1
|
Sarkisian KI, Yang JL, Marshall C, Stanczyk FZ. Allopregnanolone in the pathogenesis of the psychiatric comorbidities of polycystic ovarian syndrome. J Steroid Biochem Mol Biol 2025; 250:106719. [PMID: 40064425 DOI: 10.1016/j.jsbmb.2025.106719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 02/25/2025] [Accepted: 03/01/2025] [Indexed: 03/18/2025]
Abstract
Polycystic ovarian syndrome (PCOS) is an endocrine disorder affecting 10-15 % of women of reproductive age, with significant implications for both physical and mental health. Several recent research studies have examined the connection between PCOS and psychiatric disorders; however, the mechanism linking the two is not fully understood. Allopregnanolone is a neurosteroid that modulates GABAA receptors and is naturally affected by the pathophysiology of PCOS. It is thought to play a role in mood disorders, including premenstrual dysphoric disorder and postpartum depression. Recent research has begun to focus on the relationship between PCOS and allopregnanolone. A literature review was conducted using databases, including PubMed, MEDLINE, and Cochrane Library. Keywords included "PCOS," "psychiatric disorders," "allopregnanolone," and "neurosteroids." Articles were selected based on relevance to psychiatric implications of PCOS, with a focus on high-quality, original research studies. Quality assessment of the sources was informed using the Grades of Recommendation, Assessment, Development, and Evaluation (GRADE) Handbook criteria. The literature review revealed a growing body of evidence suggesting a strong association between PCOS and an increased risk of psychiatric disorders, particularly depression, anxiety, and mood disorders. The role of allopregnanolone, a neurosteroid, was identified as an important factor in this relationship, with some studies indicating its potential impact on mood regulation in PCOS patients. There is a dire need for clinicians to consider the mental health implications of PCOS during diagnosis and management. The integration of psychiatric screening in PCOS management could lead to earlier detection and improved outcomes. Future research should focus on the therapeutic potential of allopregnanolone and other neurosteroids in treating psychiatric disorders associated with PCOS.
Collapse
Affiliation(s)
- Karis I Sarkisian
- Department of Obstetrics and Gynecology, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States; University of California, Berkeley, United States.
| | - Jane L Yang
- Department of Obstetrics and Gynecology, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | | | - Frank Z Stanczyk
- Department of Obstetrics and Gynecology, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States.
| |
Collapse
|
2
|
Shi Q, Rammes G, Wang P, Xia C, Mou F, Zhu J, Guo H, Shao S, Wang X. Effects of Xenon on the Developing Brain: Current Insights from Pre-clinical and Clinical Studies. J Integr Neurosci 2025; 24:26388. [PMID: 40152563 DOI: 10.31083/jin26388] [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: 09/01/2024] [Revised: 10/19/2024] [Accepted: 10/31/2024] [Indexed: 03/29/2025] Open
Abstract
Research has indicated that general anesthetics may potentially induce neuroapoptosis, resulting in long-term neurological deficits in the developing brain. Fortunately, xenon, a noble gas, emerges as a promising candidate for an ideal anesthetic due to its favorable properties, offering neuroprotection and mitigating the neurotoxic effects of other general anesthetics during early life stages. Nevertheless, it is important to highlight that xenon has also been observed to cause neuroapoptosis in the neonatal brain, suggesting that xenon possesses both neuroprotective qualities (as evidenced by pre-clinical and clinical studies) and neurotoxic potential (based mainly on pre-clinical evidence) during brain development. To gain a comprehensive understanding the effects xenon, this review will explore the anesthetic properties of xenon, examine its effects on anesthesia, and elucidate its mechanisms of potential neuroprotection and neurotoxicity in the developing brain. The primary emphasis will be on xenon's application in the context of anesthetic-induced developmental neurotoxicity (AIDN), hypoxic-ischemic encephalopathy (HIE), and teratogenicity, aiming to provide valuable insights for pediatricians, pediatric anesthesiologists, and other healthcare professionals involved in the use and study of xenon anesthesia.
Collapse
Affiliation(s)
- Qinfang Shi
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022 Wuhan, Hubei, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022 Wuhan, Hubei, China
- Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, 430022 Wuhan, Hubei, China
| | - Gerhard Rammes
- Department of Anesthesiology and Intensive Care Medicine, School of Medicine and Health, Klinikum Rechts der Isar, Technical University of Munich, 81675 Munich, Germany
| | - Ping Wang
- Department of Anesthesiology, Shenzhen University General Hospital, 518071 Shenzhen, Guangdong, China
| | - Chengkun Xia
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022 Wuhan, Hubei, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022 Wuhan, Hubei, China
- Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, 430022 Wuhan, Hubei, China
| | - Fangfang Mou
- School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, 201203 Shanghai, China
| | - Jing Zhu
- School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, 201203 Shanghai, China
| | - Haidong Guo
- School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, 201203 Shanghai, China
| | - Shuijin Shao
- School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, 201203 Shanghai, China
| | - Xingxing Wang
- School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, 201203 Shanghai, China
| |
Collapse
|
3
|
Green CS, Morris JM, Magnuson JT, Leads RR, Lay CR, Gielazyn M, Rosman L, Schlenk D, Roberts AP. Exposure to the Polychlorinated biphenyl mixture Aroclor 1254 elicits neurological and cardiac developmental effects in early life stage zebrafish (Danio rerio). CHEMOSPHERE 2025; 371:144023. [PMID: 39724984 DOI: 10.1016/j.chemosphere.2024.144023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 11/25/2024] [Accepted: 12/22/2024] [Indexed: 12/28/2024]
Abstract
The goal of this study was to compare the bioaccumulation of the PCB mixture Aroclor 1254 in zebrafish to cardiac and neurologic outcomes. The establishment of effect concentrations (ECs) for cardiac and neurotoxic effects of PCBs in early life stage fish is challenging due to a lack of measured PCB concentrations in test media (e.g., fish tissue), the lack of standard exposure methods, and the propensity of PCBs to adsorb to test glassware and materials resulting in discrepancies in ECs from different studies with similar endpoints. Reporting tissue concentrations in test organisms will allow for standardization across different tests and thus may improve estimations of effect thresholds. Early life stage zebrafish (Danio rerio) are a common environmental toxicological model well represented within the literature, making them ideal for comparisons across multiple studies. Embryos were exposed at 6 h post fertilization (hpf) to aqueous Aroclor 1254 for 96 h with or without renewal in addition to a PCB 126 positive control for cardiotoxicity. PCB concentrations were measured in both exposure solutions and tissue samples. Measured concentrations of Aroclor 1254 in test solutions ranged from 8.7% to 870% of nominal concentrations. Heart rate, pericardial edema, and neurological endpoints (eye tremors) were measured in 102 hpf larvae. Pericardial edema was not present in Aroclor 1254-treated zebrafish but was observed in those exposed to PCB-126. Concentration-dependent bradycardia was observed in zebrafish exposed to Aroclor 1254 and PCB-126. Similarly, a concentration-dependent increase in eye tremor behavior was observed in embryos exposed to Aroclor 1254. Data produced by this study demonstrate novel toxicological effects of Aroclor 1254 and highlight the importance of measuring PCBs in both exposure and receptor media.
Collapse
Affiliation(s)
- Corey S Green
- Eastern New Mexico University, Department of Biological Sciences, 1500 Ave. K, Portales, NM, 88130, USA.
| | | | - Jason T Magnuson
- U.S. Geological Survey, Columbia Environmental Research Center, Columbia, MO, 65201, USA.
| | - Rachel R Leads
- Michigan State University, Department of Fisheries and Wildlife, East Lansing, MI, 48825, USA.
| | | | - Michel Gielazyn
- National Oceanic and Atmospheric Administration, Assessment and Restoration Division, St. Petersburg, FL, 33701, USA.
| | - Lisa Rosman
- National Oceanic and Atmospheric Administration, Assessment and Restoration Division, New York, NY, 10278, USA.
| | - Daniel Schlenk
- University of California Riverside, Department of Environmental Science, Riverside, CA, 92521, USA.
| | - Aaron P Roberts
- University of North Texas, Department of Biological Sciences and Advanced Environmental Research Institute, Denton, TX, 76203, USA.
| |
Collapse
|
4
|
Duffy AS, Eyo UB. Microglia and Astrocytes in Postnatal Neural Circuit Formation. Glia 2025; 73:232-250. [PMID: 39568399 DOI: 10.1002/glia.24650] [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: 07/18/2024] [Revised: 11/08/2024] [Accepted: 11/08/2024] [Indexed: 11/22/2024]
Abstract
Over the past two decades, microglia and astrocytes have emerged as critical mediators of neural circuit formation. Particularly during the postnatal period, both glial subtypes play essential roles in orchestrating nervous system development through communication with neurons. These functions include regulating synapse elimination, modulating neuronal density and activity, mediating synaptogenesis, facilitating axon guidance and organization, and actively promoting neuronal survival. Despite the vital roles of both microglia and astrocytes in ensuring homeostatic brain development, the extent to which the postnatal functions of these cells are regulated by sex and the manner in which these glial cells communicate with one another to coordinate nervous system development remain less well understood. Here, we review the critical functions of both microglia and astrocytes independently and synergistically in mediating neural circuit formation, focusing our exploration on the postnatal period from birth to early adulthood.
Collapse
Affiliation(s)
- Abigayle S Duffy
- Brain Immunology and Glia Center, University of Virginia, Charlottesville, Virginia, USA
- Department of Neuroscience, University of Virginia, Charlottesville, Virginia, USA
- Neuroscience Graduate Program, University of Virginia, Charlottesville, Virginia, USA
| | - Ukpong B Eyo
- Brain Immunology and Glia Center, University of Virginia, Charlottesville, Virginia, USA
- Department of Neuroscience, University of Virginia, Charlottesville, Virginia, USA
- Neuroscience Graduate Program, University of Virginia, Charlottesville, Virginia, USA
| |
Collapse
|
5
|
Vigil JJ, Tiemeier E, Orfila JE, Chalmers NE, Chang VN, Mitchell D, Veitch I, Falk M, Dietz RM, Herson PS, Quillinan N. Endogenous recovery of hippocampal function following global cerebral ischemia in juvenile female mice is influenced by neuroinflammation and circulating sex hormones. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.28.635301. [PMID: 39975306 PMCID: PMC11838352 DOI: 10.1101/2025.01.28.635301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Cardiac arrest-induced global cerebral ischemia (GCI) in childhood often results in learning and memory deficits. We previously demonstrated in a murine cardiac arrest and cardiopulmonary resuscitation (CA/CPR) mouse model that a cellular mechanism of learning and memory, long-term potentiation (LTP), is acutely impaired in the hippocampus of juvenile males, correlating with deficits in memory tasks. However, little is known regarding plasticity impairments in juvenile females. We performed CA/CPR in juvenile (P21-25) female mice and used slice electrophysiology and hippocampal dependent behavior to assess hippocampal function. LTP was and contextual fear were impaired 7-days after GCI and endogenously recovered by 30-days. LTP remained impaired at 30 days in ovariectomized females, suggesting the surge in gonadal sex hormones during puberty mediates endogenous recovery. Unlike juvenile males, recovery of LTP in juvenile females was not associated with BDNF expression. NanoString transcriptional analysis revealed a potential role of neuroinflammatory processes, and specifically Cd68 pathways, in LTP impairment and hormone-dependent recovery. We were able to restore LTP in ovariectomized females with chronic and acute PPT administration, implicating estrogen receptor alpha in recovery mechanisms. This study supports a mechanism of endogenous LTP recovery after GCI in juvenile female mice which differs mechanistically from juvenile males and does not occur in adults of either sex.
Collapse
|
6
|
Wu G, Zhu T, Ma C, Xu L, Qian Z, Kong G, Cui H, Zhang T, Wang J, Tang Y. Association of abnormal cortical inhibition and clinical outcomes in patients at clinical high risk for psychosis. Clin Neurophysiol 2025; 169:65-73. [PMID: 39626344 DOI: 10.1016/j.clinph.2024.11.015] [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: 10/23/2023] [Revised: 05/27/2024] [Accepted: 11/21/2024] [Indexed: 12/18/2024]
Abstract
OBJECTIVE Cortical inhibition (CI) can be in-vivo measured using transcranial magnetic stimulation (TMS), and patients with schizophrenia had abnormal CI. However, whether the abnormal CI occur early in patients with clinical high risk for psychosis (CHR) or could predict their clinical outcomes remains less known. METHODS We measured short-interval cortical inhibition (SICI), cortical silent period (CSP), and intra-cortical facilitation (ICF) over the motor cortex and neurocognitive performances in 55 CHR, 35 first-episode schizophrenia (FES), and 35 healthy controls (HC). We divided CHR patients into CHR converters (CHR-C) and CHR non-converters (CHR-NC) according to their clinical outcomes within the two-year follow-up. RESULTS CSP was longer in CHR-C (P = 0.033) and FES (P = 0.047) than in HC, while CSP was comparable between CHR-NC and HC. In CHR, CSP was negatively related to their performances in symbol coding and maze tasks. There was no significant between-group difference for either SICI or ICF. CONCLUSIONS Our findings suggested GABAB-mediated CSP was prolonged in CHR, who later converted into schizophrenia, and was associated with poor neurocognitive functions. SIGNIFICANCE CSP is prolonged before the onset of psychosis, particularly in CHR-C patients, suggesting that CSP could be a potential biomarker for predicting transition to schizophrenia.
Collapse
Affiliation(s)
- Guanfu Wu
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tianyuan Zhu
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chunyan Ma
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lihua Xu
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhenying Qian
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Gai Kong
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Huiru Cui
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tianhong Zhang
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jijun Wang
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Brain Science and Technology Research Center, Shanghai Jiao Tong University, Shanghai, China; CAS Center for Excellence in Brain Science and Intelligence Technology (CEBSIT), Chinese Academy of Science, Shanghai, China.
| | - Yingying Tang
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
7
|
S S, L.S. D, Rajendran P, N H, Singh S A. Exploring the potential of probiotics in Alzheimer's disease and gut dysbiosis. IBRO Neurosci Rep 2024; 17:441-455. [PMID: 39629018 PMCID: PMC11612366 DOI: 10.1016/j.ibneur.2024.11.004] [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: 09/15/2024] [Revised: 11/04/2024] [Accepted: 11/05/2024] [Indexed: 12/06/2024] Open
Abstract
Alzheimer's disease is a fatal neurodegenerative disorder that causes memory loss and cognitive decline in older people. There is increasing evidence suggesting that gut microbiota alteration is a cause of Alzheimer's disease pathogenesis. This review explores the link between gut dysbiosis and the development of Alzheimer's disease contributing to neuroinflammation, amyloid β accumulation, and cognitive decline. We examine the recent studies that illustrate the gut-brain axis (GBA) as a bidirectional communication between the gut and brain and how its alteration can influence neurological health. Furthermore, we discuss the potential of probiotic supplementation as a management approach to restore gut microbiota balance, and ultimately improve cognitive function in AD patients. Based on current research findings, this review aims to provide insights into the promising role of probiotics in Alzheimer's disease management and the need for further investigation into microbiota-targeted interventions.
Collapse
Affiliation(s)
- Sowmiya S
- Department of Pharmacology, Dr M.G.R. Educational and Research Institute, Poonamalle High Road, Velappanchavadi, Chennai, Tamil Nadu 600 077, India
| | - Dhivya L.S.
- Department of Pharmaceutical Chemistry, Dr M.G.R. Educational and Research Institute, Poonamalle High Road, Velappanchavadi, Chennai, Tamil Nadu 600 077, India
| | - Praveen Rajendran
- Department of Pharmacology, Dr M.G.R. Educational and Research Institute, Poonamalle High Road, Velappanchavadi, Chennai, Tamil Nadu 600 077, India
| | - Harikrishnan N
- Department of pharmaceutical analysis, Dr M.G.R. Educational and Research Institute, Poonamalle High Road, Velappanchavadi, Chennai, Tamil Nadu 600 077, India
| | - Ankul Singh S
- Department of Pharmacology, Dr M.G.R. Educational and Research Institute, Poonamalle High Road, Velappanchavadi, Chennai, Tamil Nadu 600 077, India
| |
Collapse
|
8
|
McKeon SD, Perica MI, Calabro FJ, Foran W, Hetherington H, Moon CH, Luna B. Prefrontal excitation/inhibition balance supports adolescent enhancements in circuit signal to noise ratio. Prog Neurobiol 2024; 243:102695. [PMID: 39622336 DOI: 10.1016/j.pneurobio.2024.102695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 11/18/2024] [Accepted: 11/29/2024] [Indexed: 12/06/2024]
Abstract
The development and refinement of neuronal circuitry allow for stabilized and efficient neural recruitment, supporting adult-like behavioral performance. During adolescence, the maturation of PFC is proposed to be a critical period (CP) for executive function, driven by a break in balance between glutamatergic excitation and GABAergic inhibition (E/I) neurotransmission. During CPs, cortical circuitry fine-tunes to improve information processing and reliable responses to stimuli, shifting from spontaneous to evoked activity, enhancing the SNR, and promoting neural synchronization. Harnessing 7 T MR spectroscopy and EEG in a longitudinal cohort (N = 164, ages 10-32 years, 283 neuroimaging sessions), we outline associations between age-related changes in glutamate and GABA neurotransmitters and EEG measures of cortical SNR. We find developmental decreases in spontaneous activity and increases in cortical SNR during our auditory steady state task using 40 Hz stimuli. Decreases in spontaneous activity were associated with glutamate levels in DLPFC, while increases in cortical SNR were associated with more balanced Glu and GABA levels. These changes were associated with improvements in working memory performance. This study provides evidence of CP plasticity in the human PFC during adolescence, leading to stabilized circuitry that allows for the optimal recruitment and integration of multisensory input, resulting in improved executive function.
