Open
Abstract
The effects of second-generation antipsychotics on prenatal neurodevelopment, apoptotic neurodegeneration, and postnatal developmental delays have been poorly investigated. Even at standard doses, the use of quetiapine fumarate (QEPF) in pregnant women might be detrimental to fetal development. We used primary mouse embryonic neurons to evaluate the disruption of morphogenesis and differentiation of ventral midbrain (VM) neurons after exposure to QEPF. The dopaminergic VM neurons were deliberately targeted due to their roles in cognition, motor activity, and behavior. The results revealed that exposure to QEPF during early brain development decreased the effects of the dopaminergic lineage-related genes Tyrosine hydroxylase(Th), Dopamine receptor D1 (Drd1), Dopamine transporter (Dat), LIM homeobox transcription factor 1 alfa (Lmx1a), and Cell adhesion molecule L1 (Chl1), and the senescent dopaminergic gene Pituitary homeobox 3 (Pitx3). In contrast, Brain derived neurotrophic factor (Bdnf) and Nuclear receptor-related 1 (Nurr1) expressions were significantly upregulated. Interestingly, QEPF had variable effects on the development of non-dopaminergic neurons in VM. An optimal dose of QEPF (10 µM) was found to insignificantly affect the viability of neurons isolated from the VM. It also instigated a non-significant reduction in adenosine triphosphate formation in these neuronal populations. Exposure to QEPF during the early stages of brain development could also hinder the formation of VM and their structural phenotypes. These findings could aid therapeutic decision-making when prescribing 2nd generation antipsychotics in pregnant populations.
Collapse
Affiliation(s)
- Walaa F. Alsanie
- Department of Clinical Laboratories Sciences, The Faculty of Applied Medical Sciences, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
- Centre of Biomedical Sciences Research (CBSR), Deanship of Scientific Research, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
- Correspondence:
| | - Sherin Abdelrahman
- Laboratory for Nanomedicine, Division of Biological and Environmental Science and Engineering (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal, Jeddah 23955, Saudi Arabia
| | - Majid Alhomrani
- Department of Clinical Laboratories Sciences, The Faculty of Applied Medical Sciences, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
- Centre of Biomedical Sciences Research (CBSR), Deanship of Scientific Research, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
| | - Ahmed Gaber
- Centre of Biomedical Sciences Research (CBSR), Deanship of Scientific Research, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
- Department of Biology, College of Science, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
| | - Ebtisam Abdulah Alosimi
- Centre of Biomedical Sciences Research (CBSR), Deanship of Scientific Research, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
| | - Hamza Habeeballah
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences in Rabigh, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Heba A. Alkhatabi
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Center of Excellence in Genomic Medicine Research (CEGMR), King Abdulaziz University, Jeddah 21589, Saudi Arabia
- King Fahd Medical Research Centre, Hematology Research Unit, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Raed I. Felimban
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Center of Innovation in Personalized Medicine (CIPM), 3D Bioprinting Unit, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Charlotte A. E. Hauser
- Laboratory for Nanomedicine, Division of Biological and Environmental Science and Engineering (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal, Jeddah 23955, Saudi Arabia
- Computational Bioscience Research Center (CBRC), King Abdullah University of Science and Technology (KAUST), Thuwal, Jeddah 23955, Saudi Arabia
| | - Hossam H. Tayeb
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Nanomedicine Unit, Center of Innovation in Personalized Medicine (CIPM), King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Abdulhakeem S. Alamri
- Department of Clinical Laboratories Sciences, The Faculty of Applied Medical Sciences, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
- Centre of Biomedical Sciences Research (CBSR), Deanship of Scientific Research, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
| | - Abdulwahab Alamri
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Hail, Hail 55211, Saudi Arabia
| | - Bassem M. Raafat
- Department of Radiological Sciences, College of Applied Medical Sciences, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
| | - Khaled A. Alswat
- Department of Internal Medicine, School of Medicine, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
| | - Yusuf S. Althobaiti
- Department of Pharmacology and Toxicology, College of Pharmacy, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
- Addiction and Neuroscience Research Unit, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
| | - Yousif A. Asiri
- Department of Clinical Pharmacy, College of Pharmacy, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
| |
Collapse
|
37
|
Vachalova V, Karahoda R, Ottaviani M, Anandam KY, Abad C, Albrecht C, Staud F. Functional reorganization of monoamine transport systems during villous trophoblast differentiation: evidence of distinct differences between primary human trophoblasts and BeWo cells. Reprod Biol Endocrinol 2022; 20:112. [PMID: 35927731 PMCID: PMC9351077 DOI: 10.1186/s12958-022-00981-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Accepted: 07/19/2022] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND Three primary monoamines-serotonin, norepinephrine, and dopamine-play major roles in the placenta-fetal brain axis. Analogously to the brain, the placenta has transport mechanisms that actively take up these monoamines into trophoblast cells. These transporters are known to play important roles in the differentiated syncytiotrophoblast layer, but their status and activities in the undifferentiated, progenitor cytotrophoblast cells are not well understood. Thus, we have explored the cellular handling and regulation of monoamine transporters during the phenotypic transitioning of cytotrophoblasts along the villous pathway. METHODS Experiments were conducted with two cellular models of syncytium development: primary trophoblast cells isolated from the human term placenta (PHT), and the choriocarcinoma-derived BeWo cell line. The gene and protein expression of membrane transporters for serotonin (SERT), norepinephrine (NET), dopamine (DAT), and organic cation transporter 3 (OCT3) was determined by quantitative PCR and Western blot analysis, respectively. Subsequently, the effect of trophoblast differentiation on transporter activity was analyzed by monoamine uptake into cells. RESULTS We present multiple lines of evidence of changes in the transcriptional and functional regulation of monoamine transporters associated with trophoblast differentiation. These include enhancement of SERT and DAT gene and protein expression in BeWo cells. On the other hand, in PHT cells we report negative modulation of SERT, NET, and OCT3 protein expression. We show that OCT3 is the dominant monoamine transporter in PHT cells, and its main functional impact is on serotonin uptake, while passive transport strongly contributes to norepinephrine and dopamine uptake. Further, we show that a wide range of selective serotonin reuptake inhibitors affect serotonin cellular accumulation, at pharmacologically relevant drug concentrations, via their action on both OCT3 and SERT. Finally, we demonstrate that BeWo cells do not well reflect the molecular mechanisms and properties of healthy human trophoblast cells. CONCLUSIONS Collectively, our findings provide insights into the regulation of monoamine transport during trophoblast differentiation and present important considerations regarding appropriate in vitro models for studying monoamine regulation in the placenta.
