1
|
Kajita Y, Ono K, Kaneda S, Mushiake H. Pterostilben upregulates GAD67-mediated GABA synthesis in hippocampal parvalbumin-positive cells. Neuroscience 2025; 573:482-490. [PMID: 40189131 DOI: 10.1016/j.neuroscience.2025.03.060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2025] [Revised: 02/21/2025] [Accepted: 03/25/2025] [Indexed: 04/12/2025]
Abstract
BACKGROUND AND PURPOSE Pterostilbene (PTE, 3,5-dimethoxy-4'-hydroxystilbene) is a naturally occurring polyphenol which has antiepileptic properties, and can be utilized as a prophylaxis in patients with seizures and who are at risk of developing epilepsy. However, the effects of PTE on the gamma-aminobutyric acid (GABA)-mediated (GABAergic) nervous system are poorly understood. This study aimed to evaluate the effects of PTE on the GABAergic neurons. EXPERIMENTAL APPROACH Male Long-Evans rats were orally administered PTE (200 mg/kg, 5 % Tween 80 in saline) for 10 days using a plastic sonde. The control group was treated with 5 % Tween 80 in saline in a similar manner. Approximately 24 h after the last treatment, we fixed and removed the brains and examined GABA, GAD67, and GAD65 expression in the hippocampal layers using immunohistochemical analysis. In addition, changed GAD expression was compared between the two main GABAergic subtypes, PV+ and SOM+ cells. KEY RESULTS We demonstrated that PTE treatment increased GABA expression in the hippocampus. These effects can be attributed to GAD67-mediated GABA synthesis in the PV+ cells. CONCLUSIONS AND IMPLICATIONS Our results uncovered that PTE increased GABA synthesis, suggesting that PTE has potential as a prophylactic drug for patients at risk of developing epilepsy.
Collapse
Affiliation(s)
- Yuki Kajita
- Department of Physiology, Tohoku University School of Medicine, 2-1 Seiryo-machi, Aobaku, Sendai 980-8575, Japan.
| | - Ko Ono
- Department of Physiology, Tohoku University School of Medicine, 2-1 Seiryo-machi, Aobaku, Sendai 980-8575, Japan
| | - Saya Kaneda
- Department of Physiology, Tohoku University School of Medicine, 2-1 Seiryo-machi, Aobaku, Sendai 980-8575, Japan
| | - Hajime Mushiake
- Department of Physiology, Tohoku University School of Medicine, 2-1 Seiryo-machi, Aobaku, Sendai 980-8575, Japan
| |
Collapse
|
2
|
Chen H, LaFlamme CW, Wang YD, Blan AW, Koehler N, Mendonca Moraes R, Olszewski AR, Almanza Fuerte EP, Bonkowski ES, Bajpai R, Lavado A, Pruett-Miller SM, Mefford HC. Patient-derived models of UBA5-associated encephalopathy identify defects in neurodevelopment and highlight potential therapeutic avenues. Sci Transl Med 2025; 17:eadn8417. [PMID: 40333994 DOI: 10.1126/scitranslmed.adn8417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 01/17/2025] [Accepted: 04/16/2025] [Indexed: 05/09/2025]
Abstract
UBA5 encodes for the E1 enzyme of the UFMylation cascade, which plays an essential role in endoplasmic reticulum (ER) homeostasis. The clinical phenotypes of UBA5-associated encephalopathy include developmental delays, epilepsy, and intellectual disability. To date, there is no humanized neuronal model to study the cellular and molecular consequences of UBA5 pathogenic variants. We developed and characterized patient-derived cortical organoid cultures from two patients with compound heterozygous variants in UBA5. Both shared the same missense variant, which encodes a hypomorphic allele (p.A371T), along with a nonsense variant (p.G267* or p.A123fs*4). Single-cell RNA sequencing of 100-day organoids identified defects in GABAergic interneuron development. We demonstrated aberrant neuronal firing and reduction in size of patient-derived organoids. Mechanistically, we showed that ER homeostasis is perturbed along with an exacerbated unfolded protein response pathway in engineered U87-MG cells and patient-derived organoids expressing UBA5 pathogenic variants. We also assessed two potential therapeutic modalities that augmented UBA5 protein abundance to rescue aberrant molecular and cellular phenotypes. We assessed SINEUP, a long noncoding RNA that augments translation efficiency, and CRISPRa, a modified CRISPR-Cas9 approach to augment transcription efficiency to increase UBA5 protein production. Our study provides a humanized model that allows further investigations of UBA5 variants in the brain and highlights promising approaches to alleviate cellular aberrations for this rare, developmental disorder.
Collapse
Affiliation(s)
- Helen Chen
- Center for Pediatric Neurological Disease Research, Pediatric Translational Neuroscience Initiative, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Christy W LaFlamme
- Center for Pediatric Neurological Disease Research, Pediatric Translational Neuroscience Initiative, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
- St. Jude Graduate School of Biomedical Sciences, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Yong-Dong Wang
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Aidan W Blan
- Center for Pediatric Neurological Disease Research, Pediatric Translational Neuroscience Initiative, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Nikki Koehler
- Center for Pediatric Neurological Disease Research, Pediatric Translational Neuroscience Initiative, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Renata Mendonca Moraes
- Center for Pediatric Neurological Disease Research, Pediatric Translational Neuroscience Initiative, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Athena R Olszewski
- Center for Pediatric Neurological Disease Research, Pediatric Translational Neuroscience Initiative, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
- St. Jude Graduate School of Biomedical Sciences, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Edith P Almanza Fuerte
- Center for Pediatric Neurological Disease Research, Pediatric Translational Neuroscience Initiative, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Emily S Bonkowski
- Center for Pediatric Neurological Disease Research, Pediatric Translational Neuroscience Initiative, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Richa Bajpai
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
- Center for Advanced Genome Engineering, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Alfonso Lavado
- Center for Pediatric Neurological Disease Research, Pediatric Translational Neuroscience Initiative, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Shondra M Pruett-Miller
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
- Center for Advanced Genome Engineering, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Heather C Mefford
- Center for Pediatric Neurological Disease Research, Pediatric Translational Neuroscience Initiative, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
- St. Jude Graduate School of Biomedical Sciences, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| |
Collapse
|
3
|
Zeman M, Stefanik P, Rumanova VS, Okuliarova M. Interactive effects of light at night and high fructose intake on the central circadian clock and endocrine outputs in rats. Mol Cell Endocrinol 2025; 605:112559. [PMID: 40311860 DOI: 10.1016/j.mce.2025.112559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Revised: 04/01/2025] [Accepted: 04/29/2025] [Indexed: 05/03/2025]
Abstract
Light pollution is an increasing global environmental risk factor that contributes to the recent burden of metabolic diseases. The underlying mechanisms are not understood, but disruption of circadian control of physiological and behavioural processes may be involved. The negative consequences of chronodisruption can be augmented by co-exposure to high energy intake. Therefore, we investigated the individual and combined effects of artificial light at night (ALAN) and 10 % fructose in drinking water on the central clock in the suprachiasmatic nuclei (SCN) of the hypothalamus and circadian hormonal outputs in male rats. After 10 weeks of ALAN exposure and high fructose intake, the clockwork in the SCN was attenuated as indicated by eliminated day/night differences in the core clock gene Per1. Additionally, ALAN suppressed the daily variability and fructose induced upregulation of a gamma-aminobutyric acid-synthesising enzyme (GAD65), potentially affecting inhibitory neurotransmission in the SCN. ALAN and fructose additively inhibited plasma melatonin levels revealing excessive fructose intake as a chronodisruptive factor that can be potentiated by ALAN. In contrast to melatonin, daytime plasma testosterone concentrations were increased by high fructose and supressed by ALAN. Furthermore, high fructose intake elevated the plasma levels of two adipokines, leptin and adiponectin, but this response was absent specifically during the daytime in rats exposed to ALAN, indicating that ALAN reduced adipose tissue responsiveness. Our results document the complex consequences of ALAN and high fructose intake on endocrine control mechanisms that can have a long-term negative impact on metabolic health.