Collapse
Affiliation(s)
- Shane D McKeon
- Department of Bioengineering, University of Pittsburgh, PA, USA; The Center for the Neural Basis of Cognition, University of Pittsburgh, PA, USA.
| | - Maria I Perica
- The Center for the Neural Basis of Cognition, University of Pittsburgh, PA, USA; Department of Psychology, University of Pittsburgh, PA, USA
| | - Finnegan J Calabro
- Department of Bioengineering, University of Pittsburgh, PA, USA; The Center for the Neural Basis of Cognition, University of Pittsburgh, PA, USA; Department of Psychiatry, University of Pittsburgh, PA, USA
| | - Will Foran
- Department of Psychiatry, University of Pittsburgh, PA, USA
| | - Hoby Hetherington
- Resonance Research Incorporated, Billerica, MA, USA; Department of Radiology, University of Missouri, Columbia, MO, USA
| | - Chan-Hong Moon
- Department of Radiology, University of Pittsburgh, PA, USA
| | - Beatriz Luna
- The Center for the Neural Basis of Cognition, University of Pittsburgh, PA, USA; Department of Psychiatry, University of Pittsburgh, PA, USA.
| |
Collapse
|
9
|
Ji X, Liu S, Li S, Li X, Luo A, Zhang X, Zhao Y. GABA in early brain development: A dual role review. Int J Dev Neurosci 2024; 84:843-856. [PMID: 39503050 DOI: 10.1002/jdn.10387] [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: 04/19/2024] [Revised: 09/27/2024] [Accepted: 09/30/2024] [Indexed: 01/03/2025] Open
Abstract
This comprehensive review examines the multifaceted roles of gamma-aminobutyric acid (GABA) in early brain development. GABA, traditionally recognized for its inhibitory functions in the mature brain, also exhibits excitatory effects during early neural development. This article explores the mechanisms behind GABA's dual roles, detailing its impact on the properties of the immature brain, the mechanisms of GABA-mediated excitation, the role of GABA-mediated presynaptic inhibition, the trophic actions of GABA during early development, GABA regulation of neurite growth and GABA-mediated cell differentiation in the immature brain. Emphasizing recent research findings, the review highlights the significance of GABAergic signalling in shaping the developing brain and its potential implications for understanding neurodevelopmental disorders.
Collapse
Affiliation(s)
- Xiaoyu Ji
- Department of Anesthesiology, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shuzhen Liu
- Department of Anesthesiology, Tai'an Central Hospital, Tai'an, China
| | - Shiyong Li
- Department of Anesthesiology, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xing Li
- Department of Anesthesiology, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ailin Luo
- Department of Anesthesiology, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xue Zhang
- Department of Anesthesiology, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yilin Zhao
- Department of Anesthesiology, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
10
|
Köksal-Ersöz E, Benquet P, Wendling F. Expansion of epileptogenic networks via neuroplasticity in neural mass models. PLoS Comput Biol 2024; 20:e1012666. [PMID: 39625956 PMCID: PMC11642990 DOI: 10.1371/journal.pcbi.1012666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 12/13/2024] [Accepted: 11/21/2024] [Indexed: 12/14/2024] Open
Abstract
Neuroplasticity refers to functional and structural changes in brain regions in response to healthy and pathological activity. Activity dependent plasticity induced by epileptic activity can involve healthy brain regions into the epileptogenic network by perturbing their excitation/inhibition balance. In this article, we present a new neural mass model, which accounts for neuroplasticity, for investigating the possible mechanisms underlying the epileptogenic network expansion. Our multiple-timescale model is inspired by physiological calcium-mediated synaptic plasticity and pathological extrasynaptic N-methyl-D-aspartate (NMDA) dependent plasticity dynamics. The model highlights that synaptic plasticity at excitatory connections and structural changes in the inhibitory system can transform a healthy region into a secondary epileptic focus under recurrent seizures and interictal activity occurring in the primary focus. Our results suggest that the latent period of this transformation can provide a window of opportunity to prevent the expansion of epileptogenic networks, formation of an epileptic focus, or other comorbidities associated with epileptic activity.
Collapse
|
11
|
Sambo D, Kinstler E, Lin Y, Goldman D. Differential effects of prenatal alcohol exposure on brain growth reveals early upregulation of cell cycle and apoptosis and delayed downregulation of metabolism in affected offspring. PLoS One 2024; 19:e0311683. [PMID: 39602444 PMCID: PMC11602053 DOI: 10.1371/journal.pone.0311683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Accepted: 09/06/2024] [Indexed: 11/29/2024] Open
Abstract
Fetal Alcohol Spectrum Disorder (FASD) encompasses the deleterious consequences of Prenatal Alcohol Exposure (PAE), including developmental delay, microcephaly, dysmorphologies, and cognitive and behavioral issues. The dose and timing of alcohol exposure, maternal and environmental factors, and genetics all impact FASD outcomes, but differential susceptibility and resiliency to PAE remains poorly understood. In this study, we examined the differential effects of PAE during early mouse development on brain growth and gene expression. Brains were weighed and collected either 24 hours or five days after treatment. We then performed transcriptomics to determine whether offspring differentially affected by PAE, by brain weight, also differ in gene expression, despite having the same genetic background, alcohol exposure, and maternal factors. We found within litter variation in brain weights after PAE, and classified offspring as having normal, middle, and low-weight brains relative to saline-treated controls. The normal-weight brains showed no significant differences in gene expression, suggesting these offspring were both phenotypically and transcriptionally unaffected by PAE. While both middle- and low-weight brains showed changes in gene expression, the middle-weight brains showed the most robust transcriptome differences. Twenty-four hours after PAE, we saw an upregulation of cell cycle and apoptosis in affected offspring, whereas at roughly a week later, we saw a downregulation of metabolic processes. Overall, these findings highlight variability in response to PAE and demonstrate the molecular processes involved in offspring phenotypically affected by alcohol.
Collapse
Affiliation(s)
- Danielle Sambo
- Laboratory of Neurogenetics, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Rockville, Maryland, United States of America
| | - Ethan Kinstler
- Laboratory of Neurogenetics, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Rockville, Maryland, United States of America
| | - Yuhong Lin
- Laboratory of Liver Diseases, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Rockville, Maryland, United States of America
| | - David Goldman
- Laboratory of Neurogenetics, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Rockville, Maryland, United States of America
| |
Collapse
|
12
|
Fedorov NS, Sibgatullina GV, Malomouzh AI. Impairment of Skeletal Muscle Contraction by Inhibitors of GABA Transporters. Int J Mol Sci 2024; 25:12510. [PMID: 39684222 DOI: 10.3390/ijms252312510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 11/19/2024] [Accepted: 11/19/2024] [Indexed: 12/18/2024] Open
Abstract
γ-Aminobutyric acid (GABA) has a significant impact on the functioning of not only the central but also the peripheral part of the nervous system. Recently, various elements of the GABAergic signaling system have been discovered in the area of the neuromuscular junction of mammals. At the same time, the functional activity of membrane-bound GABA transporters (GATs) and their role in neuromuscular transmission have not been identified. In the present study, performed on a neuromuscular preparation of the mouse diaphragm, the effect of GABA transporter inhibitors (nipecotic acid and β-alanine) on the force of muscle contraction was assessed. It was found that in the presence of both compounds in the bathing solution, the force of contractions caused by stimulation of the motor nerve dropped by 30-50%. However, when the muscle was stimulated directly, no effect of GABA transporter inhibitors on the contractile force was observed. The depressant effect of β-alanine induced by nerve stimulation was completely abolished by the GABAB receptor blocker CGP 55845. GABA transporters were detected at the neuromuscular junction using immunohistochemistry. Thus, our results indicate that GABA transporters are localized in the area of the neuromuscular junction, and their activity affects the muscle contraction force. This influence is most likely due to the removal of GABA released during nerve stimulation and activating GABA receptors, which leads to a decrease in the contraction force of the striated muscles.
Collapse
Affiliation(s)
- Nikita S Fedorov
- Kazan Institute of Biochemistry and Biophysics, Federal Research Center "Kazan Scientific Center of RAS", 2/31 Lobachevsky Street, P.O. Box 30, Kazan 420111, Russia
| | - Guzel V Sibgatullina
- Kazan Institute of Biochemistry and Biophysics, Federal Research Center "Kazan Scientific Center of RAS", 2/31 Lobachevsky Street, P.O. Box 30, Kazan 420111, Russia
| | - Artem I Malomouzh
- Kazan Institute of Biochemistry and Biophysics, Federal Research Center "Kazan Scientific Center of RAS", 2/31 Lobachevsky Street, P.O. Box 30, Kazan 420111, Russia
- Department of Radiophotonics and Microwave Technologies, Kazan National Research Technical University, 10 K. Marx St., Kazan 420111, Russia
| |
Collapse
|
13
|
de Groot ER, Dudink J, Austin T. Sleep as a driver of pre- and postnatal brain development. Pediatr Res 2024; 96:1503-1509. [PMID: 38956219 PMCID: PMC11624135 DOI: 10.1038/s41390-024-03371-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 06/11/2024] [Accepted: 06/17/2024] [Indexed: 07/04/2024]
Abstract
In 1966, Howard Roffwarg proposed the ontogenic sleep hypothesis, relating neural plasticity and development to rapid eye movement (REM) sleep, a hypothesis that current fetal and neonatal sleep research is still exploring. Recently, technological advances have enabled researchers to automatically quantify neonatal sleep architecture, which has caused a resurgence of research in this field as attempts are made to further elucidate the important role of sleep in pre- and postnatal brain development. This article will review our current understanding of the role of sleep as a driver of brain development and identify possible areas for future research. IMPACT: The evidence to date suggests that Roffwarg's ontogenesis hypothesis of sleep and brain development is correct. A better understanding of the relationship between sleep and the development of functional connectivity is needed. Reliable, non-invasive tools to assess sleep in the NICU and at home need to be tested in a real-world environment and the best way to promote healthy sleep needs to be understood before clinical trials promoting and optimizing sleep quality in neonates could be undertaken.
Collapse
Affiliation(s)
- Eline R de Groot
- Department of Neonatology, Wilhelmina Children's Hospital, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Jeroen Dudink
- Department of Neonatology, Wilhelmina Children's Hospital, University Medical Centre Utrecht, Utrecht, The Netherlands
- Brain Centre Rudolf Magnus, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Topun Austin
- NeoLab, Evelyn Perinatal Imaging Centre, The Rosie Hospital, Cambridge University Hospitals, Cambridge, UK.
| |
Collapse
|
14
|
Kong S, Qian X, Cai J, Wang J, Wang K. Percutaneous plasma disc decompression through a lower surgical approach for the treatment of cervicogenic headache in patients with cervical spondylotic radiculopathy: A retrospective cohort study. Biomed Rep 2024; 21:152. [PMID: 39247422 PMCID: PMC11375622 DOI: 10.3892/br.2024.1840] [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: 01/18/2024] [Accepted: 07/27/2024] [Indexed: 09/10/2024] Open
Abstract
Cervical spondylotic radiculopathy (CSR) is the most common type of cervical spondylosis, frequently accompanied by cervicogenic headache (CEH). Percutaneous plasma disc decompression (PPDD) and pulsed radiofrequency (PRF) are minimally invasive techniques targeting cervical intervertebral discs or cervical nerves, and have been proven to be effective methods for treatment of CSR and CEH. The present study aimed to evaluate clinical efficacy and practicality of percutaneous plasma disc decompression (PPDD) via a lower surgical approach for the treatment of cervicogenic headache (CEH) and upper extremity radicular pain by analyzing clinical outcomes of patients with cervical spondylotic radiculopathy (CSR) undergoing PPDD and pulsed radiofrequency (PRF). Clinical data of patients with CSR who received PPDD (n=79) or PRF (n=92) at Shanghai Traditional Chinese Medicine Hospital (Shanghai, China) and Jiashan County People's Hospital (Jiaxing, China) from January 2022 to December 2022 were retrospectively collected and analyzed. The surgical site and procedure, bleeding volume, preoperative analgesic use and upper extremity symptoms, history of nerve block treatment and duration of disease were recorded, as well as relevant postoperative complications (infection, hematoma, nerve injury). The therapeutic effects [NRS (numeric rating scale) and NDI (neck disability index) score, and CEH remission rate at 1, 3 and 6 months after treatment] of both surgical methods were investigated using the telephone follow-up. CEH remission rates at 1, 3 and 6 months after surgery in the PPDD group were significantly higher than in the PRF group (78.8 vs. 43.5, P=0.016; 84.8 vs. 34.8, P=0.003 and 75.8 vs. 26.1%, P=0.005, respectively). The PPDD group showed higher NRS scores than the PRF group at 1 month after surgery (3 vs. 2, P<0.0001) and lower NRS scores than the PRF group at 6 months after surgery (2 vs. 3, P<0.0001). NDI scores in the PPDD group were significantly lower than those in the PRF group at 1, 3 and 6 months after surgery (15.49 vs. 20.05, P=0.002; 16.06 vs. 20.10, P=0.003 and 9.90 vs. 13.80, P=0.001, respectively). There was no significant difference in postoperative complication rate between the two groups (P>0.999). PPDD could significantly relieve CEH symptoms and upper extremity radicular pain in patients with CSR treated via a lower surgical approach and PPDD was more effective than PRF for long-term CEH remission and pain alleviation.
Collapse
Affiliation(s)
- Shuyi Kong
- Department of Pain Management, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200071, P.R. China
| | - Xuantao Qian
- Shu Guang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P.R. China
| | - Jianfeng Cai
- Department of Orthopedics, Jiashan County Hospital of Traditional Chinese Medicine, Jiaxing, Zhejiang 314100, P.R. China
| | - Jing Wang
- Department of Emergency, The Second Affiliated Hospital of Shandong First Medical University, Taian, Shandong 271000, P.R. China
| | - Kaiqiang Wang
- Department of Pain Management, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200071, P.R. China
| |
Collapse
|
15
|
Cheng Y, Zhang R, Li X, Zhou X, Chen M, Liu A. The Dopamine Transporter Is a New Target for Ischemic Stroke. CNS Neurosci Ther 2024; 30:e70092. [PMID: 39467829 PMCID: PMC11518691 DOI: 10.1111/cns.70092] [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: 07/11/2024] [Revised: 09/11/2024] [Accepted: 10/10/2024] [Indexed: 10/30/2024] Open
Abstract
AIMS Dopamine transporter (DAT) can regulate DA homeostasis and has been implicated in many nervous system diseases. Whether DAT is involved in the protection against ischemic stroke is unclear. METHODS In vivo microdialysis measurements of DA were recorded in the ischemic penumbral area of mice with middle cerebral artery occlusion (MCAO). DAT coding gene, Slc6a3 mutation, and DAT overexpression animals were performed MCAO. Madopar (compound formulation of levodopa) and nomifensine (DA reuptake inhibitor) were administered in MCAO animals. Brain slices were prepared in Slc6a3 mutation or wild-type (WT) animals with MCAO to record miniature excitatory postsynaptic currents (mEPSCs) and miniature inhibitory postsynaptic currents (mIPSCs). The effects of DA and its dopamine-1 receptor (D1R) antagonists (SCH-23390) on mEPSCs, mIPSCs, and neurons protection were recorded. RESULTS MCAO caused a prominent increase in DA. Slc6a3 mutation significantly attenuated the ischemic injury, whereas DAT overexpression aggravated this injury. Both nomifensine and madopar protected against brain injury. Slc6a3 mutation and DA restored the disturbance of mEPSCs and mIPSC, and protected against neuron death, which was abolished by SCH-23390. CONCLUSION DAT inhibition might be explored as a strategy for ischemic stroke prevention. DA and D1R involve in the restoration of synaptic dysfunction and neuron protection.