Collapse
Affiliation(s)
- Veronika Vachalova
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic
| | - Rona Karahoda
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic
| | - Martina Ottaviani
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic
| | - Kasin Yadunandam Anandam
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic
| | - Cilia Abad
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic
| | - Christiane Albrecht
- Institute of Biochemistry and Molecular Medicine, University of Bern, Bern, Switzerland
- Swiss National Centre of Competence in Research, NCCR TransCure, University of Bern, Bern, Switzerland
| | - Frantisek Staud
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic.
| |
Collapse
|
38
|
Fahmy HM, Mohamed ER, Hussein AA, Khadrawy YA, Ahmed NA. Evaluation of the therapeutic effect of mesoporous silica nanoparticles loaded with Gallic acid on reserpine-induced depression in Wistar rats. BMC Pharmacol Toxicol 2022; 23:40. [PMID: 35705968 PMCID: PMC9199140 DOI: 10.1186/s40360-022-00579-1] [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: 04/01/2022] [Accepted: 06/02/2022] [Indexed: 12/14/2022] Open
Abstract
Background The current study evaluates the free gallic acid (GA) and GA-loaded mesoporous silica nanoparticles (MSNs) antidepressant efficacy in a rat model of depression caused by reserpine. Methods By using a scanning electron microscope (SEM), dynamic light scattering (DLS), and zeta potential, MSNs and GA-loaded MSNs were characterized. The efficiency of encapsulation and the release of GA-loaded MSNs were also investigated. The effect of GA, either in its free form or loaded on (MSNs) on oxidative stress biomarkers and monoamine neurotransmitters levels (serotonin (5-HT), norepinephrine (NEP), and dopamine (DA)), were evaluated in these areas (cortex, hippocampus, striatum, and hypothalamus) of control, a depression model of rat, a depression model of rat treated with either free GA, MSNs or GA loaded MSNs. The forced swimming test (FST) also the open field test (OFT) were carried out to evaluate the behavioral changes in all groups. Results Reserpine caused a decrease in the time spent in motor and swimming activity besides increasing the time of immobility, as demonstrated by OFT and FST. Significantly reductions in 5-HT, NEP, and DA were obtained in the cortex, hippocampus, hypothalamus, and striatum of reserpine-treated rats. Free GA was more effective in increasing the serotonin level in the cortex, hippocampus, and hypothalamus, while GA-loaded MSNs were more effective in increasing it in the striatum. GA-loaded MSNs also increased the level of NEP in the four studied brain areas. Free GA increased dopamine levels in the cortex and striatum, whereas GA-loaded MSNs increased DA levels in the hippocampus and hypothalamus compared with the depressed untreated group. Conclusions MSNs can be used as a drug delivery system to target GA selectively to specific brain areas.
Collapse
Affiliation(s)
- Heba M Fahmy
- Biophysics Department, Faculty of Science, Cairo University, Giza, Egypt.
| | - Eman R Mohamed
- Zoology Department, Faculty of Science, Cairo University, Giza, Egypt
| | - Aida A Hussein
- Zoology Department, Faculty of Science, Suez University, Suez, Egypt
| | - Yasser A Khadrawy
- Medical Physiology Department, Medical Division, National Research Centre, Cairo, Egypt
| | - Nawal A Ahmed
- Zoology Department, Faculty of Science, Cairo University, Giza, Egypt
| |
Collapse
|
39
|
Bellon A, Feuillet V, Cortez-Resendiz A, Mouaffak F, Kong L, Hong LE, De Godoy L, Jay TM, Hosmalin A, Krebs MO. Dopamine-induced pruning in monocyte-derived-neuronal-like cells (MDNCs) from patients with schizophrenia. Mol Psychiatry 2022; 27:2787-2802. [PMID: 35365810 PMCID: PMC9156413 DOI: 10.1038/s41380-022-01514-w] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Revised: 02/05/2022] [Accepted: 02/25/2022] [Indexed: 01/10/2023]
Abstract
The long lapse between the presumptive origin of schizophrenia (SCZ) during early development and its diagnosis in late adolescence has hindered the study of crucial neurodevelopmental processes directly in living patients. Dopamine, a neurotransmitter consistently associated with the pathophysiology of SCZ, participates in several aspects of brain development including pruning of neuronal extensions. Excessive pruning is considered the cause of the most consistent finding in SCZ, namely decreased brain volume. It is therefore possible that patients with SCZ carry an increased susceptibility to dopamine's pruning effects and that this susceptibility would be more obvious in the early stages of neuronal development when dopamine pruning effects appear to be more prominent. Obtaining developing neurons from living patients is not feasible. Instead, we used Monocyte-Derived-Neuronal-like Cells (MDNCs) as these cells can be generated in only 20 days and deliver reproducible results. In this study, we expanded the number of individuals in whom we tested the reproducibility of MDNCs. We also deepened the characterization of MDNCs by comparing its neurostructure to that of human developing neurons. Moreover, we studied MDNCs from 12 controls and 13 patients with SCZ. Patients' cells differentiate more efficiently, extend longer secondary neurites and grow more primary neurites. In addition, MDNCs from medicated patients expresses less D1R and prune more primary neurites when exposed to dopamine. Haloperidol did not influence our results but the role of other antipsychotics was not examined and thus, needs to be considered as a confounder.