Collapse
Affiliation(s)
- Michal Zeman
- Department of Animal Physiology and Ethology, Faculty of Natural Sciences, Comenius University, Bratislava, Slovakia.
| | - Peter Stefanik
- Department of Animal Physiology and Ethology, Faculty of Natural Sciences, Comenius University, Bratislava, Slovakia
| | - Valentina Sophia Rumanova
- Department of Animal Physiology and Ethology, Faculty of Natural Sciences, Comenius University, Bratislava, Slovakia; Hypothalamic Integration Mechanisms, Netherlands Institute for Neuroscience (NIN), An Institute of the Royal Netherlands Academy of Arts and Sciences (KNAW), Amsterdam, the Netherlands; Department of Endocrinology and Metabolism, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands; Laboratory of Endocrinology, Amsterdam University Medical Centers, Amsterdam Gastroenterology Endocrinology Metabolism (AGEM), Amsterdam, the Netherlands
| | - Monika Okuliarova
- Department of Animal Physiology and Ethology, Faculty of Natural Sciences, Comenius University, Bratislava, Slovakia
| |
Collapse
|
4
|
Dow CT. Proposing Bromo-Epi-Androsterone (BEA) for Stiff Person Syndrome (SPS). Microorganisms 2025; 13:824. [PMID: 40284660 PMCID: PMC12029804 DOI: 10.3390/microorganisms13040824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2025] [Revised: 03/23/2025] [Accepted: 03/24/2025] [Indexed: 04/29/2025] Open
Abstract
SPS is characterized by progressive spasmodic muscular rigidity. SPS is thought to be an autoimmune disease with a prominent feature of antibodies against glutamic acid decarboxylase (GAD). GAD is responsible for the enzymatic conversion of glutamic acid (glutamate) into the inhibitory neurotransmitter gamma-aminobutyric acid (GABA). Reduced GABA activity leads to increased excitability in the central nervous system, resulting in muscle rigidity and spasms characteristic of SPS. While SPS is rare, anti-GAD antibodies seen in SPS are also seen in the much more common autoimmune disease, type 1 diabetes (T1D). There is evolving research showing that the anti-GAD antibodies of T1D are produced in response to the presence of mycobacterial heat shock protein 65 (mHSP65), and the mHSP65 is produced in response to an occult infection by a bacterium, Mycobacterium avium subspecies Paratuberculosis (MAP). Humans are broadly exposed to MAP in food, water, and air. There are linear and conformational similarities between the epitopes of GAD and mHSP65. This article proposes that MAP is also an infectious trigger for SPS. Dehydroepiandrosterone (DHEA) is a principal component of the steroid metabolome; it plateaus in young adults and then steadily declines. Bromo-epi-androsterone (BEA) is a potent synthetic analog of DHEA; unlike DHEA, it is non-androgenic, non-anabolic, and an effective modulator of immune dysregulation. BEA is also an anti-infective agent and has been shown to benefit mycobacterial infections, including tuberculosis and leprosy. With the immune stabilizing capacity of BEA as well as its anti-mycobacterial properties, there is reason to believe that a randomized clinical trial with BEA may be beneficial for SPS.
Collapse
Affiliation(s)
- Coad Thomas Dow
- McPherson Eye Research Institute, University of Wisconsin-Madison, Madison, WI 53705, USA;
- Protibea Therapeutics, LLC., Naples, FL 34105, USA
| |
Collapse
|
5
|
Wang B, Fu W, Ueda A, Shah H, Wu CF, Chi W, Zhuang X. Genetic vitamin B6 deficiency and alcohol interaction in behavior and metabolism. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.06.641947. [PMID: 40093095 PMCID: PMC11908246 DOI: 10.1101/2025.03.06.641947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
Alcohol abuse is a leading cause of preventable deaths, affecting brain function and metabolism, including GABA transmission and vitamin B6 (VB6) levels. However, the interaction between genetic VB6 deficiency and alcohol consumption remains unexplored. Here, we utilized Drosophila models with mutations in pyridox(am)ine-5'-phosphate oxidase (PNPO), a key enzyme in VB6 metabolism, to examine this interaction at behavioral and biochemical levels. Our findings demonstrate that PNPO deficiency reduces alcohol aversion, increases consumption, and alters locomotor behavior. Biochemically, PNPO deficiency and alcohol exposure converge on amino acid metabolism, elevating inhibitory neurotransmitters GABA and glycine. Moreover, both PNPO deficiency and alcohol exposure lead to lethality with significant interaction, which can be rescued by VB6 supplementation. These results highlight a functional interaction between genetic VB6 deficiency and alcohol, suggesting potential therapeutic strategies for alcohol-related behaviors.