Collapse
Affiliation(s)
- Yan‐Qiong Cheng
- Department of Pharmacy Research, Yueyang Hospital of Integrated Traditional Chinese and Western MedicineShanghai University of Traditional Chinese MedicineShanghaiChina
| | - Ruo‐Xi Zhang
- Department of Pharmacy Research, Yueyang Hospital of Integrated Traditional Chinese and Western MedicineShanghai University of Traditional Chinese MedicineShanghaiChina
| | - Xing‐Yuan Li
- Department of Pharmacy Research, Yueyang Hospital of Integrated Traditional Chinese and Western MedicineShanghai University of Traditional Chinese MedicineShanghaiChina
| | - Xiao‐Ting Zhou
- Department of Pharmacy Research, Yueyang Hospital of Integrated Traditional Chinese and Western MedicineShanghai University of Traditional Chinese MedicineShanghaiChina
| | - Ming Chen
- MOE Frontier Center for Brain Science, Institutes of Brain Science, State Key Laboratory of Medical NeurobiologyFudan UniversityShanghaiChina
| | - Ai‐Jun Liu
- Department of Pharmacy Research, Yueyang Hospital of Integrated Traditional Chinese and Western MedicineShanghai University of Traditional Chinese MedicineShanghaiChina
| |
Collapse
|
16
|
Spathopoulou A, Sauerwein GA, Marteau V, Podlesnic M, Lindlbauer T, Kipura T, Hotze M, Gabassi E, Kruszewski K, Koskuvi M, Réthelyi JM, Apáti Á, Conti L, Ku M, Koal T, Müller U, Talmazan RA, Ojansuu I, Vaurio O, Lähteenvuo M, Lehtonen Š, Mertens J, Kwiatkowski M, Günther K, Tiihonen J, Koistinaho J, Trajanoski Z, Edenhofer F. Integrative metabolomics-genomics analysis identifies key networks in a stem cell-based model of schizophrenia. Mol Psychiatry 2024; 29:3128-3140. [PMID: 38684795 PMCID: PMC11449784 DOI: 10.1038/s41380-024-02568-8] [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: 10/17/2022] [Revised: 04/12/2024] [Accepted: 04/17/2024] [Indexed: 05/02/2024]
Abstract
Schizophrenia (SCZ) is a neuropsychiatric disorder, caused by a combination of genetic and environmental factors. The etiology behind the disorder remains elusive although it is hypothesized to be associated with the aberrant response to neurotransmitters, such as dopamine and glutamate. Therefore, investigating the link between dysregulated metabolites and distorted neurodevelopment holds promise to offer valuable insights into the underlying mechanism of this complex disorder. In this study, we aimed to explore a presumed correlation between the transcriptome and the metabolome in a SCZ model based on patient-derived induced pluripotent stem cells (iPSCs). For this, iPSCs were differentiated towards cortical neurons and samples were collected longitudinally at various developmental stages, reflecting neuroepithelial-like cells, radial glia, young and mature neurons. The samples were analyzed by both RNA-sequencing and targeted metabolomics and the two modalities were used to construct integrative networks in silico. This multi-omics analysis revealed significant perturbations in the polyamine and gamma-aminobutyric acid (GABA) biosynthetic pathways during rosette maturation in SCZ lines. We particularly observed the downregulation of the glutamate decarboxylase encoding genes GAD1 and GAD2, as well as their protein product GAD65/67 and their biochemical product GABA in SCZ samples. Inhibition of ornithine decarboxylase resulted in further decrease of GABA levels suggesting a compensatory activation of the ornithine/putrescine pathway as an alternative route for GABA production. These findings indicate an imbalance of cortical excitatory/inhibitory dynamics occurring during early neurodevelopmental stages in SCZ. Our study supports the hypothesis of disruption of inhibitory circuits to be causative for SCZ and establishes a novel in silico approach that enables for integrative correlation of metabolic and transcriptomic data of psychiatric disease models.
Collapse
Affiliation(s)
- Angeliki Spathopoulou
- Institute of Molecular Biology & CMBI, Department of Genomics, Stem Cell & Regenerative Medicine, University of Innsbruck, Innsbruck, Austria
| | - Gabriella A Sauerwein
- Institute of Molecular Biology & CMBI, Department of Genomics, Stem Cell & Regenerative Medicine, University of Innsbruck, Innsbruck, Austria
| | - Valentin Marteau
- Institute of Molecular Biology & CMBI, Department of Genomics, Stem Cell & Regenerative Medicine, University of Innsbruck, Innsbruck, Austria
| | - Martina Podlesnic
- Institute of Molecular Biology & CMBI, Department of Genomics, Stem Cell & Regenerative Medicine, University of Innsbruck, Innsbruck, Austria
| | - Theresa Lindlbauer
- Institute of Molecular Biology & CMBI, Department of Genomics, Stem Cell & Regenerative Medicine, University of Innsbruck, Innsbruck, Austria
| | - Tobias Kipura
- Institute of Biochemistry and Center for Molecular Biosciences Innsbruck, University of Innsbruck, Innsbruck, Austria
| | - Madlen Hotze
- Institute of Biochemistry and Center for Molecular Biosciences Innsbruck, University of Innsbruck, Innsbruck, Austria
| | - Elisa Gabassi
- Institute of Molecular Biology & CMBI, Department of Genomics, Stem Cell & Regenerative Medicine, University of Innsbruck, Innsbruck, Austria
| | - Katharina Kruszewski
- Institute of Molecular Biology & CMBI, Department of Genomics, Stem Cell & Regenerative Medicine, University of Innsbruck, Innsbruck, Austria
| | - Marja Koskuvi
- Neuroscience Center, University of Helsinki, Helsinki, Finland
| | - János M Réthelyi
- Department of Psychiatry and Psychotherapy, Semmelweis University, Budapest, Hungary
| | - Ágota Apáti
- HUN-REN RCNS, Institute of Molecular Life Sciences, Budapest, Hungary
| | - Luciano Conti
- Department of Cellular, Computational and Integrative Biology-CIBIO, University of Trento, Trento, Italy
| | - Manching Ku
- Department of Pediatrics and Adolescent Medicine, Division of Pediatric Hematology and Oncology, Faculty of Medicine, Medical Center - University of Freiburg, Freiburg, Germany
| | | | - Udo Müller
- biocrates life sciences AG, Innsbruck, Austria
| | | | - Ilkka Ojansuu
- Department of Forensic Psychiatry, University of Kuopio, Niuvanniemi Hospital, Kuopio, Finland
| | - Olli Vaurio
- Department of Forensic Psychiatry, University of Kuopio, Niuvanniemi Hospital, Kuopio, Finland
| | - Markku Lähteenvuo
- Department of Forensic Psychiatry, University of Kuopio, Niuvanniemi Hospital, Kuopio, Finland
| | - Šárka Lehtonen
- Neuroscience Center, University of Helsinki, Helsinki, Finland
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Jerome Mertens
- Institute of Molecular Biology & CMBI, Department of Genomics, Stem Cell & Regenerative Medicine, University of Innsbruck, Innsbruck, Austria
- Department of Neurosciences, Sanford Consortium for Regenerative Medicine, University of California San Diego, San Diego, USA
| | - Marcel Kwiatkowski
- Institute of Biochemistry and Center for Molecular Biosciences Innsbruck, University of Innsbruck, Innsbruck, Austria
| | - Katharina Günther
- Institute of Molecular Biology & CMBI, Department of Genomics, Stem Cell & Regenerative Medicine, University of Innsbruck, Innsbruck, Austria
| | - Jari Tiihonen
- Department of Forensic Psychiatry, University of Kuopio, Niuvanniemi Hospital, Kuopio, Finland
- Department of Clinical Neuroscience, Karolinska Institutet, and Center for Psychiatry Research, Stockholm City Council, Stockholm, Sweden
| | - Jari Koistinaho
- Institute of Life Science, University of Helsinki, FI-00014, Helsinki, Finland
- Drug Research Program, Division of Pharmacology and Pharmacotherapy, University of Helsinki, Helsinki, Finland
| | - Zlatko Trajanoski
- Institute of Bioinformatics, Biocenter, Medical University Innsbruck, Innsbruck, Austria
| | - Frank Edenhofer
- Institute of Molecular Biology & CMBI, Department of Genomics, Stem Cell & Regenerative Medicine, University of Innsbruck, Innsbruck, Austria.
| |
Collapse
|
17
|
Galindo-Torres P, Rosas C, Ramos-Rodríguez S, Galindo-Sánchez CE. Chronic thermal stress on Octopus maya embryos down-regulates epigenome-related genes and those involved in the nervous system development and morphogenesis. COMPARATIVE BIOCHEMISTRY AND PHYSIOLOGY. PART D, GENOMICS & PROTEOMICS 2024; 52:101332. [PMID: 39366120 DOI: 10.1016/j.cbd.2024.101332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 09/20/2024] [Accepted: 09/22/2024] [Indexed: 10/06/2024]
Abstract
Red Octopus maya is strongly influenced by temperature. Recent studies have reported negative reproduction effects on males and females when exposed to temperatures higher than 27 °C. Embryos under thermal stress show morphological and physiological alterations; similar phenotypes have been reported in embryos from stressed females, evidencing transgenerational consequences. Transcriptomic profiles were characterized along embryo development during normal-under thermal stress and epigenetic alterations through DNA methylation and damage quantification. Total RNA in organogenesis, activation, and growth stages in control and thermal stress were sequenced with Illumina RNA-Seq. Similarly, total DNA was used for DNA methylation and damage quantification between temperatures and embryo stages. Differential gene expression analyses showed that embryos express genes associated with oxygen transport, morphogenesis, nervous system, neuroendocrine cell differentiation, spermatogenesis, and male sex differentiation. Conversely, embryos turn off genes involved mainly in nervous system development, morphogenesis, and gene expression regulation when exposed to thermal stress - consistent with O. maya embryo phenotypes showing abnormal arms, eyes, and body development. No significant differences were observed in quantifying DNA methylation between temperatures but they were for DNA damage quantification. Epigenetic alterations are hypothesized to occur since several genes found downregulated belong to the epigenetic machinery but at histone tail level.
Collapse
Affiliation(s)
- Pavel Galindo-Torres
- Centro de Investigacion Cientifica y de Educacion Superior de Ensenada (CICESE), Carretera Tijuana-Ensenada No. 3918, Zona Playitas, Ensenada, Baja California CP 22860, Mexico.
| | - Carlos Rosas
- Unidad Multidisciplinaria de Docencia e Investigacion (UMDI), Facultad de Ciencias, Universidad Nacional Autonoma de Mexico (UNAM), Puerto DE Abrigo s/n, Sisal, Hunucma, Yucatan CP97355, Mexico.
| | - Sadot Ramos-Rodríguez
- Centro de Investigacion Cientifica y de Educacion Superior de Ensenada (CICESE), Carretera Tijuana-Ensenada No. 3918, Zona Playitas, Ensenada, Baja California CP 22860, Mexico.
| | - Clara E Galindo-Sánchez
- Centro de Investigacion Cientifica y de Educacion Superior de Ensenada (CICESE), Carretera Tijuana-Ensenada No. 3918, Zona Playitas, Ensenada, Baja California CP 22860, Mexico.
| |
Collapse
|
18
|
Liu Y, Chen Y, Duffy CR, VanLeuven AJ, Byers JB, Schriever HC, Ball RE, Carpenter JM, Gunderson CE, Filipov NM, Ma P, Kner PA, Lauderdale JD. Decreased GABA levels during development result in increased connectivity in the larval zebrafish tectum. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.11.612511. [PMID: 39314470 PMCID: PMC11419034 DOI: 10.1101/2024.09.11.612511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
γ-aminobutyric acid (GABA) is an abundant neurotransmitter that plays multiple roles in the vertebrate central nervous system (CNS). In the early developing CNS, GABAergic signaling acts to depolarize cells. It mediates several aspects of neural development, including cell proliferation, neuronal migration, neurite growth, and synapse formation, as well as the development of critical periods. Later in CNS development, GABAergic signaling acts in an inhibitory manner when it becomes the predominant inhibitory neurotransmitter in the brain. This behavior switch occurs due to changes in chloride/cation transporter expression. Abnormalities of GABAergic signaling appear to underlie several human neurological conditions, including seizure disorders. However, the impact of reduced GABAergic signaling on brain development has been challenging to study in mammals. Here we take advantage of zebrafish and light sheet imaging to assess the impact of reduced GABAergic signaling on the functional circuitry in the larval zebrafish optic tectum. Zebrafish have three gad genes: two gad1 paralogs known as gad1a and gad1b, and gad2. The gad1b and gad2 genes are expressed in the developing optic tectum. Null mutations in gad1b significantly reduce GABA levels in the brain and increase electrophysiological activity in the optic tectum. Fast light sheet imaging of genetically encoded calcium indicator (GCaMP)-expressing gab1b null larval zebrafish revealed patterns of neural activity that were different than either gad1b-normal larvae or gad1b-normal larvae acutely exposed to pentylenetetrazole (PTZ). These results demonstrate that reduced GABAergic signaling during development increases functional connectivity and concomitantly hyper-synchronization of neuronal networks.
Collapse
Affiliation(s)
- Yang Liu
- School of Electrical and Computer Engineering, The University of Georgia, Athens, GA 30602, USA
| | - Yongkai Chen
- Department of Statistics, The University of Georgia, Athens, GA 30602, USA
| | - Carly R Duffy
- Department of Cellular Biology, The University of Georgia, Athens, GA 30602, USA
| | - Ariel J VanLeuven
- Department of Cellular Biology, The University of Georgia, Athens, GA 30602, USA
| | - John Branson Byers
- Department of Cellular Biology, The University of Georgia, Athens, GA 30602, USA
| | - Hannah C Schriever
- Department of Cellular Biology, The University of Georgia, Athens, GA 30602, USA
| | - Rebecca E Ball
- Department of Cellular Biology, The University of Georgia, Athens, GA 30602, USA
| | - Jessica M Carpenter
- Department of Physiology and Pharmacology, The University of Georgia, College of Veterinary Medicine, Athens, GA, 30602, USA
- Neuroscience Division of the Biomedical and Translational Sciences Institute, The University of Georgia, Athens, GA 30602, USA
| | - Chelsea E Gunderson
- Department of Cellular Biology, The University of Georgia, Athens, GA 30602, USA
| | - Nikolay M Filipov
- Department of Physiology and Pharmacology, The University of Georgia, College of Veterinary Medicine, Athens, GA, 30602, USA
| | - Ping Ma
- Department of Statistics, The University of Georgia, Athens, GA 30602, USA
| | - Peter A Kner
- School of Electrical and Computer Engineering, The University of Georgia, Athens, GA 30602, USA
| | - James D Lauderdale
- Department of Cellular Biology, The University of Georgia, Athens, GA 30602, USA
- Neuroscience Division of the Biomedical and Translational Sciences Institute, The University of Georgia, Athens, GA 30602, USA
| |
Collapse
|
19
|
Looschen K, Khatri SN, Maulik M, Salisbury C, Carman AF, Corriveau K, Smith C, Manetti D, Romanelli MN, Arias HR, Gipson CD, Mitra S. Novel psychoplastogen DM506 reduces cue-induced heroin-seeking and inhibits tonic GABA currents in the Prelimbic Cortex. Neurochem Int 2024; 178:105785. [PMID: 38838988 DOI: 10.1016/j.neuint.2024.105785] [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: 04/04/2024] [Revised: 05/10/2024] [Accepted: 06/01/2024] [Indexed: 06/07/2024]
Abstract
Opioid use disorder is a major public health crisis that is manifested by persistent drug-seeking behavior and high relapse frequency. Most of the available treatments rely on targeting opioid receptors using small molecules that do not provide sustained symptom alleviation. Psychoplastogens are a novel class of non-opioid drugs that produce rapid and sustained effects on neuronal plasticity, intended to produce therapeutic benefits. Ibogalogs are synthetic derivatives of iboga alkaloids that lack hallucinogenic or adverse side effects. In the current study, we examine the therapeutic potential of DM506, a novel ibogalog lacking any cardiotoxic or hallucinogenic effects, in cue-induced seeking behavior following heroin self-administration. At a single systemic dose of 40 mg/kg, DM506 significantly decreased cue-induced seeking in both male and female rats at abstinence day 1 (AD1) following heroin self-administration. Upon re-testing for cue-induced seeking at AD14, we found that males receiving DM506 continued to show decreased cue-induced seeking, an effect not observed in females. Since there is evidence of psychedelics influencing tonic GABA currents, and opioid and psychoplastogen-mediated neuroadaptations in the medial prefrontal cortex (PrL) underlying its functional effects, we performed patch-clamp recordings on PrL slices of drug-naïve rats with an acute application or chronic incubation with DM506. Tonic GABA current was decreased in slices incubated with DM506 for 2 h. qPCR analysis did not reveal any differences in the mRNA levels of GABAA receptor α and δ subunits at AD14 in heroin and saline self-administered animals that received vehicle or DM506 at AD1. Overall, our data indicate that DM506 attenuates cue-induced heroin seeking and inhibits tonic GABA current in the prelimbic cortex.