Collapse
Affiliation(s)
- Alfredo Bellon
- Department of Psychiatry and Behavioral Health, Penn State Hershey Medical Center, Hershey, PA, USA.
- Department of Pharmacology, Penn State Hershey Medical Center, Hershey, PA, USA.
| | - Vincent Feuillet
- Aix-Marseille University, CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy, Marseille, France
- Université de Paris, Institut Cochin, CNRS, INSERM, F-75014, Paris, France
| | - Alonso Cortez-Resendiz
- Department of Psychiatry and Behavioral Health, Penn State Hershey Medical Center, Hershey, PA, USA
| | - Faycal Mouaffak
- Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM U1266, Pathophysiology of Psychiatric Disorders, Université de Paris, Paris, France
- Pôle de Psychiatrie d'Adultes 93G04, EPS Ville Evrard, Saint Denis, France
| | - Lan Kong
- Department of Public Health Sciences, Penn State Hershey Medical Center, Hershey, PA, USA
| | - L Elliot Hong
- Department of Psychiatry, Maryland Psychiatric Research Center, University of Maryland School of Medicine, Baltimore, MD, USA
| | | | - Therese M Jay
- Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM U1266, Pathophysiology of Psychiatric Disorders, Université de Paris, Paris, France
| | - Anne Hosmalin
- Université de Paris, Institut Cochin, CNRS, INSERM, F-75014, Paris, France
| | - Marie-Odile Krebs
- Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM U1266, Pathophysiology of Psychiatric Disorders, Université de Paris, Paris, France
- Groupe-Hospitalo-Universitaire de Paris, Psychiatrie et Neuroscience, Pôle PEPIT, University of Paris, Paris, France
| |
Collapse
|
40
|
Saad AK, Akour A, Mahboob A, AbuRuz S, Sadek B. Role of Brain Modulators in Neurodevelopment: Focus on Autism Spectrum Disorder and Associated Comorbidities. Pharmaceuticals (Basel) 2022; 15:612. [PMID: 35631438 PMCID: PMC9144645 DOI: 10.3390/ph15050612] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 05/08/2022] [Accepted: 05/12/2022] [Indexed: 02/04/2023] Open
Abstract
Autism spectrum disorder (ASD) and associated neurodevelopmental disorders share similar pathogenesis and clinical features. Pathophysiological changes in these diseases are rooted in early neuronal stem cells in the uterus. Several genetic and environmental factors potentially perturb neurogenesis and synaptogenesis processes causing incomplete or altered maturation of the brain that precedes the symptomology later in life. In this review, the impact of several endogenous neuromodulators and pharmacological agents on the foetus during pregnancy, manifested on numerous aspects of neurodevelopment is discussed. Within this context, some possible insults that may alter these modulators and therefore alter their role in neurodevelopment are high-lighted. Sometimes, a particular insult could influence several neuromodulator systems as is supported by recent research in the field of ASD and associated disorders. Dopaminergic hy-pothesis prevailed on the table for discussion of the pathogenesis of schizophrenia (SCH), atten-tion-deficit hyperactivity disorder (ADHD) and ASD for a long time. However, recent cumulative evidence suggests otherwise. Indeed, the neuromodulators that are dysregulated in ASD and comorbid disorders are as diverse as the causes and symptoms of this disease. Additionally, these neuromodulators have roles in brain development, further complicating their involvement in comorbidity. This review will survey the current understanding of the neuromodulating systems to serve the pharmacological field during pregnancy and to minimize drug-related insults in pa-tients with ASD and associated comorbidity disorders, e.g., SCH or ADHD.
Collapse
Affiliation(s)
- Ali K. Saad
- Department of Pharmacology & Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 17666, United Arab Emirates; (A.K.S.); (A.A.); (S.A.)
- Zayed Center for Health Sciences, United Arab Emirates University, Al Ain P.O. Box 17666, United Arab Emirates
| | - Amal Akour
- Department of Pharmacology & Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 17666, United Arab Emirates; (A.K.S.); (A.A.); (S.A.)
- Zayed Center for Health Sciences, United Arab Emirates University, Al Ain P.O. Box 17666, United Arab Emirates
- Department of Biopharmaceutics and Clinical Pharmacy, School of Pharmacy, The University of Jordan, Amman P.O. Box 11942, Jordan
| | - Abdulla Mahboob
- Department of Chemistry, College of Sciences, United Arab Emirates University, Al-Ain P.O. Box 15551, United Arab Emirates;
| | - Salahdein AbuRuz
- Department of Pharmacology & Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 17666, United Arab Emirates; (A.K.S.); (A.A.); (S.A.)
- Zayed Center for Health Sciences, United Arab Emirates University, Al Ain P.O. Box 17666, United Arab Emirates
- Department of Biopharmaceutics and Clinical Pharmacy, School of Pharmacy, The University of Jordan, Amman P.O. Box 11942, Jordan
| | - Bassem Sadek
- Department of Pharmacology & Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 17666, United Arab Emirates; (A.K.S.); (A.A.); (S.A.)