Collapse
Affiliation(s)
- Benjamin Wang
- Department of Neurobiology, University of Chicago, Chicago, IL 60637
| | - Wenqin Fu
- Department of Neurobiology, University of Chicago, Chicago, IL 60637
| | - Atsushi Ueda
- Department of Biology, College of Liberal Arts and Sciences, University of Iowa, Iowa City, IA 52242
| | - Hardik Shah
- Biological Science Division, Metabolomics Platform, Comprehensive Cancer Center, The University of Chicago, Chicago, IL 60637
| | - Chun-Fang Wu
- Department of Biology, College of Liberal Arts and Sciences, University of Iowa, Iowa City, IA 52242
| | - Wanhao Chi
- Department of Neurobiology, University of Chicago, Chicago, IL 60637
- Present address: The Ken & Ruth Davee Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611
| | - Xiaoxi Zhuang
- Department of Neurobiology, University of Chicago, Chicago, IL 60637
| |
Collapse
|
6
|
Yang Y, Jiang H, Yang W, Wang H, Wang M, Li X, Huang P, Fang S, Hao W, Yang Y, Zhao F, He W. The IVIG treatment response in autoimmune polyendocrine syndromes type 2 with anti-GAD65 antibody-associated stiff person syndrome: a case report and literature review. Front Immunol 2025; 15:1471115. [PMID: 39840065 PMCID: PMC11746041 DOI: 10.3389/fimmu.2024.1471115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Accepted: 12/12/2024] [Indexed: 01/23/2025] Open
Abstract
Autoimmune polyendocrine syndromes (APS) is a rare group of disorders caused by impaired function of multiple endocrine glands due to disruption of immune tolerance. Of which, type 2 (APS-2) is the most common. Glutamic acid decarboxylase (GAD) is the rate-limiting enzyme for the synthesis of gamma-aminobutyric acid (GABA). Anti-GAD antibodies are associated with various neurological disorders, including stiff person syndrome (SPS). SPS is characterized by axial muscle stiffness, rigidity, and intermittent painful muscle spasms, with a prevalence of one to two in a million, making it an extremely rare neurological disorder. The comorbidity of APS-2 with SPS is even rarer. Most practicing neurologists encounter only one or two cases of APS-2 combined with anti-GAD65 antibody-associated SPS in their careers, resulting in underdiagnosis and undertreatment, leading to severe disability and suffering. This case report describes a young male who initially exhibited hair loss, vitiligo, and previously unreported eosinophilia. Before his diagnosis, he was admitted multiple times, with symptoms improving following the addition of intravenous immunoglobulin (IVIG) therapy to a poor treatment regimen. This paper aims to increase physicians' awareness of this condition, enhancing the likelihood of early diagnosis and treatment.
Collapse
Affiliation(s)
- Yulong Yang
- Department of Neurology, The First Affiliated Hospital of Anhui University of Traditional, Chinese Medicine, Hefei, Anhui, China
| | - Hailin Jiang
- Department of Neurology, The First Affiliated Hospital of Anhui University of Traditional, Chinese Medicine, Hefei, Anhui, China
| | - Wenming Yang
- Department of Neurology, The First Affiliated Hospital of Anhui University of Traditional, Chinese Medicine, Hefei, Anhui, China
- Key Laboratory of Xin’An Medicine, Ministry of Education Hefei, Anhui, China
- Center for Xin’an Medicine and Modernization of Traditional Chinese Medicine, Institute of Health and Medicine Hefei Comprehensive National Science Center, Hefei, Anhui, China
| | - Han Wang
- Department of Neurology, The First Affiliated Hospital of Anhui University of Traditional, Chinese Medicine, Hefei, Anhui, China
- Key Laboratory of Xin’An Medicine, Ministry of Education Hefei, Anhui, China
| | - Meixia Wang
- Department of Neurology, The First Affiliated Hospital of Anhui University of Traditional, Chinese Medicine, Hefei, Anhui, China
- Key Laboratory of Xin’An Medicine, Ministry of Education Hefei, Anhui, China
| | - Xiang Li
- Department of Neurology, The First Affiliated Hospital of Anhui University of Traditional, Chinese Medicine, Hefei, Anhui, China
| | - Peng Huang
- Department of Neurology, The First Affiliated Hospital of Anhui University of Traditional, Chinese Medicine, Hefei, Anhui, China
| | - Shuzhen Fang
- Department of Neurology, The First Affiliated Hospital of Anhui University of Traditional, Chinese Medicine, Hefei, Anhui, China
| | - Wenjie Hao
- Department of Neurology, The First Affiliated Hospital of Anhui University of Traditional, Chinese Medicine, Hefei, Anhui, China
| | - Yue Yang
- Department of Neurology, The First Affiliated Hospital of Anhui University of Traditional, Chinese Medicine, Hefei, Anhui, China
| | - Furong Zhao
- Department of Neurology, The First Affiliated Hospital of Anhui University of Traditional, Chinese Medicine, Hefei, Anhui, China
| | - Wei He
- Department of Neurology, The First Affiliated Hospital of Anhui University of Traditional, Chinese Medicine, Hefei, Anhui, China
| |
Collapse
|
7
|
Lü J, Jiang C, Drabick JJ, Joshi M, Perimbeti S. Angelica gigas Nakai (Korean Dang-gui) Root Alcoholic Extracts in Health Promotion and Disease Therapy - active Phytochemicals and In Vivo Molecular Targets. Pharm Res 2025; 42:25-47. [PMID: 39779619 PMCID: PMC11785709 DOI: 10.1007/s11095-024-03809-9] [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: 09/20/2024] [Accepted: 12/17/2024] [Indexed: 01/11/2025]
Abstract
Angelica gigas Nakai (AGN) root is a medicinal herbal widely used in traditional medicine in Korea. AGN root ethanolic extracts have been marketed as dietary supplements in the United States for memory health and pain management. We have recently reviewed the pharmacokinetics (PK) and first-pass hepatic metabolism of ingested AGN supplements in humans for the signature pyranocoumarins decursin (D, Cmax 1x), decursinol angelate (DA, Cmax ~ 10x) and their common botanical precursor and hepatic metabolite decursinol (DOH, Cmax ~ 1000x). Here we update in vivo medicinal activities of AGN and/or its pyranocoumarins and furanocoumarin nodakenin in cancer, pain, memory loss, cerebral ischemia reperfusion stroke, metabolic syndrome and vascular endothelial dysfunctions, anxiety, sleep disorder, epilepsy, inflammatory bowel disease, osteoporosis and osteoarthritis. Given their polypharmacology nature, the pertinent mechanisms of action are likely misrepresented by many cell culture studies that did not consider the drug metabolism knowledge. We report here Rho-associated protein kinases (ROCK1/2) as novel targets for DA and DOH. Combining with published inhibitory activity of DOH on acetylcholinesterase, agonist activity of DOH and antagonist/degrader activity of DA/D on androgen and estrogen receptors, D/DA promoting activity for glutamic acid decarboxylase (GAD)- gamma-aminobutyric acid (GABA) inhibitory axis and inhibition of glutamate dehydrogenase (GDH), monoamine oxidase-A (MAO-A) and transient receptor potential vanilloid 1 (TRPV1), we postulate their contributions to neuro-cognitive, metabolic, oncologic, vascular and other beneficial bioactivities of AGN extracts. A clinical trial is being planned for an AGN extract to manage side effects of androgen deprivation therapy in prostate cancer patients.