Collapse
Affiliation(s)
- Kassandra Looschen
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV, USA
| | - Shailesh Narayan Khatri
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, USA
| | - Malabika Maulik
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV, USA; Department of Biochemistry and Microbiology, Oklahoma State University Center for Health Sciences, Tulsa, OK, USA
| | - Colin Salisbury
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV, USA
| | - Alaina F Carman
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV, USA
| | - Katilyn Corriveau
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV, USA
| | - Colton Smith
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV, USA
| | - Dina Manetti
- Department of Neurosciences, Psychology, Drug Research and Child Health Section of Pharmaceutical and Nutraceutical Sciences, University of Florence, Italy
| | - Maria Novella Romanelli
- Department of Neurosciences, Psychology, Drug Research and Child Health Section of Pharmaceutical and Nutraceutical Sciences, University of Florence, Italy
| | - Hugo R Arias
- Department of Pharmacology and Physiology, Oklahoma State University Center for Health Sciences, Tulsa, USA; Department of Pharmacology and Physiology, Oklahoma State University Center for Health Sciences, Tahlequah, USA
| | - Cassandra D Gipson
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, USA
| | - Swarup Mitra
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV, USA; Department of Pharmacology and Physiology, Oklahoma State University Center for Health Sciences, Tulsa, USA.
| |
Collapse
|
20
|
Farzan M, Abedi B, Bhia I, Madanipour A, Farzan M, Bhia M, Aghaei A, Kheirollahi I, Motallebi M, Amini-Khoei H, Ertas YN. Pharmacological Activities and Molecular Mechanisms of Sinapic Acid in Neurological Disorders. ACS Chem Neurosci 2024; 15:2966-2981. [PMID: 39082749 DOI: 10.1021/acschemneuro.4c00349] [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: 08/10/2024] Open
Abstract
Sinapic acid (SA) is a phenylpropanoid derivative found in various natural sources that exhibits remarkable versatile properties, including antioxidant, anti-inflammatory, and metal-chelating capabilities, establishing itself as a promising candidate for the prevention and treatment of conditions affecting the central nervous system, such as Alzheimer's disease (AD), Parkinson's disease (PD), ischemic stroke, and other neurological disorders. These effects also include neuroprotection in epilepsy models, as evidenced by a reduction in seizure-like behavior, cell death in specific hippocampal regions, and lowered neuroinflammatory markers. In AD, SA treatment enhances memory, reverses cognitive deficits, and attenuates astrocyte activation. SA also has positive effects on cognition by improving memory and lowering oxidative stress. This is shown by lower levels of oxidative stress markers, higher levels of antioxidant enzyme activity, and better memory retention. Additionally, in ischemic stroke and PD models, SA provides microglial protection and exerts anti-inflammatory effects. This review emphasizes SA's multifaceted neuroprotective properties and its potential role in the prevention and treatment of various brain disorders. Despite the need for further research to fully understand its mechanisms of action and clinical applicability, SA stands out as a valuable bioactive compound in the ongoing quest to combat neurodegenerative diseases and enhance the quality of life for affected individuals.
Collapse
Affiliation(s)
- Mahan Farzan
- Student Research Committee, Shahrekord University of Medical Sciences, Shahrekord 8815713471, Iran
- Medical Plants Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord 8815783657, Iran
| | - Behnaz Abedi
- Department of Basic Sciences, Faculty of Veterinary Medicine, University of Tabriz, Tabriz 5166616471, Iran
| | - Iman Bhia
- Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran 1985717443, Iran
| | - Atossa Madanipour
- Student Research Committee, Alborz University of Medical Sciences, Karaj 3146883811, Iran
| | - Mahour Farzan
- Medical Plants Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord 8815783657, Iran
| | - Mohammad Bhia
- School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran 1996835113, Iran
| | - Ava Aghaei
- Student Research Committee, Shahrekord University of Medical Sciences, Shahrekord 8815713471, Iran
| | - Iman Kheirollahi
- Student Research Committee, Isfahan University of Medical Sciences, Isfahan 8174673441, Iran
| | - Mahzad Motallebi
- Nanomedicine Research Association (NRA), Universal Scientific Education and Research Network (USERN), Tehran 7616911319, Iran
| | - Hossein Amini-Khoei
- Medical Plants Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord 8815783657, Iran
| | - Yavuz Nuri Ertas
- Department of Biomedical Engineering, Erciyes University, Kayseri 38039, Turkey
- Department of Technical Sciences, Western Caspian University, AZ1001 Baku, Azerbaijan
| |
Collapse
|
21
|
McKeon SD, Perica MI, Calabro FJ, Foran W, Hetherington H, Moon CH, Luna B. Prefrontal Excitation/ Inhibition Balance Supports Adolescent Enhancements in Circuit Signal to Noise Ratio. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.15.608100. [PMID: 39229165 PMCID: PMC11370379 DOI: 10.1101/2024.08.15.608100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
The development and refinement of neuronal circuitry allow for stabilized and efficient neural recruitment, supporting adult-like behavioral performance. During adolescence, the maturation of PFC is proposed to be a critical period (CP) for executive function, driven by a break in balance between glutamatergic excitation and GABAergic inhibition (E/I) neurotransmission. During CPs, cortical circuitry fine-tunes to improve information processing and reliable responses to stimuli, shifting from spontaneous to evoked activity, enhancing the SNR, and promoting neural synchronization. Harnessing 7T MR spectroscopy and EEG in a longitudinal cohort (N = 164, ages 10-32 years, 283 neuroimaging sessions), we outline associations between age-related changes in glutamate and GABA neurotransmitters and EEG measures of cortical SNR. We find developmental decreases in spontaneous activity and increases in cortical SNR during our auditory steady state task using 40 Hz stimuli. Decreases in spontaneous activity were associated with glutamate levels in DLPFC, while increases in cortical SNR were associated with more balanced Glu and GABA levels. These changes were associated with improvements in working memory performance. This study provides evidence of CP plasticity in the human PFC during adolescence, leading to stabilized circuitry that allows for the optimal recruitment and integration of multisensory input, resulting in improved executive function.
Collapse
Affiliation(s)
- Shane D. McKeon
- Department of Bioengineering, University of Pittsburgh, PA, USA
- The Center for the Neural Basis of Cognition, University of Pittsburgh, PA, USA
| | - Maria I. Perica
- The Center for the Neural Basis of Cognition, University of Pittsburgh, PA, USA
- Department of Psychology, University of Pittsburgh, PA, USA
| | - Finnegan J. Calabro
- Department of Bioengineering, University of Pittsburgh, PA, USA
- The Center for the Neural Basis of Cognition, University of Pittsburgh, PA, USA
- Department of Psychiatry, University of Pittsburgh, PA, USA
| | - Will Foran
- Department of Psychiatry, University of Pittsburgh, PA, USA
| | - Hoby Hetherington
- Resonance Research Incorporated, Billerica, MA, USA
- Department of Radiology, University of Missouri, Columbia, MO, USA
| | - Chan-Hong Moon
- Department of Radiology, University of Pittsburgh, PA, USA
| | - Beatriz Luna
- The Center for the Neural Basis of Cognition, University of Pittsburgh, PA, USA
- Department of Psychiatry, University of Pittsburgh, PA, USA
| |
Collapse
|
22
|
Islam SNU, Kouser S, Hassan P, Asgher M, Shah AA, Khan NA. Gamma-aminobutyric acid interactions with phytohormones and its role in modulating abiotic and biotic stress in plants. STRESS BIOLOGY 2024; 4:36. [PMID: 39158750 PMCID: PMC11333426 DOI: 10.1007/s44154-024-00180-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 06/10/2024] [Indexed: 08/20/2024]
Abstract
Gamma-aminobutyric acid (GABA), a ubiquitous non-protein 4-carbon amino acid present in both prokaryotic and eukaryotic organisms. It is conventionally recognized as a neurotransmitter in mammals and plays a crucial role in plants. The context of this review centers on the impact of GABA in mitigating abiotic stresses induced by climate change, such as drought, salinity, heat, and heavy metal exposure. Beyond its neurotransmitter role, GABA emerges as a key player in diverse metabolic processes, safeguarding plants against multifaceted abiotic as well as biotic challenges. This comprehensive exploration delves into the GABA biosynthetic pathway, its transport mechanisms, and its intricate interplay with various abiotic stresses. The discussion extends to the nuanced relationship between GABA and phytohormones during abiotic stress acclimation, offering insights into the strategic development of mitigation strategies against these stresses. The delineation of GABA's crosstalk with phytohormones underscores its pivotal role in formulating crucial strategies for abiotic stress alleviation in plants.
Collapse
Affiliation(s)
- Syed Nazar Ul Islam
- Plant Physiology and Biochemistry Laboratory, Department of Botany, School of Biosciences and Biotechnology, Baba Ghulam Shah Badshah University, Rajouri, Jammu and Kashmir, 185234, India
| | - Shaista Kouser
- Plant Physiology and Biochemistry Laboratory, Department of Botany, School of Biosciences and Biotechnology, Baba Ghulam Shah Badshah University, Rajouri, Jammu and Kashmir, 185234, India
| | - Parveena Hassan
- School of Biosciences and Biotechnology, Baba Ghulam Shah Badshah University, Rajouri, Jammu and Kashmir, 185234, India
| | - Mohd Asgher
- Plant Physiology and Biochemistry Laboratory, Department of Botany, School of Biosciences and Biotechnology, Baba Ghulam Shah Badshah University, Rajouri, Jammu and Kashmir, 185234, India.
| | - Ali Asghar Shah
- School of Biosciences and Biotechnology, Baba Ghulam Shah Badshah University, Rajouri, Jammu and Kashmir, 185234, India
| | - Nafees A Khan
- Plant Physiology and Biochemistry Laboratory, Department of Botany, Aligarh Muslim University, Aligarh, 202002, India.
| |
Collapse
|
23
|
Wang J, O'Reilly M, Cooper IA, Chehrehasa F, Moody H, Beecher K. Mapping GABAergic projections that mediate feeding. Neurosci Biobehav Rev 2024; 163:105743. [PMID: 38821151 DOI: 10.1016/j.neubiorev.2024.105743] [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: 03/25/2024] [Revised: 05/23/2024] [Accepted: 05/28/2024] [Indexed: 06/02/2024]
Abstract
Neuroscience offers important insights into the pathogenesis and treatment of obesity by investigating neural circuits underpinning appetite and feeding. Gamma-aminobutyric acid (GABA), one of the most abundant neurotransmitters in the brain, and its associated receptors represent an array of pharmacologically targetable mediators of appetite signalling. Targeting the GABAergic system is therefore an increasingly investigated approach to obesity treatment. However, the many GABAergic projections that control feeding have yet to be collectively analysed. This review provides a comprehensive analysis of the relationship between GABAergic signalling and appetite by examining both foundational studies and the results of newly emerging chemogenetic/optogenetic experiments. A current snapshot of these efforts to map GABAergic projections influencing appetite is provided, and potential avenues for further investigation are provided.
Collapse
Affiliation(s)
- Joshua Wang
- School of Clinical Sciences, Faculty of Health, Queensland University of Technology, 2 George Street, Brisbane 4000, QLD, Australia.
| | - Max O'Reilly
- UQ Centre for Clinical Research, Faculty of Medicine, University of Queensland, Building 71/918 Royal Brisbane and Women's Hospital Campus, Herston 4029, QLD, Australia
| | | | - Fatemeh Chehrehasa
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, 2 George Street, Brisbane 4000, QLD, Australia
| | - Hayley Moody
- Queensland University of Technology, 2 George Street, Brisbane 4000, QLD, Australia
| | - Kate Beecher
- UQ Centre for Clinical Research, Faculty of Medicine, University of Queensland, Building 71/918 Royal Brisbane and Women's Hospital Campus, Herston 4029, QLD, Australia
| |
Collapse
|
24
|
Verma S, Singh V, Nagampalli V, Ponsky LE, Li CSR, Chao H, Gupta S. Ligand-gated ion channels as potential biomarkers for ADT-mediated cognitive decline in prostate cancer patients. Mol Carcinog 2024; 63:1051-1063. [PMID: 38482990 PMCID: PMC11096008 DOI: 10.1002/mc.23708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 02/01/2024] [Accepted: 02/13/2024] [Indexed: 05/16/2024]
Abstract
Men with prostate cancer are at increased risk of developing cognitive decline by the use of second-generation androgen signaling inhibitors. To date, reliable and sensitive biomarkers that could distinguish men at high risk of cognitive dysfunction under androgen deprivation therapy (ADT) have not been characterized. We used high-throughput transcriptional profiling utilizing human prostate cancer cell culture models mimicking ADT, biomarker selection using minimal common oncology data elements-cytoscape, and bioinformatic analyses employing Advaita® iPathwayGuide and DisGeNET for identification of disease-related gene associations. Validation analysis of genes was performed on brain neuronal and glial cells by quantitative real-time polymerase chain reaction assay. Our systematic analysis of androgen deprivation-associated genes involved multiple biological processes, including neuroactive ligand-receptor interaction, axon guidance, cytokine-cytokine receptor interaction, and metabolic and cancer signaling pathways. Genes associated with neuroreceptor ligand interaction, including gamma-aminobutyric acid (GABA) A and B receptors and nuclear core proteins, were identified as top upstream regulators. Functional enrichment and protein-protein interaction network analysis highlighted the role of ligand-gated ion channels (LGICs) and their receptors in cognitive dysfunction. Gene-disease association assigned forgetfulness, intellectual disability, visuospatial deficit, bipolar disorder, and other neurocognitive impairment with upregulation of type-1 angiotensin II receptor, brain-derived neurotrophic factor, GABA type B receptor subunit 2 (GABBR2), GABRA3, GABRA5, GABRB1, glycine receptor beta, glutamate ionotropic receptor N-methyl-D-aspartate receptor (NMDA) type subunit 1, glutamate ionotropic receptor NMDA type subunit 2D, 5-hydroxytryptamine receptor 1D, interferon beta 1, and nuclear receptor subfamily 3 group C member 1 as top differentially expressed genes. Validation studies of brain glial cells, neurons, and patients on ADT demonstrated the association of these genes with cognitive decline. Our findings highlight LGICs as potential biomarkers for ADT-mediated cognitive decline. Further validation of these biomarkers may lead to future practical clinical use.
Collapse
Affiliation(s)
- Shiv Verma
- Department of Urology, Case Western Reserve University, School of Medicine, Cleveland, OH 44106, USA
- The Urology Institute, University Hospitals Cleveland Medical Center, Cleveland, OH 44106 USA
| | - Vaibhav Singh
- Department of Pathology, Case Western Reserve University, School of Medicine, Cleveland, OH 44106, USA
| | | | - Lee E Ponsky
- Department of Urology, Case Western Reserve University, School of Medicine, Cleveland, OH 44106, USA
- The Urology Institute, University Hospitals Cleveland Medical Center, Cleveland, OH 44106 USA
| | - Chiang-Shan R Li
- Department of Psychiatry and of Neuroscience, Yale University School of Medicine, New Haven, CT 06519
| | - Herta Chao
- Department of Medicine & Yale Comprehensive Cancer Center, Yale University, New Haven, CT 06510, USA
| | - Sanjay Gupta
- Department of Urology, Case Western Reserve University, School of Medicine, Cleveland, OH 44106, USA
- The Urology Institute, University Hospitals Cleveland Medical Center, Cleveland, OH 44106 USA
- Department of Pathology, Case Western Reserve University, School of Medicine, Cleveland, OH 44106, USA
- Department of Pharmacology, Case Western Reserve University, School of Medicine, Cleveland, OH 44106, USA
- Department of Nutrition, Case Western Reserve University, School of Medicine, Cleveland, OH 44106, USA
- Division of General Medical Sciences, Case Comprehensive Cancer Center, Cleveland, OH 44106 USA
| |
Collapse
|
25
|
Topchiy I, Mohbat J, Folorunso OO, Wang ZZ, Lazcano-Etchebarne C, Engin E. GABA system as the cause and effect in early development. Neurosci Biobehav Rev 2024; 161:105651. [PMID: 38579901 PMCID: PMC11081854 DOI: 10.1016/j.neubiorev.2024.105651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 03/05/2024] [Accepted: 04/01/2024] [Indexed: 04/07/2024]
Abstract
GABA is the primary inhibitory neurotransmitter in the adult brain and through its actions on GABAARs, it protects against excitotoxicity and seizure activity, ensures temporal fidelity of neurotransmission, and regulates concerted rhythmic activity of neuronal populations. In the developing brain, the development of GABAergic neurons precedes that of glutamatergic neurons and the GABA system serves as a guide and framework for the development of other brain systems. Despite this early start, the maturation of the GABA system also continues well into the early postnatal period. In this review, we organize evidence around two scenarios based on the essential and protracted nature of GABA system development: 1) disruptions in the development of the GABA system can lead to large scale disruptions in other developmental processes (i.e., GABA as the cause), 2) protracted maturation of this system makes it vulnerable to the effects of developmental insults (i.e., GABA as the effect). While ample evidence supports the importance of GABA/GABAAR system in both scenarios, large gaps in existing knowledge prevent strong mechanistic conclusions.