- Zayed Center for Health Sciences, United Arab Emirates University, Al Ain P.O. Box 17666, United Arab Emirates
| |
Collapse
|
41
|
Catale C, Lo Iacono L, Martini A, Heil C, Guatteo E, Mercuri NB, Viscomi MT, Palacios D, Carola V. Early Life Social Stress Causes Sex- and Region-Dependent Dopaminergic Changes that Are Prevented by Minocycline. Mol Neurobiol 2022; 59:3913-3932. [PMID: 35435618 PMCID: PMC9148283 DOI: 10.1007/s12035-022-02830-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 04/02/2022] [Indexed: 02/03/2023]
Abstract
Early life stress (ELS) is known to modify trajectories of brain dopaminergic development, but the mechanisms underlying have not been determined. ELS perturbs immune system and microglia reactivity, and inflammation and microglia influence dopaminergic transmission and development. Whether microglia mediate the effects of ELS on dopamine (DA) system development is still unknown. We explored the effects of repeated early social stress on development of the dopaminergic system in male and female mice through histological, electrophysiological, and transcriptomic analyses. Furthermore, we tested whether these effects could be mediated by ELS-induced altered microglia/immune activity through a pharmacological approach. We found that social stress in early life altered DA neurons morphology, reduced dopamine transporter (DAT) and tyrosine hydroxylase expression, and lowered DAT-mediated currents in the ventral tegmental area but not substantia nigra of male mice only. Notably, stress-induced DA alterations were prevented by minocycline, an inhibitor of microglia activation. Transcriptome analysis in the developing male ventral tegmental area revealed that ELS caused downregulation of dopaminergic transmission and alteration in hormonal and peptide signaling pathways. Results from this study offer new insight into the mechanisms of stress response and altered brain dopaminergic maturation after ELS, providing evidence of neuroimmune interaction, sex differences, and regional specificity.
Collapse
Affiliation(s)
- Clarissa Catale
- Division of Experimental Neuroscience, Neurobiology of Behavior Laboratory, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Luisa Lo Iacono
- Department of Dynamic and Clinical Psychology, and Health Studies, Sapienza University of Rome, Via degli Apuli 1, Rome, Italy
| | - Alessandro Martini
- Division of Experimental Neuroscience, Experimental Neurology Laboratory, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Constantin Heil
- Division of Experimental Neuroscience, Epigenetics and Signal Transduction Laboratory, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Ezia Guatteo
- Division of Experimental Neuroscience, Experimental Neurology Laboratory, IRCCS Santa Lucia Foundation, Rome, Italy
- Department of Motor Science and Wellness, University of Naples Parthenope, Naples, Italy
| | - Nicola Biagio Mercuri
- Division of Experimental Neuroscience, Experimental Neurology Laboratory, IRCCS Santa Lucia Foundation, Rome, Italy
- Department of Systems Medicine, Università Degli Studi Di Roma Tor Vergata, Rome, Italy
| | - Maria Teresa Viscomi
- Department of Life Science and Public Health, Section of Histology and Embryology, Università Cattolica Del S. Cuore, Rome, Italy
- IRCCS Fondazione Policlinico Universitario A. Gemelli, Rome, Italy
| | - Daniela Palacios
- Division of Experimental Neuroscience, Epigenetics and Signal Transduction Laboratory, IRCCS Santa Lucia Foundation, Rome, Italy
- IRCCS Fondazione Policlinico Universitario A. Gemelli, Rome, Italy
- Department of Life Science and Public Health, Section of Biology, Università Cattolica Del S. Cuore, Rome, Italy
| | - Valeria Carola
- Division of Experimental Neuroscience, Neurobiology of Behavior Laboratory, IRCCS Santa Lucia Foundation, Rome, Italy.
- Department of Dynamic and Clinical Psychology, and Health Studies, Sapienza University of Rome, Via degli Apuli 1, Rome, Italy.
| |
Collapse
|
42
|
Oldehinkel M, Llera A, Faber M, Huertas I, Buitelaar JK, Bloem BR, Marquand AF, Helmich R, Haak KV, Beckmann CF. Mapping dopaminergic projections in the human brain with resting-state fMRI. eLife 2022; 11:71846. [PMID: 35113016 PMCID: PMC8843090 DOI: 10.7554/elife.71846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 01/26/2022] [Indexed: 12/02/2022] Open
Abstract
The striatum receives dense dopaminergic projections, making it a key region of the dopaminergic system. Its dysfunction has been implicated in various conditions including Parkinson’s disease (PD) and substance use disorder. However, the investigation of dopamine-specific functioning in humans is problematic as current MRI approaches are unable to differentiate between dopaminergic and other projections. Here, we demonstrate that ‘connectopic mapping’ – a novel approach for characterizing fine-grained, overlapping modes of functional connectivity – can be used to map dopaminergic projections in striatum. We applied connectopic mapping to resting-state functional MRI data of the Human Connectome Project (population cohort; N = 839) and selected the second-order striatal connectivity mode for further analyses. We first validated its specificity to dopaminergic projections by demonstrating a high spatial correlation (r = 0.884) with dopamine transporter availability – a marker of dopaminergic projections – derived from DaT SPECT scans of 209 healthy controls. Next, we obtained the subject-specific second-order modes from 20 controls and 39 PD patients scanned under placebo and under dopamine replacement therapy (L-DOPA), and show that our proposed dopaminergic marker tracks PD diagnosis, symptom severity, and sensitivity to L-DOPA. Finally, across 30 daily alcohol users and 38 daily smokers, we establish strong associations with self-reported alcohol and nicotine use. Our findings provide evidence that the second-order mode of functional connectivity in striatum maps onto dopaminergic projections, tracks inter-individual differences in PD symptom severity and L-DOPA sensitivity, and exhibits strong associations with levels of nicotine and alcohol use, thereby offering a new biomarker for dopamine-related (dys)function in the human brain.