Collapse
Affiliation(s)
- Junxuan Lü
- Department of Pharmacology, Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA.
- Penn State Cancer Institute, Pennsylvania State University, Hershey, PA, 17033, USA.
- Center for Cannabis and Natural Product Pharmaceutics, Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA.
| | - Cheng Jiang
- Department of Pharmacology, Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA
- Center for Cannabis and Natural Product Pharmaceutics, Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA
| | - Joseph J Drabick
- Penn State Cancer Institute, Pennsylvania State University, Hershey, PA, 17033, USA
- Department of Medicine Division of Hematology and Oncology, Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA
| | - Monika Joshi
- Penn State Cancer Institute, Pennsylvania State University, Hershey, PA, 17033, USA
- Department of Medicine Division of Hematology and Oncology, Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA
| | - Stuthi Perimbeti
- Penn State Cancer Institute, Pennsylvania State University, Hershey, PA, 17033, USA
- Department of Medicine Division of Hematology and Oncology, Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA
| |
Collapse
|
8
|
Bortolami A, Forzisi Kathera-Ibarra E, Balatsky A, Dubey M, Amin R, Venkateswaran S, Dutto S, Seth I, Ashor A, Nwandiko A, Pan PY, Crockett DP, Sesti F. Abnormal cytoskeletal remodeling but normal neuronal excitability in a mouse model of the recurrent developmental and epileptic encephalopathy-susceptibility KCNB1-p.R312H variant. Commun Biol 2024; 7:1713. [PMID: 39738805 DOI: 10.1038/s42003-024-07344-6] [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: 06/26/2024] [Accepted: 12/02/2024] [Indexed: 01/02/2025] Open
Abstract
Integrin_K+ Channel_Complexes (IKCs), are implicated in neurodevelopment and cause developmental and epileptic encephalopathy (DEE) through mechanisms that were poorly understood. Here, we investigate the function of neocortical IKCs formed by voltage-gated potassium (Kv) channels Kcnb1 and α5β5 integrin dimers in wild-type (WT) and homozygous knock-in (KI) Kcnb1R312H(+/+) mouse model of DEE. Kcnb1R312H(+/+) mice suffer from severe cognitive deficit and compulsive behavior. Their brains show neuronal damage in multiple areas and disrupted corticocortical and corticothalamic connectivity along with aberrant glutamatergic vesicular transport. Surprisingly, the electrical properties of Kcnb1R312H(+/+) pyramidal neurons are similar to those of WT neurons, indicating that the arginine to histidine replacement does not affect the conducting properties of the mutant channel. In contrast, fluorescence recovery after photobleaching, biochemistry, and immunofluorescence, reveal marked differences in the way WT and Kcnb1R312H(+/+) neurons modulate the remodeling of the actin cytoskeleton, a key player in the processes underlying neurodevelopment. Together these results demonstrate that Kv channels can cause multiple conditions, including epileptic seizures, through mechanisms that do not involve their conducting functions and put forward the idea that the etiology of DEE may be primarily non-ionic.
Collapse
Affiliation(s)
- Alessandro Bortolami
- Department of Neuroscience and Cell Biology, Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ, USA
| | - Elena Forzisi Kathera-Ibarra
- Department of Neuroscience and Cell Biology, Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ, USA
| | - Anastasia Balatsky
- Department of Neuroscience and Cell Biology, Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ, USA
| | - Mansi Dubey
- Department of Neuroscience and Cell Biology, Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ, USA
| | - Rusheel Amin
- Department of Neuroscience and Cell Biology, Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ, USA
| | - Srinidi Venkateswaran
- Department of Neuroscience and Cell Biology, Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ, USA
| | - Stefania Dutto
- Department of Neuroscience and Cell Biology, Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ, USA
| | - Ishan Seth
- Department of Neuroscience and Cell Biology, Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ, USA
| | - Adam Ashor
- Department of Neuroscience and Cell Biology, Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ, USA
- Nilo Therapeutics, New York, NY, USA
| | - Angel Nwandiko
- Department of Neuroscience and Cell Biology, Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ, USA
| | - Ping-Yue Pan
- Department of Neuroscience and Cell Biology, Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ, USA
| | - David P Crockett
- Department of Neuroscience and Cell Biology, Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ, USA
| | - Federico Sesti
- Department of Neuroscience and Cell Biology, Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ, USA.
| |
Collapse
|
9
|
Tong J, Gao J, Qi Y, Gao Z, Wang Q, Liu Y, Yuan T, Ren M, Yang G, Li Z, Li J, Sun H, Zhao X, Leung YY, Mu Y, Xu J, Lu C, Peng S, Ge L. GABA AR-PPT1 palmitoylation homeostasis controls synaptic transmission and circuitry oscillation. Transl Psychiatry 2024; 14:488. [PMID: 39695089 DOI: 10.1038/s41398-024-03206-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 12/05/2024] [Accepted: 12/10/2024] [Indexed: 12/20/2024] Open
Abstract
The infantile neuronal ceroid lipofuscinosis, also called CLN1 disease, is a fatal neurodegenerative disease caused by mutations in the CLN1 gene encoding palmitoyl protein thioesterase 1 (PPT1). Identifying the depalmitoylation substrates of PPT1 is crucial for understanding CLN1 disease. In this study, we found that GABAAR, the critical synaptic protein essential for inhibitory neurotransmission, is a substrate of PPT1. PPT1 depalmitoylates GABAAR α1 subunit at Cystein-260, while binding to Cystein-165 and -179. Mutations of PPT1 or its GABAAR α1 subunit binding site enhanced inhibitory synaptic transmission and strengthened oscillations powers but disrupted phase coupling in CA1 region and impaired learning and memory in 1- to 2-months-old PPT1-deficient and Gabra1em1 mice. Our study highlights the critical role of PPT1 in maintaining GABAAR palmitoylation homeostasis and reveals a previously unknown molecular pathway in CLN1 diseases induced by PPT1 mutations.
Collapse
Affiliation(s)
- Jia Tong
- The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, He'nan, China
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, He'nan, China
| | - Jingjing Gao
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, He'nan, China
- School of Pharmacy, Lanzhou University, Lanzhou, China
| | - Yawei Qi
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, He'nan, China
| | - Ziyan Gao
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, He'nan, China
| | - Qianqian Wang
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, He'nan, China
| | - Yang Liu
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, He'nan, China
| | - Tiangang Yuan
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, He'nan, China
| | - Minglong Ren
- Institute of Brain Functional Genomics, East China Normal University, Shanghai, China
| | - Guixia Yang
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, He'nan, China
| | - Zhaoyue Li
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, He'nan, China
| | - Jin Li
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, He'nan, China
| | - Hongyuan Sun
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, He'nan, China
| | - Xing Zhao
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, He'nan, China
| | - Yeung-Yeung Leung
- Division of Brain Sciences, Imperial College Faculty of Medicine, Du Cane Road, London, UK
| | - Yonghui Mu
- Basic Medical College, Xinxiang Medical University, Xinxiang, He'nan, China
| | - Jiamin Xu
- Institute of Brain Functional Genomics, East China Normal University, Shanghai, China
| | - Chengbiao Lu
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, He'nan, China.