Collapse
Affiliation(s)
- Irina Topchiy
- Division of Basic Neuroscience, McLean Hospital, Belmont, MA 02478, USA; Department of Psychiatry, Harvard Medical School, Boston, MA 02215, USA
| | - Julie Mohbat
- Division of Basic Neuroscience, McLean Hospital, Belmont, MA 02478, USA; Department of Psychiatry, Harvard Medical School, Boston, MA 02215, USA; School of Life Sciences, Ecole Polytechnique Federale de Lausanne, Lausanne CH-1015, Switzerland
| | - Oluwarotimi O Folorunso
- Division of Basic Neuroscience, McLean Hospital, Belmont, MA 02478, USA; Department of Psychiatry, Harvard Medical School, Boston, MA 02215, USA
| | - Ziyi Zephyr Wang
- Division of Basic Neuroscience, McLean Hospital, Belmont, MA 02478, USA; Department of Psychiatry, Harvard Medical School, Boston, MA 02215, USA
| | | | - Elif Engin
- Division of Basic Neuroscience, McLean Hospital, Belmont, MA 02478, USA; Department of Psychiatry, Harvard Medical School, Boston, MA 02215, USA.
| |
Collapse
|
26
|
Ng ACH, Chahine M, Scantlebury MH, Appendino JP. Channelopathies in epilepsy: an overview of clinical presentations, pathogenic mechanisms, and therapeutic insights. J Neurol 2024; 271:3063-3094. [PMID: 38607431 DOI: 10.1007/s00415-024-12352-x] [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: 03/17/2024] [Revised: 03/24/2024] [Accepted: 03/25/2024] [Indexed: 04/13/2024]
Abstract
Pathogenic variants in genes encoding ion channels are causal for various pediatric and adult neurological conditions. In particular, several epilepsy syndromes have been identified to be caused by specific channelopathies. These encompass a spectrum from self-limited epilepsies to developmental and epileptic encephalopathies spanning genetic and acquired causes. Several of these channelopathies have exquisite responses to specific antiseizure medications (ASMs), while others ASMs may prove ineffective or even worsen seizures. Some channelopathies demonstrate phenotypic pleiotropy and can cause other neurological conditions outside of epilepsy. This review aims to provide a comprehensive exploration of the pathophysiology of seizure generation, ion channels implicated in epilepsy, and several genetic epilepsies due to ion channel dysfunction. We outline the clinical presentation, pathogenesis, and the current state of basic science and clinical research for these channelopathies. In addition, we briefly look at potential precision therapy approaches emerging for these disorders.
Collapse
Affiliation(s)
- Andy Cheuk-Him Ng
- Clinical Neuroscience and Pediatric Neurology, Department of Pediatrics, Cumming School of Medicine, Alberta Children's Hospital, University of Calgary, 28 Oki Drive NW, Calgary, AB, T3B 6A8, Canada
- Division of Neurology, Department of Pediatrics, Faculty of Medicine and Dentistry, University of Alberta and Stollery Children's Hospital, Edmonton, AB, Canada
| | - Mohamed Chahine
- Department of Medicine, Faculty of Medicine, Université Laval, Quebec City, QC, Canada
- CERVO, Brain Research Centre, Quebec City, Canada
| | - Morris H Scantlebury
- Clinical Neuroscience and Pediatric Neurology, Department of Pediatrics, Cumming School of Medicine, Alberta Children's Hospital, University of Calgary, 28 Oki Drive NW, Calgary, AB, T3B 6A8, Canada
- Hotchkiss Brain Institute and Alberta Children's Hospital Research Institute, Calgary, Canada
| | - Juan P Appendino
- Clinical Neuroscience and Pediatric Neurology, Department of Pediatrics, Cumming School of Medicine, Alberta Children's Hospital, University of Calgary, 28 Oki Drive NW, Calgary, AB, T3B 6A8, Canada.
| |
Collapse
|
27
|
Park H, Lee CH. The Impact of Pulmonary Disorders on Neurological Health (Lung-Brain Axis). Immune Netw 2024; 24:e20. [PMID: 38974208 PMCID: PMC11224666 DOI: 10.4110/in.2024.24.e20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 04/30/2024] [Accepted: 05/23/2024] [Indexed: 07/09/2024] Open
Abstract
The brain and lungs, vital organs in the body, play essential roles in maintaining overall well-being and survival. These organs interact through complex and sophisticated bi-directional pathways known as the 'lung-brain axis', facilitated by their close proximity and neural connections. Numerous studies have underscored the mediation of the lung-brain axis by inflammatory responses and hypoxia-induced damage, which are pivotal to the progression of both pulmonary and neurological diseases. This review aims to delve into how pulmonary diseases, including acute/chronic airway diseases and pulmonary conditions, can instigate neurological disorders such as stroke, Alzheimer's disease, and Parkinson's disease. Additionally, we highlight the emerging research on the lung microbiome which, drawing parallels between the gut and lungs in terms of microbiome contents, may play a significant role in modulating brain health. Ultimately, this review paves the way for exciting avenues of future research and therapeutics in addressing respiratory and neurological diseases.
Collapse
Affiliation(s)
- Hongryeol Park
- Department of Tissue Morphogenesis, Max-Planck Institute for Molecular Biomedicine, Muenster 48149, Germany
| | - Chan Hee Lee
- Department of Biomedical Science, Hallym University, Chuncheon 24252, Korea
- Program of Material Science for Medicine and Pharmaceutics, Hallym University, Chuncheon 24252, Korea
| |
Collapse
|
28
|
Chujan S, Vajeethaveesin N, Satayavivad J, Kitkumthorn N. Identification of Molecular Mechanisms of Ameloblastoma and Drug Repositioning by Integration of Bioinformatics Analysis and Molecular Docking Simulation. Bioinform Biol Insights 2024; 18:11779322241256459. [PMID: 38812739 PMCID: PMC11135093 DOI: 10.1177/11779322241256459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Accepted: 05/02/2024] [Indexed: 05/31/2024] Open
Abstract
Background Ameloblastoma (AM) is a benign tumor locally originated from odontogenic epithelium that is commonly found in the jaw. This tumor makes aggressive invasions and has a high recurrence rate. This study aimed to investigate the differentially expressed genes (DEGs), biological function alterations, disease targets, and existing drugs for AM using bioinformatics analysis. Methods The data set of AM was retrieved from the GEO database (GSE132474) and identified the DEGs using bioinformatics analysis. The biological alteration analysis was applied to Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways. Protein-protein interaction (PPI) network analysis and hub gene identification were screened through NetworkAnalyst. The transcription factor-protein network was constructed via OmicsNet. We also identified candidate compounds from L1000CDS2 database. The target of AM and candidate compounds were verified using docking simulation. Results Totally, 611 DEGs were identified. The biological function enrichment analysis revealed glycosaminoglycan and GABA (γ-aminobutyric acid) signaling were most significantly up-regulated and down-regulated in AM, respectively. Subsequently, hub genes and transcription factors were screened via the network and showed FOS protein was found in both networks. Furthermore, we evaluated FOS protein to be a therapeutic target in AMs. Candidate compounds were screened and verified using docking simulation. Tanespimycin showed the greatest affinity binding value to bind FOS protein. Conclusions This study presented the underlying molecular mechanisms of disease pathogenesis, biological alteration, and important pathways of AMs and provided a candidate compound, Tanespimycin, targeting FOS protein for the treatment of AMs.
Collapse
Affiliation(s)
- Suthipong Chujan
- Laboratory of Pharmacology, Chulabhorn Research Institute, Bangkok, Thailand
- Center of Excellence on Environmental Health and Toxicology (EHT), Office of the Permanent Secretary (OPS), Ministry of Higher Education, Science, Research and Innovation (MHESI), Bangkok, Thailand
| | | | - Jutamaad Satayavivad
- Laboratory of Pharmacology, Chulabhorn Research Institute, Bangkok, Thailand
- Center of Excellence on Environmental Health and Toxicology (EHT), Office of the Permanent Secretary (OPS), Ministry of Higher Education, Science, Research and Innovation (MHESI), Bangkok, Thailand
| | - Nakarin Kitkumthorn
- Department of Oral Biology, Faculty of Dentistry, Mahidol University, Bangkok, Thailand
| |
Collapse
|
29
|
Eisen A, Pioro EP, Goutman SA, Kiernan MC. Nanoplastics and Neurodegeneration in ALS. Brain Sci 2024; 14:471. [PMID: 38790450 PMCID: PMC11119293 DOI: 10.3390/brainsci14050471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 05/02/2024] [Accepted: 05/06/2024] [Indexed: 05/26/2024] Open
Abstract
Plastic production, which exceeds one million tons per year, is of global concern. The constituent low-density polymers enable spread over large distances and micro/nano particles (MNPLs) induce organ toxicity via digestion, inhalation, and skin contact. Particles have been documented in all human tissues including breast milk. MNPLs, especially weathered particles, can breach the blood-brain barrier, inducing neurotoxicity. This has been documented in non-human species, and in human-induced pluripotent stem cell lines. Within the brain, MNPLs initiate an inflammatory response with pro-inflammatory cytokine production, oxidative stress with generation of reactive oxygen species, and mitochondrial dysfunction. Glutamate and GABA neurotransmitter dysfunction also ensues with alteration of excitatory/inhibitory balance in favor of reduced inhibition and resultant neuro-excitation. Inflammation and cortical hyperexcitability are key abnormalities involved in the pathogenic cascade of amyotrophic lateral sclerosis (ALS) and are intricately related to the mislocalization and aggregation of TDP-43, a hallmark of ALS. Water and many foods contain MNPLs and in humans, ingestion is the main form of exposure. Digestion of plastics within the gut can alter their properties, rendering them more toxic, and they cause gut microbiome dysbiosis and a dysfunctional gut-brain axis. This is recognized as a trigger and/or aggravating factor for ALS. ALS is associated with a long (years or decades) preclinical period and neonates and infants are exposed to MNPLs through breast milk, milk substitutes, and toys. This endangers a time of intense neurogenesis and establishment of neuronal circuitry, setting the stage for development of neurodegeneration in later life. MNPL neurotoxicity should be considered as a yet unrecognized risk factor for ALS and related diseases.
Collapse
Affiliation(s)
- Andrew Eisen
- Division of Neurology, Department of Medicine, University of British Columbia, Vancouver, BC V6S 1Z3, Canada;
| | - Erik P. Pioro
- Division of Neurology, Department of Medicine, University of British Columbia, Vancouver, BC V6S 1Z3, Canada;
| | - Stephen A. Goutman
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109, USA;
| | | |
Collapse
|
30
|
Cheng H, Chen D, Li X, Al-Sheikh U, Duan D, Fan Y, Zhu L, Zeng W, Hu Z, Tong X, Zhao G, Zhang Y, Zou W, Duan S, Kang L. Phasic/tonic glial GABA differentially transduce for olfactory adaptation and neuronal aging. Neuron 2024; 112:1473-1486.e6. [PMID: 38447577 DOI: 10.1016/j.neuron.2024.02.006] [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: 05/05/2023] [Revised: 11/11/2023] [Accepted: 02/06/2024] [Indexed: 03/08/2024]
Abstract
Phasic (fast) and tonic (sustained) inhibition of γ-aminobutyric acid (GABA) are fundamental for regulating day-to-day activities, neuronal excitability, and plasticity. However, the mechanisms and physiological functions of glial GABA transductions remain poorly understood. Here, we report that the AMsh glia in Caenorhabditis elegans exhibit both phasic and tonic GABAergic signaling, which distinctively regulate olfactory adaptation and neuronal aging. Through genetic screening, we find that GABA permeates through bestrophin-9/-13/-14 anion channels from AMsh glia, which primarily activate the metabolic GABAB receptor GBB-1 in the neighboring ASH sensory neurons. This tonic action of glial GABA regulates the age-associated changes of ASH neurons and olfactory responses via a conserved signaling pathway, inducing neuroprotection. In addition, the calcium-evoked, vesicular glial GABA release acts upon the ionotropic GABAA receptor LGC-38 in ASH neurons to regulate olfactory adaptation. These findings underscore the fundamental significance of glial GABA in maintaining healthy aging and neuronal stability.
Collapse
Affiliation(s)
- Hankui Cheng
- Department of Neurology of the Fourth Affiliated Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Yiwu 322000, China; MOE Frontier Science Center for Brain Science and Brain machine Integration, NHC and CAMS Key Laboratory of Medical Neurobiology, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310053, China
| | - Du Chen
- Department of Neurology of the Fourth Affiliated Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Yiwu 322000, China; MOE Frontier Science Center for Brain Science and Brain machine Integration, NHC and CAMS Key Laboratory of Medical Neurobiology, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310053, China
| | - Xiao Li
- Department of Neurology of the Fourth Affiliated Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Yiwu 322000, China; MOE Frontier Science Center for Brain Science and Brain machine Integration, NHC and CAMS Key Laboratory of Medical Neurobiology, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310053, China
| | - Umar Al-Sheikh
- Department of Neurology of the Fourth Affiliated Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Yiwu 322000, China; MOE Frontier Science Center for Brain Science and Brain machine Integration, NHC and CAMS Key Laboratory of Medical Neurobiology, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310053, China
| | - Duo Duan
- Department of Neurology of the Fourth Affiliated Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Yiwu 322000, China; MOE Frontier Science Center for Brain Science and Brain machine Integration, NHC and CAMS Key Laboratory of Medical Neurobiology, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310053, China
| | - Yuedan Fan
- Department of Neurology of the Fourth Affiliated Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Yiwu 322000, China; MOE Frontier Science Center for Brain Science and Brain machine Integration, NHC and CAMS Key Laboratory of Medical Neurobiology, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310053, China
| | - Linhui Zhu
- Department of Neurology of the Fourth Affiliated Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Yiwu 322000, China; MOE Frontier Science Center for Brain Science and Brain machine Integration, NHC and CAMS Key Laboratory of Medical Neurobiology, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310053, China
| | - Wanxin Zeng
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Zhitao Hu
- Department of Neuroscience, City University of Hong Kong, Kowloon, China
| | - Xiajing Tong
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Guohua Zhao
- Department of Neurology of the Fourth Affiliated Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Yiwu 322000, China
| | - Yongming Zhang
- Department of Ophthalmology of the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu 322000, China
| | - Wenjuan Zou
- Department of Neurology of the Fourth Affiliated Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Yiwu 322000, China; MOE Frontier Science Center for Brain Science and Brain machine Integration, NHC and CAMS Key Laboratory of Medical Neurobiology, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310053, China
| | - Shumin Duan
- MOE Frontier Science Center for Brain Science and Brain machine Integration, NHC and CAMS Key Laboratory of Medical Neurobiology, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310053, China
| | - Lijun Kang
- Department of Neurology of the Fourth Affiliated Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Yiwu 322000, China; MOE Frontier Science Center for Brain Science and Brain machine Integration, NHC and CAMS Key Laboratory of Medical Neurobiology, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310053, China.
| |
Collapse
|
31
|
Thirion A, Loots DT, Williams ME, Solomons R, Mason S. 1H-NMR metabolomics investigation of CSF from children with HIV reveals altered neuroenergetics due to persistent immune activation. Front Neurosci 2024; 18:1270041. [PMID: 38745940 PMCID: PMC11091326 DOI: 10.3389/fnins.2024.1270041] [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: 07/31/2023] [Accepted: 04/15/2024] [Indexed: 05/16/2024] Open
Abstract
Background HIV can invade the central nervous system (CNS) early during infection, invading perivascular macrophages and microglia, which, in turn, release viral particles and immune mediators that dysregulate all brain cell types. Consequently, children living with HIV often present with neurodevelopmental delays. Methods In this study, we used proton nuclear magnetic resonance (1H-NMR) spectroscopy to analyze the neurometabolic profile of HIV infection using cerebrospinal fluid samples obtained from 17 HIV+ and 50 HIV- South African children. Results Nine metabolites, including glucose, lactate, glutamine, 1,2-propanediol, acetone, 3-hydroxybutyrate, acetoacetate, 2-hydroxybutyrate, and myo-inositol, showed significant differences when comparing children infected with HIV and those uninfected. These metabolites may be associated with activation of the innate immune response and disruption of neuroenergetics pathways. Conclusion These results elucidate the neurometabolic state of children infected with HIV, including upregulation of glycolysis, dysregulation of ketone body metabolism, and elevated reactive oxygen species production. Furthermore, we hypothesize that neuroinflammation alters astrocyte-neuron communication, lowering neuronal activity in children infected with HIV, which may contribute to the neurodevelopmental delay often observed in this population.
Collapse
Affiliation(s)
- Anicia Thirion
- Department of Biochemistry, Human Metabolomics, Faculty of Natural and Agricultural Sciences, North-West University, Potchefstroom, South Africa
| | - Du Toit Loots
- Department of Biochemistry, Human Metabolomics, Faculty of Natural and Agricultural Sciences, North-West University, Potchefstroom, South Africa
| | - Monray E. Williams
- Department of Biochemistry, Human Metabolomics, Faculty of Natural and Agricultural Sciences, North-West University, Potchefstroom, South Africa
| | - Regan Solomons
- Department of Pediatrics and Child Health, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg, South Africa
| | - Shayne Mason
- Department of Biochemistry, Human Metabolomics, Faculty of Natural and Agricultural Sciences, North-West University, Potchefstroom, South Africa
| |
Collapse
|
32
|
Yang SH, Kim YJ, Yang HR, Park SU, Kim JG, Kim JK. Metabolic Profiling in Plasma and Brain Induced by 17β-Estradiol Supplementation in Ovariectomized Mice. ACS OMEGA 2024; 9:18212-18223. [PMID: 38680363 PMCID: PMC11044158 DOI: 10.1021/acsomega.3c10399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 03/25/2024] [Accepted: 03/27/2024] [Indexed: 05/01/2024]
Abstract
17β-Estradiol is an ovarian hormone that regulates energy circulation and storage by acting on the central nervous system. However, the metabolic differences between the blood and brain when stimulated by 17β-estradiol are poorly understood. Moreover, research using menopause-induced models to investigate primary metabolites in the blood and brain is limited. Thus, this study aimed to identify metabolic changes in the plasma and brain resulting from 17β-estradiol supplementation in an estrogen-deficient mouse model. Three groups of mice were utilized: sham-operated mice (Sham), ovariectomized mice (OVX), and ovariectomized mice that received a weekly supplementation of 17β-estradiol (E2). Plasma and brain samples from these mice were subjected to metabolic analysis using gas chromatography-time-of-flight-mass spectrometry. Compared with the plasma samples from the Sham and OVX groups, the plasma samples from the E2 group contained higher contents of branched-chain amino acids (BCAAs), such as valine, isoleucine, and leucine. Meanwhile, the brain samples from the E2 group contained higher contents of most metabolites, including BCAAs, neurotransmitters, tricarboxylic acid cycle intermediates, and fatty acids, than those from the two other groups. This study is the first to reveal differences in energy metabolism induced by 17β-estradiol supplementation through brain metabolic profiling of ovariectomized mice, emphasizing the importance of brain metabolic profiling in menopausal hormone research.