Collapse
Affiliation(s)
- Marianne Oldehinkel
- Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Centre, Nijmegen, Netherlands
| | - Alberto Llera
- Donders Institute for Brain, Cognition and Behaviour, Radboud, Radboud University Medical Centre, Nijmegen, Netherlands
| | - Myrthe Faber
- Donders Institute for Brain, Cognition and Behaviour, Radboud, Radboud University Medical Centre, Nijmegen, Netherlands
| | - Ismael Huertas
- Institute of Biomedicine of Seville (IBiS), Seville, Spain
| | - Jan K Buitelaar
- Donders Institute for Brain, Cognition and Behaviour, Radboud, Radboud University Medical Center, Nijmegen, Netherlands
| | - Bastiaan R Bloem
- Department of Neurology, Radboud University Medical Centre, Nijmegen, Netherlands
| | - Andre F Marquand
- Donders Institute for Brain, Cognition and Behaviour, Radboud, Radboud University Medical Centre, Nijmegen, Netherlands
| | - Rick Helmich
- Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Centre, Nijmegen, Netherlands
| | - Koen V Haak
- Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Centre, Nijmegen, Netherlands
| | - Christian F Beckmann
- Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Centre, Nijmegen, Netherlands
| |
Collapse
|
43
|
Philyaw TJ, Rothenfluh A, Titos I. The Use of Drosophila to Understand Psychostimulant Responses. Biomedicines 2022; 10:119. [PMID: 35052798 PMCID: PMC8773124 DOI: 10.3390/biomedicines10010119] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 12/31/2021] [Accepted: 12/31/2021] [Indexed: 01/27/2023] Open
Abstract
The addictive properties of psychostimulants such as cocaine, amphetamine, methamphetamine, and methylphenidate are based on their ability to increase dopaminergic neurotransmission in the reward system. While cocaine and methamphetamine are predominately used recreationally, amphetamine and methylphenidate also work as effective therapeutics to treat symptoms of disorders including attention deficit and hyperactivity disorder (ADHD) and autism spectrum disorder (ASD). Although both the addictive properties of psychostimulant drugs and their therapeutic efficacy are influenced by genetic variation, very few genes that regulate these processes in humans have been identified. This is largely due to population heterogeneity which entails a requirement for large samples. Drosophila melanogaster exhibits similar psychostimulant responses to humans, a high degree of gene conservation, and allow performance of behavioral assays in a large population. Additionally, amphetamine and methylphenidate reduce impairments in fly models of ADHD-like behavior. Therefore, Drosophila represents an ideal translational model organism to tackle the genetic components underlying the effects of psychostimulants. Here, we break down the many assays that reliably quantify the effects of cocaine, amphetamine, methamphetamine, and methylphenidate in Drosophila. We also discuss how Drosophila is an efficient and cost-effective model organism for identifying novel candidate genes and molecular mechanisms involved in the behavioral responses to psychostimulant drugs.
Collapse
Affiliation(s)
- Travis James Philyaw
- Molecular Biology Graduate Program, University of Utah, Salt Lake City, UT 84112, USA;
| | - Adrian Rothenfluh
- Department of Psychiatry, Huntsman Mental Health Institute, University of Utah, Salt Lake City, UT 84108, USA
- Molecular Medicine Program, University of Utah, Salt Lake City, UT 84112, USA
- Department of Neurobiology, University of Utah, Salt Lake City, UT 84132, USA
- Department of Human Genetics, University of Utah, Salt Lake City, UT 84112, USA
| | - Iris Titos
- Molecular Medicine Program, University of Utah, Salt Lake City, UT 84112, USA
| |
Collapse
|
44
|
Abstract
During evolution, the cerebral cortex advances by increasing in surface and the introduction of new cytoarchitectonic areas among which the prefrontal cortex (PFC) is considered to be the substrate of highest cognitive functions. Although neurons of the PFC are generated before birth, the differentiation of its neurons and development of synaptic connections in humans extend to the 3rd decade of life. During this period, synapses as well as neurotransmitter systems including their receptors and transporters, are initially overproduced followed by selective elimination. Advanced methods applied to human and animal models, enable investigation of the cellular mechanisms and role of specific genes, non-coding regulatory elements and signaling molecules in control of prefrontal neuronal production and phenotypic fate, as well as neuronal migration to establish layering of the PFC. Likewise, various genetic approaches in combination with functional assays and immunohistochemical and imaging methods reveal roles of neurotransmitter systems during maturation of the PFC. Disruption, or even a slight slowing of the rate of neuronal production, migration and synaptogenesis by genetic or environmental factors, can induce gross as well as subtle changes that eventually can lead to cognitive impairment. An understanding of the development and evolution of the PFC provide insight into the pathogenesis and treatment of congenital neuropsychiatric diseases as well as idiopathic developmental disorders that cause intellectual disabilities.
Collapse
Affiliation(s)
- Sharon M Kolk
- Department of Molecular Neurobiology, Donders Institute for Brain, Cognition and Behaviour and Faculty of Science, Radboud University, Nijmegen, The Netherlands.
| | - Pasko Rakic
- Department of Neuroscience and Kavli Institute for Neuroscience, Yale University, New Haven, Connecticut, USA.