- He'nan International Joint Laboratory for Non-invasive Neural Modulation, Department of Physiology and Pathology, School of Basic Medical Science, Xinxiang Medical University, Xinxiang, He'nan, China.
| | - Shiyong Peng
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, He'nan, China.
| | - Lihao Ge
- The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, He'nan, China.
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, He'nan, China.
| |
Collapse
|
10
|
Zhang Q, Zhu L, Li H, Chen Q, Li N, Li J, Zhao Z, Xiao D, Tang T, Bi C, Zhang Y, Zhang H, Zhang G, Li M, Zhu Y, Zhang J, Kong J. Insights and progress on the biosynthesis, metabolism, and physiological functions of gamma-aminobutyric acid (GABA): a review. PeerJ 2024; 12:e18712. [PMID: 39703920 PMCID: PMC11657192 DOI: 10.7717/peerj.18712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Accepted: 11/24/2024] [Indexed: 12/21/2024] Open
Abstract
GABA (γ-aminobutyric acid) is a non-protein amino acid that occurs naturally in the human brain, animals, plants and microorganisms. It is primarily produced by the irreversible action of glutamic acid decarboxylase (GAD) on the α-decarboxylation of L-glutamic acid. As a major neurotransmitter in the brain, GABA plays a crucial role in behavior, cognition, and the body's stress response. GABA is mainly synthesized through the GABA shunt and the polyamine degradation pathways. It works through three receptors (GABAA, GABAB, and GABAC), each exhibiting different pharmacological and physiological characteristics. GABA has a variety of physiological roles and applications. In plants, it regulates growth, development and stress responses. In mammals, it influences physiological functions such as nervous system regulation, blood pressure equilibrium, liver and kidneys enhancement, hormone secretion regulation, immunity enhancement, cancer prevention, as well as anti-aging effects. As a biologically active ingredient, GABA possesses unique physiological effects and medicinal value, leading to its widespread application and substantially increased market demand in the food and pharmaceutical industries. GABA is primarily produced through chemical synthesis, plant enrichment and microbial fermentation. In this review, we first make an overview of GABA, focusing on its synthesis, metabolism, GABA receptors and physiological functions. Next, we describe the industrial production methods of GABA. Finally, we discuss the development of ligands for the GABA receptor binding site, the prospects of GABA production and application, as well as its clinical trials in potential drugs or compounds targeting GABA for the treatment of epilepsy. The purpose of this review is to attract researchers from various fields to focus on GABA research, promote multidisciplinary communications and collaborations, break down disciplinary barriers, stimulate innovative research ideas and methods, and advance the development and application of GABA in medicine, agriculture, food and other fields.
Collapse
Affiliation(s)
- Qingli Zhang
- Department of Medicine, Qingdao Binhai University, Qingdao, Shandong, China
| | - Lei Zhu
- Department of Medicine, Qingdao Binhai University, Qingdao, Shandong, China
| | - Hailong Li
- Department of Medicine, Qingdao Binhai University, Qingdao, Shandong, China
| | - Qu Chen
- Department of Medicine, Qingdao Binhai University, Qingdao, Shandong, China
| | - Nan Li
- Department of Rehabilitation, Qingdao Binhai College Affiliated Hospital, Qingdao, China
| | - Jiansheng Li
- Department of Nephrology, Gansu Provincial Hospital of Traditional Chinese Medicine, Lanzhou, Gansu, China
| | - Zichu Zhao
- Department of Medicine, Qingdao Binhai University, Qingdao, Shandong, China
| | - Di Xiao
- Department of Medicine, Qingdao Binhai University, Qingdao, Shandong, China
| | - Tingting Tang
- Department of Medicine, Qingdao Binhai University, Qingdao, Shandong, China
| | - Chunhua Bi
- Department of Medicine, Qingdao Binhai University, Qingdao, Shandong, China
| | - Yan Zhang
- Department of Medicine, Qingdao Binhai University, Qingdao, Shandong, China
| | - Haili Zhang
- Department of Medicine, Qingdao Binhai University, Qingdao, Shandong, China
| | - Guizhen Zhang
- Department of Medicine, Qingdao Binhai University, Qingdao, Shandong, China
| | - Mingyang Li
- Department of Medicine, Qingdao Binhai University, Qingdao, Shandong, China
| | - Yanli Zhu
- College of Life Sciences and Engineering, Northwest Minzu University, Lanzhou, China
| | - Jingjing Zhang
- Department of Medicine, Qingdao Binhai University, Qingdao, Shandong, China
| | - Jingjing Kong
- Department of Medicine, Qingdao Binhai University, Qingdao, Shandong, China
| |
Collapse
|
11
|
Wen ZH, Chen NF, Cheng HJ, Kuo HM, Chen PY, Feng CW, Yao ZK, Chen WF, Sung CS. Upregulated spinal histone deacetylases induce nociceptive sensitization by inhibiting the GABA system in chronic constriction injury-induced neuropathy in rats. Pain Rep 2024; 9:e1209. [PMID: 39512583 PMCID: PMC11543203 DOI: 10.1097/pr9.0000000000001209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Accepted: 08/29/2024] [Indexed: 11/15/2024] Open
Abstract
Introduction Neuropathic pain (NP) affects countless people worldwide; however, few effective treatments are currently available. Histone deacetylases (HDACs) participate in epigenetic modifications in neuropathy-induced nociceptive sensitization. Gamma-aminobutyric acid (GABA) is a major inhibitory neurotransmitter that can inhibit NP. The present study aimed to examine the role of spinal HDAC and its isoforms in neuropathy. Methods Male Wistar Rat with chronic constriction injury (CCI)-induced peripheral neuropathy and HDAC inhibitor, panobinostat, was administrated intrathecally. We performed quantitative real-time polymerase chain reaction (RT-qPCR), western blot, and immunohistochemical analysis of lumbar spinal cord dorsal horn and nociceptive behaviors (thermal hyperalgesia and mechanical allodynia) measurements. Results Herein, RT-qPCR analysis revealed that spinal hdac3, hdac4, and hdac6 were upregulated in CCI rats. Western blotting and immunofluorescence staining further confirmed that HDAC3, HDAC4, and HDAC6 were significantly upregulated, whereas GABA and its synthesis key enzyme glutamic acid decarboxylase (GAD) 65 were dramatically downregulated. Intrathecal panobinostat attenuated nociceptive behavior and restored the downregulated spinal GAD65 and GABA expression in CCI rats. Conclusions HDAC upregulation might induce nociception through GAD65 and GABA inhibition in CCI-induced neuropathy. These findings strongly suggest that HDACs negatively regulate inhibitory neurotransmitters, constituting a potential therapeutic strategy for an epigenetic approach to manage NP.