Collapse
Affiliation(s)
- So Hwi Yang
- Division
of Life Sciences, College of Life Sciences, and Bioengineering, Incheon National University, Incheon 22012, Republic of Korea
| | - Ye Jin Kim
- Division
of Life Sciences, College of Life Sciences, and Bioengineering, Incheon National University, Incheon 22012, Republic of Korea
| | - Hye Rim Yang
- Division
of Life Sciences, College of Life Sciences, and Bioengineering, Incheon National University, Incheon 22012, Republic of Korea
| | - Sang Un Park
- Department
of Crop Science, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon 34134, Republic of Korea
| | - Jae Geun Kim
- Division
of Life Sciences, College of Life Sciences, and Bioengineering, Incheon National University, Incheon 22012, Republic of Korea
| | - Jae Kwang Kim
- Division
of Life Sciences, College of Life Sciences, and Bioengineering, Incheon National University, Incheon 22012, Republic of Korea
- Convergence
Research Center for Insect Vectors, Incheon
National University, Incheon 22012, Republic
of Korea
| |
Collapse
|
33
|
Hussain N, Muccee F. In-silico characterization of GABAT protein found in gut-brain axis associated bacteria of healthy individuals and multiple sclerosis patients. Saudi J Biol Sci 2024; 31:103939. [PMID: 38352114 PMCID: PMC10859293 DOI: 10.1016/j.sjbs.2024.103939] [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: 06/20/2023] [Revised: 01/10/2024] [Accepted: 01/26/2024] [Indexed: 02/16/2024] Open
Abstract
Background Multiple sclerosis (MS) is a neurodegenerative disease characterized by inflammation and demyelination of neurons. There is evidence to suggest that level of a neurotransmitter gamma-aminobutyric acid (GABA), due to the degradation by γ-aminobutyric acid transaminase (GABAT), is reduced in certain areas of the brain in MS patients. MS is always accompanied by gut bacteria dysbiosis. In healthy individuals, Faecalibacterium sp. while in MS patients A. calcoaceticus, Clostridium sp. and S. typhimurium are found abundantly. Although all these microbes produce GABAT but only in MS patients this enzyme significantly degrades GABA. Objective Present study is an attempt to characterize the GABAT protein sequences of these bacteria. Methodology Sequences of GABAT protein were retrieved from Uniprot database. Sequences were analyzed by Protparam, Gneg-mPLoc, SOSUI, PFP-FunDSeqE, Pepwheel program, PROTEUS and Alphafold and SAVES servers, MEME suite and HDOCK server. Results In healthy individuals gastrointestinal tract (GIT) bacteria, GABAT protein was present in inner-membrane with α helix content (61 and 62%) and β sheet content (5%), 4-helical cytokines functional domains. It has greater number of B-cell epitopes and more complex 3D configuration as compared to MS patients GIT bacterial enzymes. Conclusion Present study might enable us to modify the GABAT encoding gene and enzyme through site-directed mutagenesis in pathogenic bacteria thus reducing their potential of causing MS.
Collapse
Affiliation(s)
- Nadia Hussain
- Department of Pharmaceutical Sciences, College of Pharmacy, Al Ain University, Al Ain Campus, Al Ain 64141, United Arab Emirates
- AAU Health and Biomedical Research Center, Al Ain University, Abu Dhabi Campus, Abu Dhabi P. O. Box 112612, United Arab Emirates
| | - Fatima Muccee
- School of Biochemistry and Biotechnology, University of Punjab, Lahore 52254, Pakistan
| |
Collapse
|
34
|
Sinha JK, Trisal A, Ghosh S, Gupta S, Singh KK, Han SS, Mahapatra M, Abomughaid MM, Abomughayedh AM, Almutary AG, Iqbal D, Bhaskar R, Mishra PC, Jha SK, Jha NK, Singh AK. Psychedelics for alzheimer's disease-related dementia: Unveiling therapeutic possibilities and pathways. Ageing Res Rev 2024; 96:102211. [PMID: 38307424 DOI: 10.1016/j.arr.2024.102211] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 01/25/2024] [Accepted: 01/26/2024] [Indexed: 02/04/2024]
Abstract
Psychedelics have traditionally been used for spiritual and recreational purposes, but recent developments in psychotherapy have highlighted their potential as therapeutic agents. These compounds, which act as potent 5-hydroxytryptamine (5HT) agonists, have been recognized for their ability to enhance neural plasticity through the activation of the serotoninergic and glutamatergic systems. However, the implications of these findings for the treatment of neurodegenerative disorders, particularly dementia, have not been fully explored. In recent years, studies have revealed the modulatory and beneficial effects of psychedelics in the context of dementia, specifically Alzheimer's disease (AD)-related dementia, which lacks a definitive cure. Psychedelics such as N,N-dimethyltryptamine (DMT), lysergic acid diethylamide (LSD), and Psilocybin have shown potential in mitigating the effects of this debilitating disease. These compounds not only target neurotransmitter imbalances but also act at the molecular level to modulate signalling pathways in AD, including the brain-derived neurotrophic factor signalling pathway and the subsequent activation of mammalian target of rapamycin and other autophagy regulators. Therefore, the controlled and dose-dependent administration of psychedelics represents a novel therapeutic intervention worth exploring and considering for the development of drugs for the treatment of AD-related dementia. In this article, we critically examined the literature that sheds light on the therapeutic possibilities and pathways of psychedelics for AD-related dementia. While this emerging field of research holds great promise, further studies are necessary to elucidate the long-term safety, efficacy, and optimal treatment protocols. Ultimately, the integration of psychedelics into the current treatment paradigm may provide a transformative approach for addressing the unmet needs of individuals living with AD-related dementia and their caregivers.
Collapse
Affiliation(s)
| | - Anchal Trisal
- Department of Biosciences, Jamia Millia Islamia, New Delhi 110025, India
| | - Shampa Ghosh
- GloNeuro, Sector 107, Vishwakarma Road, Noida 201301, India
| | - Saurabh Gupta
- Department of Biotechnology, GLA University, Mathura, Uttar Pradesh, India
| | - Krishna Kumar Singh
- Symbiosis Centre for Information Technology (SCIT), Rajiv Gandhi InfoTech Park, Hinjawadi, Pune, Maharashtra 411057, India
| | - Sung Soo Han
- School of Chemical Engineering, Yeungnam University, Gyeonsang 38541, the Republic of Korea; Research Institute of Cell Culture, Yeungnam University, 280 Daehak-Ro, Gyeongsan 38541, the Republic of Korea
| | | | - Mosleh Mohammad Abomughaid
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, University of Bisha, Bisha 61922, Saudi Arabia
| | - Ali M Abomughayedh
- Pharmacy Department, Aseer Central Hospital, Ministry of Health, Saudi Arabia
| | - Abdulmajeed G Almutary
- Department of Biomedical Sciences, College of Health Sciences, Abu Dhabi University, Abu Dhabi P.O. Box 59911, United Arab Emirates
| | - Danish Iqbal
- Department of Health Information Management, College of Applied Medical Sciences, Buraydah Private Colleges, Buraydah 51418, Saudi Arabia
| | - Rakesh Bhaskar
- School of Chemical Engineering, Yeungnam University, Gyeonsang 38541, the Republic of Korea; Research Institute of Cell Culture, Yeungnam University, 280 Daehak-Ro, Gyeongsan 38541, the Republic of Korea.
| | - Prabhu Chandra Mishra
- Department of Biotechnology, School of Engineering and Technology, Sharda University, Greater Noida, India
| | - Saurabh Kumar Jha
- Department of Zoology, Kalindi College, University of Delhi, 110008, India.
| | - Niraj Kumar Jha
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India; Centre of Research Impact and Outcome, Chitkara University, Rajpura 140401, Punjab, India; School of Bioengineering & Biosciences, Lovely Professional University, Phagwara 144411, India; Department of Biotechnology Engineering and Food Technology, Chandigarh University, Mohali, India.
| | - Abhishek Kumar Singh
- Manipal Centre for Biotherapeutics Research, Manipal Academy of Higher Education, Manipal, Karnataka 576104, India.
| |
Collapse
|
35
|
Issahaku AR, Wilhelm A, Schutte-Smith M, Erasmus E, Visser H. Elucidating the binding mechanisms of GABA and Muscimol as an avenue to discover novel GABA-mimetic small molecules. J Biomol Struct Dyn 2024:1-16. [PMID: 38520326 DOI: 10.1080/07391102.2024.2331088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 03/10/2024] [Indexed: 03/25/2024]
Abstract
Gamma-aminobutyric acid (GABA) signaling is the principal inhibitory pathway in the central nervous system. It is critical in neuronal cell proliferation and fate determination. Any aberration in GABA inhibition results in psychiatric and neurological diseases. Thus, modulating GABAergic neurotransmission has become the basis of drug therapy for psychiatric and several neurological diseases. Though GABA and muscimol are classical inhibitors of GABA receptors, the search for novel inhibitors continues unabated. In this study, the binding mechanism of GABA and muscimol was elucidated and applied in the search for small molecule GABAergic inhibitors using comprehensive computational techniques. It was revealed that a high-affinity binding of GABA and muscimol was mediated by a water molecule involving α1Thr129 and then stabilized by strong interactions including salt bridges with β2Glu155 and α1Arg66 amidst hydrogen bonds, π-π stacking, and π -cation interactions with other residues. The binding of GABA and muscimol was also characterized by stability and deeper penetration into the hydrophobic core of the protein which resulted in conformational changes of the binding pocket and domain, by inducing correlated motions of the residues. Thermodynamics analysis showed GABA and muscimol exhibited total binding free energies of -19.85 ± 8.83 Kcal/mol and -26.55 ± 3.42 Kcal/mol, respectively. A pharmacophore model search, based on the energy contributions of implicating binding residues, resulted in the identification of ZINC68604167, ZINC19735138, ZINC04202466, ZINC00901626, and ZINC01532854 as potential GABA-mimetic compounds from metabolites and natural products libraries. This study has elucidated the binding mechanisms of GABA and muscimol and successfully applied in the identification of GABA-mimetic compounds.
Collapse
Affiliation(s)
| | - Anke Wilhelm
- Department of Chemistry, University of the Free State, Bloemfontein, South Africa
| | | | - Elizabeth Erasmus
- Department of Chemistry, University of the Free State, Bloemfontein, South Africa
| | - Hendrik Visser
- Department of Chemistry, University of the Free State, Bloemfontein, South Africa
| |
Collapse
|
36
|
AbdEl-Raouf K, Farrag HSH, Rashed R, Ismail MA, El-Ganzuri MA, El-Sayed WM. New bithiophene derivative attenuated Alzheimer's disease induced by aluminum in a rat model via antioxidant activity and restoration of neuronal and synaptic transmission. J Trace Elem Med Biol 2024; 82:127352. [PMID: 38070385 DOI: 10.1016/j.jtemb.2023.127352] [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: 10/03/2023] [Revised: 11/27/2023] [Accepted: 12/01/2023] [Indexed: 01/10/2024]
Abstract
BACKGROUND One of the hypotheses that leads to an increased incidence of Alzheimer's disease (AD) is the accumulation of aluminum in the brain's frontal cortex. The present study aimed to evaluate the therapeutic role of a novel bithiophene derivative at two doses against AlCl3-induced AD in a rat model. METHODOLOGY Adult male rats were divided into six groups, 18 rats each. Group 1: naïve animals, group 2: animals received a daily oral administration of bithiophene dissolved in DMSO (1 mg/kg) for 30 days every other day, groups 3-6: animals received a daily oral administration of AlCl3 (100 mg/kg/day) for 45 consecutive days. Groups 4 and 5 received an oral administration of low or high dose of the bithiophene (0.5 or 1 mg/kg, respectively). Group 6; Animals were treated with a daily oral dose of memantine (20 mg/kg) for 30 consecutive days. MAIN FINDINGS Al disturbed the antioxidant milieu, elevated the lipid peroxidation, and depleted the antioxidants. It also disturbed the synaptic neurotransmission by elevating the activities of acetylcholine esterase and monoamine oxidase resulting in the depletion of dopamine and serotonin and accumulation of glutamate and norepinephrine. Al also deteriorated the expression of genes involved in apoptosis and the production of amyloid-β plaques as well as phosphorylation of tau. The new bithiophene at the low dose reversed most of the previous deleterious effects of aluminum in the cerebral cortex and was in many instances superior to the reference drug; memantine. CONCLUSION Taking together, the bithiophene modulated the AD etiology through antioxidant activity, prevention of neuronal and synaptic loss, and probably mitigating the formation of amyloid-β plaques and phosphorylation of tau.
Collapse
Affiliation(s)
- Kholoud AbdEl-Raouf
- Department of Zoology, Faculty of Science, Ain Shams University, Abbassia, 11566 Cairo, Egypt
| | | | - Rashed Rashed
- Department of Zoology, Faculty of Science, Ain Shams University, Abbassia, 11566 Cairo, Egypt
| | - Mohamed A Ismail
- Department of Chemistry, Faculty of Science, Mansoura University, Mansoura 35516, Egypt
| | - Monir A El-Ganzuri
- Department of Zoology, Faculty of Science, Ain Shams University, Abbassia, 11566 Cairo, Egypt
| | - Wael M El-Sayed
- Department of Zoology, Faculty of Science, Ain Shams University, Abbassia, 11566 Cairo, Egypt.
| |
Collapse
|
37
|
Deslauriers JC, Ghotkar RP, Russ LA, Jarman JA, Martin RM, Tippett RG, Sumathipala SH, Burton DF, Cole DC, Marsden KC. Cyfip2 controls the acoustic startle threshold through FMRP, actin polymerization, and GABA B receptor function. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.12.22.573054. [PMID: 38187577 PMCID: PMC10769380 DOI: 10.1101/2023.12.22.573054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
Animals process a constant stream of sensory input, and to survive they must detect and respond to dangerous stimuli while ignoring innocuous or irrelevant ones. Behavioral responses are elicited when certain properties of a stimulus such as its intensity or size reach a critical value, and such behavioral thresholds can be a simple and effective mechanism to filter sensory information. For example, the acoustic startle response is a conserved and stereotyped defensive behavior induced by sudden loud sounds, but dysregulation of the threshold to initiate this behavior can result in startle hypersensitivity that is associated with sensory processing disorders including schizophrenia and autism. Through a previous forward genetic screen for regulators of the startle threshold a nonsense mutation in Cytoplasmic Fragile X Messenger Ribonucleoprotein (FMRP)-interacting protein 2 (cyfip2) was found that causes startle hypersensitivity in zebrafish larvae, but the molecular mechanisms by which Cyfip2 establishes the acoustic startle threshold are unknown. Here we used conditional transgenic rescue and CRISPR/Cas9 to determine that Cyfip2 acts though both Rac1 and FMRP pathways, but not the closely related FXR1 or FXR2, to establish the acoustic startle threshold during early neurodevelopment. To identify proteins and pathways that may be downstream effectors of Rac1 and FMRP, we performed a candidate-based drug screen that indicated that Cyfip2 can also act acutely to maintain the startle threshold branched actin polymerization and N-methyl D-aspartate receptors (NMDARs). To complement this approach, we used unbiased discovery proteomics to determine that loss of Cyfip2 alters cytoskeletal and extracellular matrix components while also disrupting oxidative phosphorylation and GABA receptor signaling. Finally, we functionally validated our proteomics findings by showing that activating GABAB receptors, which like NMDARs are also FMRP targets, restores normal startle sensitivity in cyfip2 mutants. Together, these data reveal multiple mechanisms by which Cyfip2 regulates excitatory/inhibitory balance in the startle circuit to control the processing of acoustic information.