| |
Collapse
|
45
|
Guo Z, Li S, Wu J, Zhu X, Zhang Y. Maternal Deprivation Increased Vulnerability to Depression in Adult Rats Through DRD2 Promoter Methylation in the Ventral Tegmental Area. Front Psychiatry 2022; 13:827667. [PMID: 35308874 PMCID: PMC8924051 DOI: 10.3389/fpsyt.2022.827667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 01/21/2022] [Indexed: 11/13/2022] Open
Abstract
OBJECTIVE Early life adversity is a risk factor for depression in adulthood; however, the underlying mechanisms are not well understood. This study aims to investigate the effect of DNA methylation of DRD2 gene on early life stress-induced depression in adult rats. METHODS Newborn Sprague-Dawley rats were randomly assigned to four groups: maternal deprivation group (MD), chronic unpredictable stress (CUS) group, maternal deprivation plus chronic unpredictable stress (MD/CUS) group, and normal control group (NOR). Behaviors were measured by open field test (OFT), sucrose preference test (SPT), and Original Research Article forced swimming test (FST). Fecal CORT level was detected by ELISA. Bisulfite amplicon sequencing PCR was used to assess methylation levels of DRD2 promoter. RESULTS CUS and MD/CUS rats had a significantly shorter total distance, longer immobility time, and higher CORT level, while MD and MD/CUS rats had a significantly lower percentage of central distance, more feces, lower rate of sucrose preference, and lower levels of DRD2 protein and mRNA in the VTA than NOR rats. CUS rats showed a significantly higher DRD2 mRNA and protein levels in the VTA than NOR rats. CUS, MD, and MD/CUS rats showed a significantly higher level of DRD2 promoter methylation than NOR rats. CORT level was significantly correlated with the sucrose preference rate in SPT, the immobility time in FST, the total distance, and the number of fecal pellets in OFT. DRD2 protein level was significantly correlated with the sucrose preference rate and the number of fecal pellets. DRD2 mRNA level was significantly correlated with the percentage of central distance and the number of fecal pellets in OFT. The level of DRD2 promoter methylation was significantly correlated with the sucrose preference rate, immobility time, total distance, the percentage of central distance, and the number of fecal pellets. CONCLUSIONS Early life MD increased vulnerability to stress-induced depressive-like behavior in adult rats. Enhanced DRD2 promoter methylation in the VTA may increase the susceptibility to depression.
Collapse
Affiliation(s)
- Zhenli Guo
- Medical Psychological Center, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Shansi Li
- Medical Psychological Center, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Jialing Wu
- Medical Psychological Center, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Xiongzhao Zhu
- Medical Psychological Center, The Second Xiangya Hospital, Central South University, Changsha, China.,Medical Psychological Institute of Central South University, Central South University, Changsha, China.,National Clinical Research Center for Mental Disorders, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Yi Zhang
- Medical Psychological Center, The Second Xiangya Hospital, Central South University, Changsha, China.,Medical Psychological Institute of Central South University, Central South University, Changsha, China.,National Clinical Research Center for Mental Disorders, The Second Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
46
|
Dopamine signaling impairs ROS modulation by mitochondrial hexokinase in human neural progenitor cells. Biosci Rep 2021; 41:230295. [PMID: 34821365 PMCID: PMC8661505 DOI: 10.1042/bsr20211191] [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: 05/19/2021] [Revised: 10/20/2021] [Accepted: 11/09/2021] [Indexed: 12/17/2022] Open
Abstract
Dopamine signaling has numerous roles during brain development. In addition, alterations in dopamine signaling may be also involved in the pathophysiology of psychiatric disorders. Neurodevelopment is modulated in multiple steps by reactive oxygen species (ROS), byproducts of oxidative metabolism that are signaling factors involved in proliferation, differentiation, and migration. Hexokinase (HK), when associated with the mitochondria (mt-HK), is a potent modulator of the generation of mitochondrial ROS in the brain. In the present study, we investigated whether dopamine could affect both the activity and redox function of mt-HK in human neural progenitor cells (NPCs). We found that dopamine signaling via D1R decreases mt-HK activity and impairs ROS modulation, which is followed by an expressive release of H2O2 and impairment in calcium handling by the mitochondria. Nevertheless, mitochondrial respiration is not affected, suggesting specificity for dopamine on mt-HK function. In neural stem cells (NSCs) derived from induced-pluripotent stem cells (iPSCs) of schizophrenia patients, mt-HK is unable to decrease mitochondrial ROS, in contrast with NSCs derived from healthy individuals. Our data point to mitochondrial hexokinase as a novel target of dopaminergic signaling, as well as a redox modulator in human neural progenitor cells, which may be relevant to the pathophysiology of neurodevelopmental disorders such as schizophrenia.
Collapse
|
47
|
van Rhijn JR, Shi Y, Bormann M, Mossink B, Frega M, Recaioglu H, Hakobjan M, Klein Gunnewiek T, Schoenmaker C, Palmer E, Faivre L, Kittel-Schneider S, Schubert D, Brunner H, Franke B, Nadif Kasri N. Brunner syndrome associated MAOA mutations result in NMDAR hyperfunction and increased network activity in human dopaminergic neurons. Neurobiol Dis 2021; 163:105587. [PMID: 34923109 DOI: 10.1016/j.nbd.2021.105587] [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: 07/13/2021] [Revised: 12/01/2021] [Accepted: 12/15/2021] [Indexed: 01/15/2023] Open
Abstract
Monoamine neurotransmitter abundance affects motor control, emotion, and cognitive function and is regulated by monoamine oxidases. Among these, Monoamine oxidase A (MAOA) catalyzes the degradation of dopamine, norepinephrine, and serotonin into their inactive metabolites. Loss-of-function mutations in the X-linked MAOA gene have been associated with Brunner syndrome, which is characterized by various forms of impulsivity, maladaptive externalizing behavior, and mild intellectual disability. Impaired MAOA activity in individuals with Brunner syndrome results in bioamine aberration, but it is currently unknown how this affects neuronal function, specifically in dopaminergic (DA) neurons. Here we generated human induced pluripotent stem cell (hiPSC)-derived DA neurons from three individuals with Brunner syndrome carrying different mutations and characterized neuronal properties at the single cell and neuronal network level in vitro. DA neurons of Brunner syndrome patients showed reduced synaptic density but exhibited hyperactive network activity. Intrinsic functional properties and α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor (AMPAR)-mediated synaptic transmission were not affected in DA neurons of individuals with Brunner syndrome. Instead, we show that the neuronal network hyperactivity is mediated by upregulation of the GRIN2A and GRIN2B subunits of the N-methyl-d-aspartate receptor (NMDAR), resulting in increased NMDAR-mediated currents. By correcting a MAOA missense mutation with CRISPR/Cas9 genome editing we normalized GRIN2A and GRIN2B expression, NMDAR function and neuronal population activity to control levels. Our data suggest that MAOA mutations in Brunner syndrome increase the activity of dopaminergic neurons through upregulation of NMDAR function, which may contribute to the etiology of Brunner syndrome associated phenotypes.