Collapse
Affiliation(s)
- Zhi-Hong Wen
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung, Taiwan
- Institute of Biopharmaceutical Sciences, National Sun Yat-sen University, Kaohsiung, Taiwan
| | - Nan-Fu Chen
- Institute of Medical Science and Technology, National Sun Yat-sen University, Kaohsiung, Taiwan
- Division of Neurosurgery, Department of Surgery, Kaohsiung Armed Forces General Hospital, Kaohsiung, Taiwan
| | - Hao-Jung Cheng
- Institute of Biopharmaceutical Sciences, National Sun Yat-sen University, Kaohsiung, Taiwan
| | - Hsiao-Mei Kuo
- Department of Neurosurgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Pei-Yu Chen
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung, Taiwan
| | - Chien-Wei Feng
- Department of Obstetrics and Gynecology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Zhi-Kang Yao
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung, Taiwan
- Department of Orthopedics, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
| | - Wu-Fu Chen
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung, Taiwan
- Department of Neurosurgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Chun-Sung Sung
- Division of Pain Management, Department of Anesthesiology, Taipei Veterans General Hospital, Taipei, Taiwan
- School of Medicine, National Yang-Ming Chiao Tung University, Taipei, Taiwan
| |
Collapse
|
12
|
Mikami A, Huang H, Hyodo A, Horie K, Yasumatsu K, Ninomiya Y, Mitoh Y, Iida S, Yoshida R. The role of GABA in modulation of taste signaling within the taste bud. Pflugers Arch 2024; 476:1761-1775. [PMID: 39210062 PMCID: PMC11461785 DOI: 10.1007/s00424-024-03007-x] [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: 04/24/2024] [Revised: 08/03/2024] [Accepted: 08/07/2024] [Indexed: 09/04/2024]
Abstract
Taste buds contain 2 types of GABA-producing cells: sour-responsive Type III cells and glial-like Type I cells. The physiological role of GABA, released by Type III cells is not fully understood. Here, we investigated the role of GABA released from Type III cells using transgenic mice lacking the expression of GAD67 in taste bud cells (Gad67-cKO mice). Immunohistochemical experiments confirmed the absence of GAD67 in Type III cells of Gad67-cKO mice. Furthermore, no difference was observed in the expression and localization of cell type markers, ectonucleoside triphosphate diphosphohydrolase 2 (ENTPD2), gustducin, and carbonic anhydrase 4 (CA4) in taste buds between wild-type (WT) and Gad67-cKO mice. Short-term lick tests demonstrated that both WT and Gad67-cKO mice exhibited normal licking behaviors to each of the five basic tastants. Gustatory nerve recordings from the chorda tympani nerve demonstrated that both WT and Gad67-cKO mice similarly responded to five basic tastants when they were applied individually. However, gustatory nerve responses to sweet-sour mixtures were significantly smaller than the sum of responses to each tastant in WT mice but not in Gad67-cKO mice. In summary, elimination of GABA signalling by sour-responsive Type III taste cells eliminates the inhibitory cell-cell interactions seen with application of sour-sweet mixtures.
Collapse
Affiliation(s)
- Ayaka Mikami
- Department of Oral Physiology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
- Department of Oral and Maxillofacial Reconstructive Surgery, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Hai Huang
- Department of Oral Physiology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Aiko Hyodo
- Department of Oral Physiology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
- Department of Oral and Maxillofacial Reconstructive Surgery, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Kengo Horie
- Department of Oral Physiology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
- Faculty of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 2-5-1, Shikata-Cho, Kita-Ku, Okayama, 700-8525, Japan
| | | | - Yuzo Ninomiya
- Department of Oral Physiology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
- Graduate School of Dental Science, Kyushu University, Fukuoka, Japan
- Monell Chemical Senses Center, Philadelphia, PA, USA
| | - Yoshihiro Mitoh
- Department of Oral Physiology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
- Faculty of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 2-5-1, Shikata-Cho, Kita-Ku, Okayama, 700-8525, Japan
| | - Seiji Iida
- Department of Oral and Maxillofacial Reconstructive Surgery, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
- Faculty of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 2-5-1, Shikata-Cho, Kita-Ku, Okayama, 700-8525, Japan
| | - Ryusuke Yoshida
- Department of Oral Physiology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan.
- Faculty of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 2-5-1, Shikata-Cho, Kita-Ku, Okayama, 700-8525, Japan.
| |
Collapse
|
13
|
Hanssen KS, Winter-Hjelm N, Niethammer SN, Kobro-Flatmoen A, Witter MP, Sandvig A, Sandvig I. Reverse engineering of feedforward cortical-Hippocampal microcircuits for modelling neural network function and dysfunction. Sci Rep 2024; 14:26021. [PMID: 39472479 PMCID: PMC11522409 DOI: 10.1038/s41598-024-77157-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Accepted: 10/21/2024] [Indexed: 11/02/2024] Open
Abstract
Engineered biological neural networks are indispensable models for investigation of neural function and dysfunction from the subcellular to the network level. Notably, advanced neuroengineering approaches are of significant interest for their potential to replicate the topological and functional organization of brain networks. In this study, we reverse engineered feedforward neural networks of primary cortical and hippocampal neurons, using a custom-designed multinodal microfluidic device with Tesla valve inspired microtunnels. By interfacing this device with nanoporous microelectrodes, we show that the reverse engineered multinodal neural networks exhibit capacity for both segregated and integrated functional activity, mimicking brain network dynamics. To advocate the broader applicability of our model system, we induced localized perturbations with amyloid beta to study the impact of pathology on network functionality. Additionally, we demonstrate long-term culturing of subregion- and layer specific neurons extracted from the entorhinal cortex and hippocampus of adult Alzheimer's-model mice and rats. Our results thus highlight the potential of our approach for reverse engineering of anatomically relevant multinodal neural networks to study dynamic structure-function relationships in both healthy and pathological conditions.
Collapse
Affiliation(s)
- Katrine Sjaastad Hanssen
- Department of Neuromedicine and Movement Science, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology (NTNU), Trondheim, Norway.