Collapse
Affiliation(s)
- Jacob C. Deslauriers
- Department of Biological Sciences, North Carolina State University, Raleigh, North Carolina, USA
| | - Rohit P. Ghotkar
- Department of Biological Sciences, North Carolina State University, Raleigh, North Carolina, USA
- Current address: Putnam Associates, Boston, Massachusetts, USA
| | - Lindsey A. Russ
- Department of Biological Sciences, North Carolina State University, Raleigh, North Carolina, USA
- Current address: Department of Pharmacology & Physiology, Georgetown University, Washington D.C., USA
| | - Jordan A. Jarman
- Department of Biological Sciences, North Carolina State University, Raleigh, North Carolina, USA
- Current address: Department of Physiology and Biophysics, Boston University, Boston, MA, USA
| | - Rubia M. Martin
- Department of Biological Sciences, North Carolina State University, Raleigh, North Carolina, USA
- Current address: U.S. Environmental Protection Agency, Raleigh-Durham-Chapel Hill, North Carolina, USA
| | - Rachel G. Tippett
- Department of Biological Sciences, North Carolina State University, Raleigh, North Carolina, USA
| | - Sureni H. Sumathipala
- Department of Biological Sciences, North Carolina State University, Raleigh, North Carolina, USA
| | - Derek F. Burton
- Department of Biological Sciences, North Carolina State University, Raleigh, North Carolina, USA
| | - D. Chris Cole
- Department of Biological Sciences, North Carolina State University, Raleigh, North Carolina, USA
| | - Kurt C. Marsden
- Department of Biological Sciences, North Carolina State University, Raleigh, North Carolina, USA
- Center for Human Health and the Environment (CHHE), North Carolina State University, Raleigh, North Carolina, USA
| |
Collapse
|
38
|
Sérgio Galina Spilla C, Luiza Decanini Miranda de Souza A, Maria Guissoni Campos L, da Silveira Cruz-Machado S, Pinato L. LPS-induced inflammation in rats during pregnancy reduces maternal melatonin and impairs neurochemistry and behavior of adult male offspring. Brain Res 2024; 1824:148692. [PMID: 38036237 DOI: 10.1016/j.brainres.2023.148692] [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: 04/30/2023] [Revised: 11/22/2023] [Accepted: 11/27/2023] [Indexed: 12/02/2023]
Abstract
Inflammation during pregnancy can induce neurodevelopmental changes that affect the neurological health of offspring. Elevated levels of circulating inflammatory cytokines have been shown to decrease nocturnal melatonin synthesis by the pineal gland, potentially impacting fetal development. This study aimed to assess the effects of LPS-induced inflammation on melatonin concentrations in the plasma of pregnant female rats and explore resulting neurochemical and behavioral changes in their offspring. Our findings revealed that pregnant rats injected with LPS experienced decreased nocturnal melatonin levels in their plasma, with an increase in diurnal melatonin content. The offspring exhibited reduced performance in tests evaluating motor coordination and spatial memory compared to control subjects. Immunohistochemical analysis indicated a decline in calbindin immunoreactivity in Purkinje cells in the cerebellum. Additionally, the hippocampus displayed an increase in IBA-1 and calretinin expression, coupled with a reduction in parvalbumin expression in the offspring of the LPS group. Collectively, this study provides compelling evidence that an inflammatory state can lead to a reduction in melatonin synthesis in pregnant females, potentially impacting the neurodevelopment of offspring, including neuronal, glial, motor, and cognitive aspects. Subsequent studies will further elucidate the mechanisms underlying inflammation-induced maternal melatonin reduction and its impact on offspring neurodevelopment.
Collapse
Affiliation(s)
| | | | | | | | - Luciana Pinato
- Department of Speech, Language and Hearing Sciences, São Paulo State University (UNESP), Marília, São Paulo 17525-900, Brazil.
| |
Collapse
|
39
|
Tempone MH, Borges-Martins VP, César F, Alexandrino-Mattos DP, de Figueiredo CS, Raony Í, dos Santos AA, Duarte-Silva AT, Dias MS, Freitas HR, de Araújo EG, Ribeiro-Resende VT, Cossenza M, P. Silva H, P. de Carvalho R, Ventura ALM, Calaza KC, Silveira MS, Kubrusly RCC, de Melo Reis RA. The Healthy and Diseased Retina Seen through Neuron-Glia Interactions. Int J Mol Sci 2024; 25:1120. [PMID: 38256192 PMCID: PMC10817105 DOI: 10.3390/ijms25021120] [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: 12/21/2023] [Revised: 01/10/2024] [Accepted: 01/12/2024] [Indexed: 01/24/2024] Open
Abstract
The retina is the sensory tissue responsible for the first stages of visual processing, with a conserved anatomy and functional architecture among vertebrates. To date, retinal eye diseases, such as diabetic retinopathy, age-related macular degeneration, retinitis pigmentosa, glaucoma, and others, affect nearly 170 million people worldwide, resulting in vision loss and blindness. To tackle retinal disorders, the developing retina has been explored as a versatile model to study intercellular signaling, as it presents a broad neurochemical repertoire that has been approached in the last decades in terms of signaling and diseases. Retina, dissociated and arranged as typical cultures, as mixed or neuron- and glia-enriched, and/or organized as neurospheres and/or as organoids, are valuable to understand both neuronal and glial compartments, which have contributed to revealing roles and mechanisms between transmitter systems as well as antioxidants, trophic factors, and extracellular matrix proteins. Overall, contributions in understanding neurogenesis, tissue development, differentiation, connectivity, plasticity, and cell death are widely described. A complete access to the genome of several vertebrates, as well as the recent transcriptome at the single cell level at different stages of development, also anticipates future advances in providing cues to target blinding diseases or retinal dysfunctions.
Collapse
Affiliation(s)
- Matheus H. Tempone
- Laboratory of Neurochemistry, Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro 21949-000, Brazil; (M.H.T.); (F.C.); (D.P.A.-M.); (V.T.R.-R.)
| | - Vladimir P. Borges-Martins
- Department of Physiology and Pharmacology, Biomedical Institute and Program of Neurosciences, Federal Fluminense University, Niterói 24020-150, Brazil; (V.P.B.-M.); (A.A.d.S.); (M.C.); (R.C.C.K.)
| | - Felipe César
- Laboratory of Neurochemistry, Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro 21949-000, Brazil; (M.H.T.); (F.C.); (D.P.A.-M.); (V.T.R.-R.)
| | - Dio Pablo Alexandrino-Mattos
- Laboratory of Neurochemistry, Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro 21949-000, Brazil; (M.H.T.); (F.C.); (D.P.A.-M.); (V.T.R.-R.)
| | - Camila S. de Figueiredo
- Department of Neurobiology and Program of Neurosciences, Institute of Biology, Federal Fluminense University, Niterói 24020-141, Brazil; (C.S.d.F.); (A.T.D.-S.); (E.G.d.A.); (R.P.d.C.); (A.L.M.V.); (K.C.C.)
| | - Ícaro Raony
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (Í.R.); (H.R.F.)
| | - Aline Araujo dos Santos
- Department of Physiology and Pharmacology, Biomedical Institute and Program of Neurosciences, Federal Fluminense University, Niterói 24020-150, Brazil; (V.P.B.-M.); (A.A.d.S.); (M.C.); (R.C.C.K.)
| | - Aline Teixeira Duarte-Silva
- Department of Neurobiology and Program of Neurosciences, Institute of Biology, Federal Fluminense University, Niterói 24020-141, Brazil; (C.S.d.F.); (A.T.D.-S.); (E.G.d.A.); (R.P.d.C.); (A.L.M.V.); (K.C.C.)
| | - Mariana Santana Dias
- Laboratory of Gene Therapy and Viral Vectors, Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro 21949-000, Brazil; (M.S.D.); (H.P.S.)
| | - Hércules Rezende Freitas
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (Í.R.); (H.R.F.)
| | - Elisabeth G. de Araújo
- Department of Neurobiology and Program of Neurosciences, Institute of Biology, Federal Fluminense University, Niterói 24020-141, Brazil; (C.S.d.F.); (A.T.D.-S.); (E.G.d.A.); (R.P.d.C.); (A.L.M.V.); (K.C.C.)
- National Institute of Science and Technology on Neuroimmunomodulation—INCT-NIM, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro 21040-360, Brazil
| | - Victor Tulio Ribeiro-Resende
- Laboratory of Neurochemistry, Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro 21949-000, Brazil; (M.H.T.); (F.C.); (D.P.A.-M.); (V.T.R.-R.)
| | - Marcelo Cossenza
- Department of Physiology and Pharmacology, Biomedical Institute and Program of Neurosciences, Federal Fluminense University, Niterói 24020-150, Brazil; (V.P.B.-M.); (A.A.d.S.); (M.C.); (R.C.C.K.)
| | - Hilda P. Silva
- Laboratory of Gene Therapy and Viral Vectors, Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro 21949-000, Brazil; (M.S.D.); (H.P.S.)
| | - Roberto P. de Carvalho
- Department of Neurobiology and Program of Neurosciences, Institute of Biology, Federal Fluminense University, Niterói 24020-141, Brazil; (C.S.d.F.); (A.T.D.-S.); (E.G.d.A.); (R.P.d.C.); (A.L.M.V.); (K.C.C.)
| | - Ana L. M. Ventura
- Department of Neurobiology and Program of Neurosciences, Institute of Biology, Federal Fluminense University, Niterói 24020-141, Brazil; (C.S.d.F.); (A.T.D.-S.); (E.G.d.A.); (R.P.d.C.); (A.L.M.V.); (K.C.C.)
| | - Karin C. Calaza
- Department of Neurobiology and Program of Neurosciences, Institute of Biology, Federal Fluminense University, Niterói 24020-141, Brazil; (C.S.d.F.); (A.T.D.-S.); (E.G.d.A.); (R.P.d.C.); (A.L.M.V.); (K.C.C.)
| | - Mariana S. Silveira
- Laboratory for Investigation in Neuroregeneration and Development, Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro 21949-000, Brazil;
| | - Regina C. C. Kubrusly
- Department of Physiology and Pharmacology, Biomedical Institute and Program of Neurosciences, Federal Fluminense University, Niterói 24020-150, Brazil; (V.P.B.-M.); (A.A.d.S.); (M.C.); (R.C.C.K.)
| | - Ricardo A. de Melo Reis
- Laboratory of Neurochemistry, Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro 21949-000, Brazil; (M.H.T.); (F.C.); (D.P.A.-M.); (V.T.R.-R.)
| |
Collapse
|
40
|
Singh S, Hämäläinen RH. The Roles of Cystatin B in the Brain and Pathophysiological Mechanisms of Progressive Myoclonic Epilepsy Type 1. Cells 2024; 13:170. [PMID: 38247861 PMCID: PMC10814315 DOI: 10.3390/cells13020170] [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: 12/07/2023] [Revised: 01/03/2024] [Accepted: 01/15/2024] [Indexed: 01/23/2024] Open
Abstract
Progressive myoclonic epilepsy type 1 (EPM1) is an autosomal recessive disorder, also known as Unverricht-Lundborg disease (ULD). EPM1 patients suffer from photo-sensitive seizures, stimulus-sensitive myoclonus, nocturnal myoclonic seizures, ataxia and dysarthria. In addition, cerebral ataxia and impaired GABAergic inhibition are typically present. EPM1 is caused by mutations in the Cystatin B gene (CSTB). The CSTB protein functions as an intracellular thiol protease inhibitor and inhibits Cathepsin function. It also plays a crucial role in brain development and regulates various functions in neurons beyond maintaining cellular proteostasis. These include controlling cell proliferation and differentiation, synaptic functions and protection against oxidative stress, likely through regulation of mitochondrial function. Depending on the differentiation stage and status of neurons, the protein localizes either to the cytoplasm, nucleus, lysosomes or mitochondria. Further, CSTB can also be secreted to the extracellular matrix for interneuron rearrangement and migration. In this review, we will review the various functions of CSTB in the brain and discuss the putative pathophysiological mechanism underlying EPM1.
Collapse
Affiliation(s)
| | - Riikka H. Hämäläinen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Neulaniementie 2, 70211 Kuopio, Finland;
| |
Collapse
|
41
|
Kiernan MC, Farrar MA. Emerging role for sphingolipids in the genetics of amyotrophic lateral sclerosis. J Neurol Neurosurg Psychiatry 2024; 95:101-102. [PMID: 38041659 DOI: 10.1136/jnnp-2023-332719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Accepted: 10/17/2023] [Indexed: 12/03/2023]
Affiliation(s)
- Matthew C Kiernan
- Bushell Chair of Neurology, Brain and Mind Centre, University of Sydney, Sydney, New South Wales, Australia
- Neurology, Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia
| | - Michelle Anne Farrar
- Neurology, Sydney Children's Hospital Randwick, Randwick, New South Wales, Australia
- School of Women's and Children's Health, UNSW Medicine, UNSW, Sydney, New South Wales, Australia
| |
Collapse
|
42
|
Alkhayat D, Khawaji ZY, Sunyur AM, Sanyour OA, Badawi AS. Overview of Paraneoplastic Autoantibody-Mediated Cognitive Impairment and Behavioral Changes: A Narrative Review. Cureus 2024; 16:e51787. [PMID: 38322089 PMCID: PMC10846349 DOI: 10.7759/cureus.51787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/06/2024] [Indexed: 02/08/2024] Open
Abstract
Cognitive dysfunction and behavioral change can be some of the manifestations of cancer, occurring as a part of paraneoplastic neurological syndrome, most commonly in small cell lung cancer. Paraneoplastic limbic encephalitis is the leading cause of cognitive disturbance and abnormal behavior in paraneoplastic syndromes, which is usually autoantibody-mediated. Autoantibodies are the main contributors to the development of cognitive dysfunction and behavioral change in cancer patients, with studies suggesting a higher liability for antibody-positive cancer patients to be affected. Anti-NMDAR and anti-AMPAR are antibodies targeted against surface antigens, manifesting predominantly as memory disturbance, abnormal behavior, psychiatric symptoms, and seizures. Other surface antigen-targeted antibodies include anti-GABA, anti-CASPR2, and anti-LGI1, which were shown to have cognitive function impairment and abnormal behavior as some of the main presentations, predominantly affecting memory. Cognitive deterioration and changes in behavior were also relatively common with some of the intracellular antigen-targeted antibodies, including anti-Hu, anti-SOX1, anti-PCA2, and anti-Zic2. Affected behavior and cognition, however, were reported less commonly in other paraneoplastic antibodies against intracellular antigens (anti-Yo, anti-GAD, anti-Ma2, anti-Ri, anti-CV2, and anti-KLHL11). Our article will provide a comprehensive review of the clinical manifestations of cognitive impairment and behavioral changes among cancer patients who develop paraneoplastic syndrome. Additionally, this review will discuss the role of specific paraneoplastic autoantibodies and the clinical spectrum linked to each separately.
Collapse
Affiliation(s)
| | | | - Amal M Sunyur
- Medicine and Surgery, Taibah University, Medina, SAU
| | | | | |
Collapse
|
43
|
Wang L, Lu Z, Teng Y, Pan W, Li Y, Su S, Chang J, Zhao M. Cognitive impairment is associated with BDNF-TrkB signaling mediating synaptic damage and reduction of amino acid neurotransmitters in heart failure. FASEB J 2024; 38:e23351. [PMID: 38085181 DOI: 10.1096/fj.202301699rr] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 11/14/2023] [Accepted: 11/21/2023] [Indexed: 12/18/2023]
Abstract
Heart failure (HF) is often accompanied by cognitive impairment (CI). Brain-derived neurotrophic factor (BDNF) deficiency is closely associated with CI. However, the role and mechanism of BDNF in HF with CI is still not fully understood. Here, the case-control study was designed including 25 HF without CI patients (HF-NCI) and 50 HF with CI patients (HF-CI) to investigate the predictive value of BDNF in HF-CI while animal and cell experiments were used for mechanism research. Results found that BDNF levels in serum neuronal-derived exosomes were downregulated in HF-CI patients. There was no significant difference in serum BDNF levels among the two groups. HF rats showed obvious impairment in learning and memory; also, they had reduced thickness and length of postsynaptic density (PSD) and increased synaptic cleft width. Expression of BDNF, TrkB, PSD95, and VGLUT1 was significantly decreased in HF rats brain. In addition, compared with sham rats, amino acids were significantly reduced with no changes in the acetylcholine and monoamine neurotransmitters. Further examination showed that the number of synaptic bifurcations and the expression of BDNF, TrkB, PSD95, and VGLUT1 were all decreased in the neurons that interfered with BDNF-siRNA compared with those in the negative control neurons. Together, our results demonstrated that neuronal-derived exosomal BDNF act as effective biomarkers for prediction of HF-CI. The decrease of BDNF in the brain triggers synaptic structural damage and a decline in amino acid neurotransmitters via the BDNF-TrkB-PSD95/VGLUT1 pathway. This discovery unveils a novel pathological mechanism underlying cognitive impairment following heart failure.