Collapse
Affiliation(s)
- Jon-Ruben van Rhijn
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition and Behavior, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Yan Shi
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behavior, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Maren Bormann
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition and Behavior, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Britt Mossink
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition and Behavior, Radboud University Medical Center, Nijmegen, the Netherlands; Department of Human Genetics, Donders Institute for Brain, Cognition and Behavior, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Monica Frega
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behavior, Radboud University Medical Center, Nijmegen, the Netherlands; Department of Clinical neurophysiology, University of Twente, 7522 NB Enschede, Netherlands
| | - Hatice Recaioglu
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition and Behavior, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Marina Hakobjan
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behavior, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Teun Klein Gunnewiek
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behavior, Radboud University Medical Center, Nijmegen, the Netherlands; Department of Anatomy, Donders Institute for Brain, Cognition and Behavior, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Chantal Schoenmaker
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behavior, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Elizabeth Palmer
- Genetics of Learning Disability Service, Hunter Genetics, Waratah, NSW, Australia; School of Women's and Children's Health, University of New South Wales, Randwick, NSW, Australia
| | - Laurence Faivre
- Centre de Référence Anomalies du développement et Syndromes malformatifs and FHU TRANSLAD, Hôpital d'Enfants, Dijon, France; INSERM UMR1231 GAD, Faculté de Médecine, Université de Bourgogne, Dijon, France
| | - Sarah Kittel-Schneider
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital, Goethe-University, Frankfurt, Germany; Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital Würzburg, Würzburg, Germany
| | - Dirk Schubert
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition and Behavior, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Han Brunner
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behavior, Radboud University Medical Center, Nijmegen, the Netherlands; Department of Clinical Genetics, MUMC+, GROW School of Oncology and Developmental Biology, and MHeNS School of Neuroscience and Maastricht University, Maastricht, the Netherlands
| | - Barbara Franke
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behavior, Radboud University Medical Center, Nijmegen, the Netherlands; Department of Psychiatry, Donders Institute for Brain, Cognition and Behavior, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Nael Nadif Kasri
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition and Behavior, Radboud University Medical Center, Nijmegen, the Netherlands; Department of Human Genetics, Donders Institute for Brain, Cognition and Behavior, Radboud University Medical Center, Nijmegen, the Netherlands.
| |
Collapse
|
48
|
da Silva SR, Kalaba P, Fabišiková A, Zehl M, Dragačević V, Dos Anjos LR, Neill PJ, Wieder M, Prado-Roller A, Gajic N, Palaretti V, da Silva GVJ, Pifl C, Lubec G, Gonzalez ERP. Synthesis and dopamine receptor binding of dihydrexidine and SKF 38393 catecholamine-based analogues. Amino Acids 2021; 54:85-98. [PMID: 34842969 DOI: 10.1007/s00726-021-03106-4] [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: 10/11/2021] [Accepted: 11/17/2021] [Indexed: 10/19/2022]
Abstract
Dopamine is an important neurotransmitter that regulates numerous essential functions, including cognition and voluntary movement. As such, it serves as an important scaffold for synthesis of novel analogues as part of drug development effort to obtain drugs for treatment of neurodegenerative diseases, such as Parkinson's disease. To that end, similarity search of the ZINC database based on two known dopamine-1 receptor (D1R) agonists, dihydrexidine (DHX) and SKF 38393, respectively, was used to predict novel chemical entities with potential binding to D1R. Three compounds that showed the highest similarity index were selected for synthesis and bioactivity profiling. All main synthesis products as well as the isolated intermediates, were properly characterized. The physico-chemical analyses were performed using HRESIMS, GC/MS, LC/MS with UV-Vis detection, and FTIR, 1H NMR and 13C NMR spectroscopy. Binding to D1 and D2 receptors and inhibition of dopamine reuptake via dopamine transporter were measured for the synthesized analogues of DHX and SKF 38393.