- Kavli Institute for Systems Neuroscience, Centre for Neural Computation, Egil and Pauline Braathen and Fred Kavli Centre for Cortical Microcircuits, Norwegian University of Science and Technology (NTNU), Trondheim, Norway.
| | - Nicolai Winter-Hjelm
- Department of Neuromedicine and Movement Science, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology (NTNU), Trondheim, Norway.
| | - Salome Nora Niethammer
- Division of Neuronal Cell Biology, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Asgeir Kobro-Flatmoen
- Kavli Institute for Systems Neuroscience, Centre for Neural Computation, Egil and Pauline Braathen and Fred Kavli Centre for Cortical Microcircuits, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
- K.G. Jebsen Centre for Alzheimer's Disease, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Menno P Witter
- Kavli Institute for Systems Neuroscience, Centre for Neural Computation, Egil and Pauline Braathen and Fred Kavli Centre for Cortical Microcircuits, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
- K.G. Jebsen Centre for Alzheimer's Disease, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Axel Sandvig
- Department of Neuromedicine and Movement Science, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
- Department of Neurology and Clinical Neurophysiology, St Olav's University Hospital, Trondheim, Norway
| | - Ioanna Sandvig
- Department of Neuromedicine and Movement Science, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology (NTNU), Trondheim, Norway.
| |
Collapse
|
14
|
Habela CW, Liu S, Taga A, Dastgheyb R, Haughey N, Bergles D, Song H, Ming GL, Maragakis NJ. Altered development and network connectivity in a human neuronal model of 15q11.2 deletion-related neurodevelopmental disorders. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.19.613912. [PMID: 39345567 PMCID: PMC11429947 DOI: 10.1101/2024.09.19.613912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
The chromosome 15q11.2 locus is deleted in 1.5% of patients with genetic epilepsy and confers a risk for intellectual disability and schizophrenia. Individuals with this deletion demonstrate increased cortical thickness, decreased cortical surface area and white matter abnormalities. Human induced pluripotent stem cell (iPSC)-derived neural progenitor cells (NPC) from 15q11.2 deletion individuals exhibit early adhesion junction and migration abnormalities, but later neuronal development and function have not been fully assessed. Imaging studies indicating altered structure and network connectivity in the anterior brain regions and the cingulum suggest that in addition to alterations in progenitor dynamics, there may also be structural and functional changes within discrete networks of mature neurons. To explore this, we generated human forebrain cortical neurons from iPSCs derived from individuals with or without 15q11.2 deletion and used longitudinal imaging and multielectrode array analysis to evaluate neuronal development over time. 15q11.2 deleted neurons exhibited fewer connections and an increase in inhibitory neurons. Individual neurons had decreased neurite complexity and overall decreased neurite length. These structural changes were associated with a reduction in multiunit action potential generation, bursting and synchronization. The 15q11.2 deleted neurons also demonstrated specific functional deficits in glutamate and GABA mediated network activity and synchronization with a delay in the maturation of the inhibitory response to GABA. These data indicate that deletion of the 15q11.2 region is sufficient to impair the structural and functional maturation of cortical neuron networks which likely underlies the pathologic changes in humans with the 15q11.2 deletion.
Collapse
|
15
|
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
|
16
|
Colson C, Wang Y, Atherton J, Su X. SLC45A4 encodes a mitochondrial putrescine transporter that promotes GABA de novo synthesis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.23.604788. [PMID: 39091866 PMCID: PMC11291067 DOI: 10.1101/2024.07.23.604788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/04/2024]
Abstract
Solute carriers (SLC) are membrane proteins that facilitate the transportation of ions and metabolites across either the plasma membrane or the membrane of intracellular organelles. With more than 450 human genes annotated as SLCs, many of them are still orphan transporters without known biochemical functions. We developed a metabolomic-transcriptomic association analysis, and we found that the expression of SLC45A4 has a strong positive correlation with the cellular level of γ-aminobutyric acid (GABA). Using mass spectrometry and the stable isotope tracing approach, we demonstrated that SLC45A4 promotes GABA de novo synthesis through the Arginine/Ornithine/Putrescine (AOP) pathway. SLC45A4 functions as a putrescine transporter localized to the mitochondrial membrane to facilitate GABA production. Taken together, our results revealed a new biochemical mechanism where SLC45A4 controls GABA production.
Collapse
Affiliation(s)
- Cecilia Colson
- Department of Medicine, Rutgers-Robert Wood Johnson Medical School, New Brunswick, NJ 08901, USA
| | - Yujue Wang
- Department of Medicine, Rutgers-Robert Wood Johnson Medical School, New Brunswick, NJ 08901, USA
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ 08903, USA
| | - James Atherton
- Department of Medicine, Rutgers-Robert Wood Johnson Medical School, New Brunswick, NJ 08901, USA
| | - Xiaoyang Su
- Department of Medicine, Rutgers-Robert Wood Johnson Medical School, New Brunswick, NJ 08901, USA
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ 08903, USA
| |
Collapse
|
17
|
Lennartz M, Benjamin Dünnebier N, Höflmayer D, Dwertmann Rico S, Kind S, Reiswich V, Viehweger F, Lutz F, Fraune C, Gorbokon N, Luebke AM, Hube-Magg C, Büscheck F, Menz A, Uhlig R, Krech T, Hinsch A, Burandt E, Sauter G, Simon R, Kluth M, Steurer S, Marx AH, Lebok P, Dum D, Minner S, Jacobsen F, Clauditz TS, Bernreuther C. GAD2 Is a Highly Specific Marker for Neuroendocrine Neoplasms of the Pancreas. Am J Surg Pathol 2024; 48:377-386. [PMID: 38271200 PMCID: PMC10930383 DOI: 10.1097/pas.0000000000002186] [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: 01/27/2024]
Abstract
Glutamate decarboxylase 2 (GAD2) is the most important inhibitory neurotransmitter and plays a role in insulin-producing β cells of pancreatic islets. The limitation of GAD2 expression to a few normal cell types makes GAD2 a potential immunohistochemical diagnostic marker. To evaluate the diagnostic utility of GAD2 immunohistochemistry, a tissue microarray containing 19,202 samples from 152 different tumor entities and 608 samples of 76 different normal tissue types was analyzed. In normal tissues, GAD2 staining was restricted to brain and pancreatic islet cells. GAD2 staining was seen in 20 (13.2%) of 152 tumor categories, including 5 (3.3%) tumor categories containing at least 1 strongly positive case. GAD2 immunostaining was most commonly seen in neuroendocrine carcinomas (58.3%) and neuroendocrine tumors (63.2%) of the pancreas, followed by granular cell tumors (37.0%) and neuroendocrine tumors of the lung (11.1%). GAD2 was only occasionally (<10% of cases) seen in 16 other tumor entities including paraganglioma, medullary thyroid carcinoma, and small cell neuroendocrine carcinoma of the urinary bladder. Data on GAD2 and progesterone receptor (PR) expression (from a previous study) were available for 95 pancreatic and 380 extrapancreatic neuroendocrine neoplasms. For determining a pancreatic origin of a neuroendocrine neoplasm, the sensitivity of GAD2 was 64.2% and specificity 96.3%, while the sensitivity of PR was 56.8% and specificity 92.6%. The combination of PR and GAD2 increased both sensitivity and specificity. GAD2 immunohistochemistry is a highly useful diagnostic tool for the identification of pancreatic origin in case of neuroendocrine neoplasms with unknown site of origin.