Collapse
Affiliation(s)
- Lei Wang
- Key Laboratory of Chinese Internal Medicine of Ministry of Education, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Ziwen Lu
- Key Laboratory of Chinese Internal Medicine of Ministry of Education, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Yu Teng
- Key Laboratory of Chinese Internal Medicine of Ministry of Education, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Weibing Pan
- Key Laboratory of Chinese Internal Medicine of Ministry of Education, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Yang Li
- Key Laboratory of Chinese Internal Medicine of Ministry of Education, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Sha Su
- Key Laboratory of Chinese Internal Medicine of Ministry of Education, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Jingling Chang
- Department of Neurology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Mingjing Zhao
- Key Laboratory of Chinese Internal Medicine of Ministry of Education, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
44
|
Parrella NF, Hill AT, Dipnall LM, Loke YJ, Enticott PG, Ford TC. Inhibitory dysfunction and social processing difficulties in autism: A comprehensive narrative review. J Psychiatr Res 2024; 169:113-125. [PMID: 38016393 DOI: 10.1016/j.jpsychires.2023.11.014] [Citation(s) in RCA: 1] [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: 04/13/2023] [Revised: 09/04/2023] [Accepted: 11/15/2023] [Indexed: 11/30/2023]
Abstract
The primary inhibitory neurotransmitter γ-aminobutyric acid (GABA) has a prominent role in regulating neural development and function, with disruption to GABAergic signalling linked to behavioural phenotypes associated with neurodevelopmental disorders, particularly autism. Such neurochemical disruption, likely resulting from diverse genetic and molecular mechanisms, particularly during early development, can subsequently affect the cellular balance of excitation and inhibition in neuronal circuits, which may account for the social processing difficulties observed in autism and related conditions. This comprehensive narrative review integrates diverse streams of research from several disciplines, including molecular neurobiology, genetics, epigenetics, and systems neuroscience. In so doing it aims to elucidate the relevance of inhibitory dysfunction to autism, with specific focus on social processing difficulties that represent a core feature of this disorder. Many of the social processing difficulties experienced in autism have been linked to higher levels of the excitatory neurotransmitter glutamate and/or lower levels of inhibitory GABA. While current therapeutic options for social difficulties in autism are largely limited to behavioural interventions, this review highlights the psychopharmacological studies that explore the utility of GABA modulation in alleviating such difficulties.
Collapse
Affiliation(s)
| | - Aron T Hill
- Cognitive Neuroscience Unit, School of Psychology, Deakin University, Geelong, Australia; Department of Psychiatry, Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Lillian M Dipnall
- Cognitive Neuroscience Unit, School of Psychology, Deakin University, Geelong, Australia; Early Life Epigenetics Group, Deakin University, Geelong, Australia
| | - Yuk Jing Loke
- Epigenetics Group, Murdoch Children's Research Institute, Melbourne, Victoria, Australia; Department of Paediatrics, The University of Melbourne, Melbourne, Victoria, Australia
| | - Peter G Enticott
- Cognitive Neuroscience Unit, School of Psychology, Deakin University, Geelong, Australia
| | - Talitha C Ford
- Cognitive Neuroscience Unit, School of Psychology, Deakin University, Geelong, Australia; Centre for Human Psychopharmacology, Faculty of Health, Arts and Design, Swinburne University of Technology, Melbourne, Victoria, Australia
| |
Collapse
|
45
|
Havranek T, Bacova Z, Bakos J. Oxytocin, GABA, and dopamine interplay in autism. Endocr Regul 2024; 58:105-114. [PMID: 38656256 DOI: 10.2478/enr-2024-0012] [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] [Indexed: 04/26/2024] Open
Abstract
Oxytocin plays an important role in brain development and is associated with various neurotransmitter systems in the brain. Abnormalities in the production, secretion, and distribution of oxytocin in the brain, at least during some stages of the development, are critical for the pathogenesis of neuropsychiatric diseases, particularly in the autism spectrum disorder. The etiology of autism includes changes in local sensory and dopaminergic areas of the brain, which are also supplied by the hypothalamic sources of oxytocin. It is very important to understand their mutual relationship. In this review, the relationship of oxytocin with several components of the dopaminergic system, gamma-aminobutyric acid (GABA) inhibitory neurotransmission and their alterations in the autism spectrum disorder is discussed. Special attention has been paid to the results describing a reduced expression of inhibitory GABAergic markers in the brain in the context of dopaminergic areas in various models of autism. It is presumed that the altered GABAergic neurotransmission, due to the absence or dysfunction of oxytocin at certain developmental stages, disinhibits the dopaminergic signaling and contributes to the autism symptoms.
Collapse
Affiliation(s)
- Tomas Havranek
- Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, Bratislava, Slovakia
- Faculty of Medicine, Comenius University, Bratislava, Slovakia
| | - Zuzana Bacova
- Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Jan Bakos
- Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, Bratislava, Slovakia
- Faculty of Medicine, Comenius University, Bratislava, Slovakia
| |
Collapse
|
46
|
Loukil A, Ebright E, Uezu A, Gao Y, Soderling SH, Goetz SC. Identification of new ciliary signaling pathways in the brain and insights into neurological disorders. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.20.572700. [PMID: 38187761 PMCID: PMC10769350 DOI: 10.1101/2023.12.20.572700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
Primary cilia are conserved sensory hubs essential for signaling transduction and embryonic development. Ciliary dysfunction causes a variety of developmental syndromes with neurological features and cognitive impairment, whose basis mostly remains unknown. Despite connections to neural function, the primary cilium remains an overlooked organelle in the brain. Most neurons have a primary cilium; however, it is still unclear how this organelle modulates brain architecture and function, given the lack of any systemic dissection of neuronal ciliary signaling. Here, we present the first in vivo glance at the molecular composition of cilia in the mouse brain. We have adapted in vivo BioID (iBioID), targeting the biotin ligase BioID2 to primary cilia in neurons. We identified tissue-specific signaling networks enriched in neuronal cilia, including Eph/Ephrin and GABA receptor signaling pathways. Our iBioID ciliary network presents a wealth of neural ciliary hits that provides new insights into neurological disorders. Our findings are a promising first step in defining the fundamentals of ciliary signaling and their roles in shaping neural circuits and behavior. This work can be extended to pathological conditions of the brain, aiming to identify the molecular pathways disrupted in the brain cilium. Hence, finding novel therapeutic strategies will help uncover and leverage the therapeutic potential of the neuronal cilium.
Collapse
Affiliation(s)
- Abdelhalim Loukil
- Pediatrics and Rare Diseases Group, Sanford Research, Sioux Falls, SD 57104, USA
- Department of Pediatrics, Sanford School of Medicine, University of South Dakota, Sioux Falls, SD 57105, USA
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Emma Ebright
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Akiyoshi Uezu
- Department of Cell Biology, Duke University Medical School, Durham, NC 27710, USA
| | - Yudong Gao
- Department of Cell Biology, Duke University Medical School, Durham, NC 27710, USA
| | - Scott H Soderling
- Department of Cell Biology, Duke University Medical School, Durham, NC 27710, USA
| | - Sarah C. Goetz
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC 27710, USA
| |
Collapse
|
47
|
Hotha A, Ganesh CB. GABA-immunoreactive neurons in the Central Nervous System of the viviparous teleost Poecilia sphenops. J Chem Neuroanat 2023; 133:102339. [PMID: 37689218 DOI: 10.1016/j.jchemneu.2023.102339] [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: 07/19/2023] [Revised: 09/06/2023] [Accepted: 09/06/2023] [Indexed: 09/11/2023]
Abstract
Gamma-aminobutyric acid (GABA) functions as the primary inhibitory neurotransmitter within the central nervous system (CNS) of vertebrates. In this study, we examined the distribution pattern of GABA-immunoreactive (GABA-ir) cells and fibres in the CNS of the viviparous teleost Poecilia sphenops using immunofluorescence method. GABA immunoreactivity was seen in the glomerular, mitral, and granular layers of the olfactory bulbs, as well as in most parts of the dorsal and ventral telencephalon. The preoptic area consisted of a small cluster of GABA-ir cells, whereas extensively labelled GABA-ir neurons were observed in the hypothalamic areas, including the paraventricular organ, tuberal hypothalamus, nucleus recessus lateralis, nucleus recessus posterioris, and inferior lobes. In the thalamus, GABA-positive neurons were only found in the ventral thalamic and central posterior thalamic nuclei, whereas the dorsal part of the nucleus pretectalis periventricularis consisted of a few GABA-ir cells. GABA-immunoreactivity was extensively seen in the alar and basal subdivisions of the midbrain, whereas in the rhombencephalon, GABA-ir cells and fibres were found in the cerebellum, motor nucleus of glossopharyngeal and vagal nerves, nucleus commissuralis of Cajal, and reticular formation. In the spinal cord, GABA-ir cells and fibres were observed in the dorsal horn, ventral horn, and around the central canal. Overall, the extensive distribution of GABA-ir cells and fibres throughout the CNS suggests several roles for GABA, including the neuroendocrine, viscerosensory, and somatosensory functions, for the first time in a viviparous teleost.
Collapse
Affiliation(s)
- Achyutham Hotha
- Neuroendocrinology Research Laboratory, Department of Studies in Zoology, Karnatak University, Dharwad 580 003, India
| | - C B Ganesh
- Neuroendocrinology Research Laboratory, Department of Studies in Zoology, Karnatak University, Dharwad 580 003, India.
| |
Collapse
|
48
|
Abstract
Rett syndrome is a neurodevelopmental disorder caused by loss-of-function mutations in the methyl-CpG binding protein-2 (MeCP2) gene that is characterized by epilepsy, intellectual disability, autistic features, speech deficits, and sleep and breathing abnormalities. Neurologically, patients with all three disorders display microcephaly, aberrant dendritic morphology, reduced spine density, and an imbalance of excitatory/inhibitory signaling. Loss-of-function mutations in the cyclin-dependent kinase-like 5 (CDKL5) and FOXG1 genes also cause similar behavioral and neurobiological defects and were referred to as congenital or variant Rett syndrome. The relatively recent realization that CDKL5 deficiency disorder (CDD), FOXG1 syndrome, and Rett syndrome are distinct neurodevelopmental disorders with some distinctive features have resulted in separate focus being placed on each disorder with the assumption that distinct molecular mechanisms underlie their pathogenesis. However, given that many of the core symptoms and neurological features are shared, it is likely that the disorders share some critical molecular underpinnings. This review discusses the possibility that deregulation of common molecules in neurons and astrocytes plays a central role in key behavioral and neurological abnormalities in all three disorders. These include KCC2, a chloride transporter, vGlut1, a vesicular glutamate transporter, GluD1, an orphan-glutamate receptor subunit, and PSD-95, a postsynaptic scaffolding protein. We propose that reduced expression or activity of KCC2, vGlut1, PSD-95, and AKT, along with increased expression of GluD1, is involved in the excitatory/inhibitory that represents a key aspect in all three disorders. In addition, astrocyte-derived brain-derived neurotrophic factor (BDNF), insulin-like growth factor 1 (IGF-1), and inflammatory cytokines likely affect the expression and functioning of these molecules resulting in disease-associated abnormalities.
Collapse
Affiliation(s)
- Santosh R D’Mello
- Department of Biological Sciences, Louisiana State University Shreveport, Shreveport, LA 71104, USA
| |
Collapse
|
49
|
Basu SK, Kapse KJ, Murnick J, Pradhan S, Spoehr E, Zhang A, Andescavage N, Nino G, du Plessis AJ, Limperopoulos C. Impact of bronchopulmonary dysplasia on brain GABA concentrations in preterm infants: Prospective cohort study. Early Hum Dev 2023; 186:105860. [PMID: 37757548 PMCID: PMC10843009 DOI: 10.1016/j.earlhumdev.2023.105860] [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: 08/31/2023] [Accepted: 09/19/2023] [Indexed: 09/29/2023]
Abstract
BACKGROUND Bronchopulmonary dysplasia (BPD) is associated with cognitive-behavioral deficits in very preterm (VPT) infants, often in the absence of structural brain injury. Advanced GABA-editing techniques like Mescher-Garwood point resolved spectroscopy (MEGA-PRESS) can quantify in-vivo gamma-aminobutyric acid (GABA+, with macromolecules) and glutamate (Glx, with glutamine) concentrations to investigate for neurophysiologic perturbations in the developing brain of VPT infants. OBJECTIVE To investigate the relationship between the severity of BPD and basal-ganglia GABA+ and Glx concentrations in VPT infants. METHODS MRI studies were performed on a 3 T scanner in a cohort of VPT infants [born ≤32 weeks gestational age (GA)] without major structural brain injury and healthy-term infants (>37 weeks GA) at term-equivalent age. MEGA-PRESS (TE68ms, TR2000ms, 256averages) sequence was acquired from the right basal-ganglia voxel (∼3cm3) and metabolite concentrations were quantified in institutional units (i.u.). We stratified VPT infants into no/mild (grade 0/1) and moderate-severe (grade 2/3) BPD. RESULTS Reliable MEGA-PRESS data was available from 63 subjects: 29 healthy-term and 34 VPT infants without major structural brain injury. VPT infants with moderate-severe BPD (n = 20) had the lowest right basal-ganglia GABA+ (median 1.88 vs. 2.28 vs. 2.12 i.u., p = 0.025) and GABA+/choline (0.73 vs. 0.99 vs. 0.88, p = 0.004) in comparison to infants with no/mild BPD and healthy-term infants. The GABA+/Glx ratio was lower (0.34 vs. 0.44, p = 0.034) in VPT infants with moderate-severe BPD than in infants with no/mild BPD. CONCLUSIONS Reduced GABA+ and GABA+/Glx in VPT infants with moderate-severe BPD indicate neurophysiologic perturbations which could serve as early biomarkers of future cognitive deficits.
Collapse
Affiliation(s)
- Sudeepta K Basu
- Neonatology, Children's National Hospital, Washington, D.C., USA; Developing Brain Institute, Children's National Hospital, Washington, D.C., USA; The George Washington University School of Medicine, Washington, D.C., USA
| | - Kushal J Kapse
- Developing Brain Institute, Children's National Hospital, Washington, D.C., USA
| | - Jonathan Murnick
- The George Washington University School of Medicine, Washington, D.C., USA; Division of Diagnostic Imaging and Radiology, Children's National Hospital, Washington, D.C., USA
| | - Subechhya Pradhan
- Developing Brain Institute, Children's National Hospital, Washington, D.C., USA; The George Washington University School of Medicine, Washington, D.C., USA
| | - Emma Spoehr
- Developing Brain Institute, Children's National Hospital, Washington, D.C., USA
| | - Anqing Zhang
- The George Washington University School of Medicine, Washington, D.C., USA; Division of Biostatistics and Epidemiology, Children's National Hospital, Washington, D.C., USA
| | - Nickie Andescavage
- Neonatology, Children's National Hospital, Washington, D.C., USA; Developing Brain Institute, Children's National Hospital, Washington, D.C., USA; The George Washington University School of Medicine, Washington, D.C., USA; Division of Neurology, Children's National Hospital, Washington, D.C., USA
| | - Gustavo Nino
- The George Washington University School of Medicine, Washington, D.C., USA; Division of Pulmonary and Sleep Medicine, Children's National Hospital, Washington, D.C., USA
| | - Adre J du Plessis
- The George Washington University School of Medicine, Washington, D.C., USA; Division of Neurology, Children's National Hospital, Washington, D.C., USA; Perinatal Pediatrics institute, Children's National Hospital, Washington, D.C., USA
| | - Catherine Limperopoulos
- Developing Brain Institute, Children's National Hospital, Washington, D.C., USA; The George Washington University School of Medicine, Washington, D.C., USA; Division of Diagnostic Imaging and Radiology, Children's National Hospital, Washington, D.C., USA; Division of Neurology, Children's National Hospital, Washington, D.C., USA.
| |
Collapse
|
50
|
Tizabi Y, Bennani S, El Kouhen N, Getachew B, Aschner M. Interaction of Heavy Metal Lead with Gut Microbiota: Implications for Autism Spectrum Disorder. Biomolecules 2023; 13:1549. [PMID: 37892231 PMCID: PMC10605213 DOI: 10.3390/biom13101549] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 10/17/2023] [Indexed: 10/29/2023] Open
Abstract
Autism Spectrum Disorder (ASD), a neurodevelopmental disorder characterized by persistent deficits in social interaction and communication, manifests in early childhood and is followed by restricted and stereotyped behaviors, interests, or activities in adolescence and adulthood (DSM-V). Although genetics and environmental factors have been implicated, the exact causes of ASD have yet to be fully characterized. New evidence suggests that dysbiosis or perturbation in gut microbiota (GM) and exposure to lead (Pb) may play important roles in ASD etiology. Pb is a toxic heavy metal that has been linked to a wide range of negative health outcomes, including anemia, encephalopathy, gastroenteric diseases, and, more importantly, cognitive and behavioral problems inherent to ASD. Pb exposure can disrupt GM, which is essential for maintaining overall health. GM, consisting of trillions of microorganisms, has been shown to play a crucial role in the development of various physiological and psychological functions. GM interacts with the brain in a bidirectional manner referred to as the "Gut-Brain Axis (GBA)". In this review, following a general overview of ASD and GM, the interaction of Pb with GM in the context of ASD is emphasized. The potential exploitation of this interaction for therapeutic purposes is also touched upon.
Collapse
Affiliation(s)
- Yousef Tizabi
- Department of Pharmacology, Howard University College of Medicine, Washington, DC 20059, USA
| | - Samia Bennani
- Faculty of Medicine and Pharmacy of Casablanca, Hassan II University, Casablanca 20100, Morocco
| | - Nacer El Kouhen
- Faculty of Medicine and Pharmacy of Casablanca, Hassan II University, Casablanca 20100, Morocco
| | - Bruk Getachew
- Department of Pharmacology, Howard University College of Medicine, Washington, DC 20059, USA
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, USA;
| |
Collapse
|