Collapse
Affiliation(s)
- Suzane Rosa da Silva
- Laboratory of Fine Organic Chemistry, Department of Chemistry and Biochemistry, Faculty of Sciences and Technology of São Paulo State University, Presidente Prudente, São Paulo, 19060-900, Brazil
| | - Predrag Kalaba
- Department of Pharmaceutical Chemistry, Faculty of Life Sciences, University of Vienna, Althanstraße 14, 1090, Vienna, Austria
| | - Anna Fabišiková
- Mass Spectrometry Centre, Faculty of Chemistry, University of Vienna, Währinger Straße 38, 1090, Vienna, Austria
| | - Martin Zehl
- Mass Spectrometry Centre, Faculty of Chemistry, University of Vienna, Währinger Straße 38, 1090, Vienna, Austria.,Department of Analytical Chemistry, Faculty of Chemistry, University of Vienna, Währinger Straße 38, 1090, Vienna, Austria
| | - Vladimir Dragačević
- Department of Pharmaceutical Chemistry, Faculty of Life Sciences, University of Vienna, Althanstraße 14, 1090, Vienna, Austria
| | - Luana Ribeiro Dos Anjos
- Laboratory of Fine Organic Chemistry, Department of Chemistry and Biochemistry, Faculty of Sciences and Technology of São Paulo State University, Presidente Prudente, São Paulo, 19060-900, Brazil
| | - Philip John Neill
- Department of Pharmaceutical Chemistry, Faculty of Life Sciences, University of Vienna, Althanstraße 14, 1090, Vienna, Austria
| | - Marcus Wieder
- Department of Pharmaceutical Chemistry, Faculty of Life Sciences, University of Vienna, Althanstraße 14, 1090, Vienna, Austria
| | - Alexander Prado-Roller
- Centre for X-Ray Structure Analysis at Faculty of Chemistry, University of Vienna, Währinger Straße 40-42, 1090, Vienna, Austria
| | - Natalie Gajic
- Centre for X-Ray Structure Analysis at Faculty of Chemistry, University of Vienna, Währinger Straße 40-42, 1090, Vienna, Austria
| | - Vinicius Palaretti
- Department of Chemistry, Faculty of Philosophy, Sciences and Letters of Ribeirão Preto, University of São Paulo, Avenida dos Bandeirantes, 3900, Ribeirão Preto, SP, 14040-901, Brazil
| | - Gil Valdo Jose da Silva
- Department of Chemistry, Faculty of Philosophy, Sciences and Letters of Ribeirão Preto, University of São Paulo, Avenida dos Bandeirantes, 3900, Ribeirão Preto, SP, 14040-901, Brazil
| | - Christian Pifl
- Department of Molecular Neurosciences, Centre for Brain Research, Medical University of Vienna, Spitalgasse 4, 1090, Vienna, Austria
| | - Gert Lubec
- Department of Neuroproteomics, Paracelsus Medical University, 5020, Salzburg, Austria.
| | - Eduardo R Perez Gonzalez
- Laboratory of Fine Organic Chemistry, Department of Chemistry and Biochemistry, Faculty of Sciences and Technology of São Paulo State University, Presidente Prudente, São Paulo, 19060-900, Brazil.
| |
Collapse
|
49
|
Zhang S, Jiao Z, Zhao X, Sun M, Feng X. Environmental exposure to 17β-trenbolone during adolescence inhibits social interaction in male mice. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2021; 289:117710. [PMID: 34243057 DOI: 10.1016/j.envpol.2021.117710] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 06/10/2021] [Accepted: 07/02/2021] [Indexed: 06/13/2023]
Abstract
Puberty is a critical period for growth and development. This period is sensitive to external stimuli, which ultimately affects the development of nerves and the formation of social behaviour. 17β-Trenbolone (17β-TBOH) is an endocrine disrupting chemicals (EDCs), which had been widely reported in aquatic vertebrates. But there is little known about the effects of 17β-TBOH on mammals, especially on adolescent neurodevelopment. In this study, we found that 17β-TBOH acute 1 h exposure can cause the activation of the dopamine circuit in pubertal male balb/c mice. At present, there is little known about the effects of puberty exposure of endocrine disruptors on these neurons/nerve pathways. Through a series of behavioural tests, exposure to 80 μgkg-1 d-1 of 17β-TBOH during adolescence increased the anxiety-like behaviour of mice and reduced the control of wheel-running behaviour and the response of social interaction behaviour. The results of TH immunofluorescence staining showed that exposure to 17β-TBOH reduced dopamine axon growth in the medial prefrontal cortex (mPFC). In addition, the results of real-time PCR showed that exposure to 17β-TBOH not only down-regulated the expression of dopamine axon development genes, but also affected the balance of excitatory/inhibitory signals in mPFC. In this research, we reveal the effects of 17β-TBOH exposure during adolescence on mammalian behaviour and neurodevelopment, and provide a reference for studying the origin of adolescent diseases.
Collapse
Affiliation(s)
- Shaozhi Zhang
- College of Life Science, The Key Laboratory of Bioactive Materials, Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300071, China
| | - Zihao Jiao
- The Institute of Robotics and Automatic Information Systems, Nankai University, Tianjin, 300071, China
| | - Xin Zhao
- The Institute of Robotics and Automatic Information Systems, Nankai University, Tianjin, 300071, China
| | - Mingzhu Sun
- The Institute of Robotics and Automatic Information Systems, Nankai University, Tianjin, 300071, China
| | - Xizeng Feng
- College of Life Science, The Key Laboratory of Bioactive Materials, Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300071, China.
| |
Collapse
|
50
|
Vassoler FM, Wimmer ME. Consequences of Parental Opioid Exposure on Neurophysiology, Behavior, and Health in the Next Generations. Cold Spring Harb Perspect Med 2021; 11:a040436. [PMID: 32601130 PMCID: PMC8485740 DOI: 10.1101/cshperspect.a040436] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Substance abuse and the ongoing opioid epidemic represents a large societal burden. This review will consider the long-term impact of opioid exposure on future generations. Prenatal, perinatal, and preconception exposure are reviewed with discussion of both maternal and paternal influences. Opioid exposure can have long-lasting effects on reproductive function, gametogenesis, and germline epigenetic programming, which can influence embryogenesis and alter the developmental trajectory of progeny. The potential mechanisms by which preconception maternal and paternal opioid exposure produce deleterious consequences on the health, behavior, and physiology of offspring that have been identified by clinical and animal studies will be discussed. The timing, nature, dosing, and duration of prenatal opioid exposure combined with other important environmental considerations influence the extent to which these manipulations affect parents and their progeny. Epigenetic inheritance refers to the transmission of environmental insults across generations via mechanisms independent of the DNA sequence. This topic will be further explored in the context of prenatal, perinatal, and preconception opioid exposure for both the maternal and paternal lineage.
Collapse
Affiliation(s)
- Fair M Vassoler
- Tufts University, Cummings School of Veterinary Medicine, Grafton, Massachusetts 01536, USA
| | - Mathieu E Wimmer
- Department of Psychology and Program in Neuroscience, Temple University, Philadelphia, Pennsylvania 19122, USA
| |
Collapse
|