Collapse
Affiliation(s)
- Maximilian Lennartz
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg
| | | | - Doris Höflmayer
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg
| | | | - Simon Kind
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg
| | - Viktor Reiswich
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg
| | - Florian Viehweger
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg
| | - Florian Lutz
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg
| | - Christoph Fraune
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg
| | - Natalia Gorbokon
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg
| | - Andreas M. Luebke
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg
| | - Claudia Hube-Magg
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg
| | - Franziska Büscheck
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg
| | - Anne Menz
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg
| | - Ria Uhlig
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg
| | - Till Krech
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg
- Institute of Pathology, Clinical Center Osnabrueck, Osnabrueck
| | - Andrea Hinsch
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg
| | - Eike Burandt
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg
| | - Guido Sauter
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg
| | - Ronald Simon
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg
| | - Martina Kluth
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg
| | - Stefan Steurer
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg
| | - Andreas H. Marx
- Department of Pathology, Academic Hospital Fuerth, Fuerth Germany
| | - Patrick Lebok
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg
- Institute of Pathology, Clinical Center Osnabrueck, Osnabrueck
| | - David Dum
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg
| | - Sarah Minner
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg
| | - Frank Jacobsen
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg
| | - Till S. Clauditz
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg
| | | |
Collapse
|
18
|
Cieslik-Starkiewicz A, Noworyta K, Solich J, Korlatowicz A, Faron-Górecka A, Rygula R. Identification of genes regulated by trait sensitivity to negative feedback and prolonged alcohol consumption in rats. Pharmacol Rep 2024; 76:207-215. [PMID: 38172401 PMCID: PMC10830829 DOI: 10.1007/s43440-023-00563-4] [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: 09/10/2023] [Revised: 11/24/2023] [Accepted: 11/27/2023] [Indexed: 01/05/2024]
Abstract
BACKGROUND The results of our previous studies demonstrated that low sensitivity to negative feedback (NF) is associated with increased vulnerability to the development of compulsive alcohol-seeking in rats. In the present study, we investigated the molecular underpinnings of this relationship. METHODS Using TaqMan Gene Expression Array Cards, we analyzed the expression of the genes related to NF sensitivity and alcohol metabolism in three cortical regions (medial prefrontal cortex [mPFC], anterior cingulate cortex [ACC], orbitofrontal cortex [OFC]) and two subcortical regions (nucleus accumbens [Nacc], amygdala [Amy]). Gene expression differences were confirmed at the protein level with Western blot. RESULTS Sensitivity to NF was characterized by differences in Gad2, Drd2, and Slc6a4 expression in the ACC, Maoa in the mPFC, and Gria1, Htr3a, and Maoa in the OFC. Chronic alcohol consumption was associated with differences in the expression of Comt and Maoa in the ACC, Comt, Adh1, and Htr2b in the mPFC, Adh1, and Slc6a4 in the Nacc, Gad2, and Htr1a in the OFC, and Drd2 in the Amy. Interactions between the sensitivity to NF and alcohol consumption were observed in the expression of Gabra1, Gabbr2, Grin2a, Grin2b, and Grm3 in the ACC, and Grin2a in the OFC. The observed differences were confirmed at the protein level for MAO-A in the mPFC, and ADH1 in the mPFC and Nacc. CONCLUSIONS Our findings contribute to a better understanding of the molecular mechanisms underlying the relationship between trait sensitivity to NF and compulsive alcohol consumption.
Collapse
Affiliation(s)
- Agata Cieslik-Starkiewicz
- Affective Cognitive Neuroscience Laboratory, Department of Pharmacology, Maj Institute of Pharmacology Polish Academy of Sciences, 12 Smętna Street, 31-343, Kraków, Poland
| | - Karolina Noworyta
- Affective Cognitive Neuroscience Laboratory, Department of Pharmacology, Maj Institute of Pharmacology Polish Academy of Sciences, 12 Smętna Street, 31-343, Kraków, Poland
| | - Joanna Solich
- Biochemical Pharmacology Laboratory, Department of Pharmacology, Maj Institute of Pharmacology Polish Academy of Sciences, 12 Smętna Street, 31-343, Kraków, Poland
| | - Agata Korlatowicz
- Biochemical Pharmacology Laboratory, Department of Pharmacology, Maj Institute of Pharmacology Polish Academy of Sciences, 12 Smętna Street, 31-343, Kraków, Poland
| | - Agata Faron-Górecka
- Biochemical Pharmacology Laboratory, Department of Pharmacology, Maj Institute of Pharmacology Polish Academy of Sciences, 12 Smętna Street, 31-343, Kraków, Poland
| | - Rafal Rygula
- Affective Cognitive Neuroscience Laboratory, Department of Pharmacology, Maj Institute of Pharmacology Polish Academy of Sciences, 12 Smętna Street, 31-343, Kraków, Poland.
| |
Collapse
|
19
|
Chen H, Wang YD, Blan AW, Almanza-Fuerte EP, Bonkowski ES, Bajpai R, Pruett-Miller SM, Mefford HC. Patient derived model of UBA5-associated encephalopathy identifies defects in neurodevelopment and highlights potential therapies. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.25.577254. [PMID: 38328212 PMCID: PMC10849720 DOI: 10.1101/2024.01.25.577254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/09/2024]
Abstract
UBA5 encodes for the E1 enzyme of the UFMylation cascade, which plays an essential role in ER homeostasis. The clinical phenotypes of UBA5-associated encephalopathy include developmental delays, epilepsy and intellectual disability. To date, there is no humanized neuronal model to study the cellular and molecular consequences of UBA5 pathogenic variants. We developed and characterized patient-derived cortical organoid cultures and identified defects in GABAergic interneuron development. We demonstrated aberrant neuronal firing and microcephaly phenotypes in patient-derived organoids. Mechanistically, we show that ER homeostasis is perturbed along with exacerbated unfolded protein response pathway in cells and organoids expressing UBA5 pathogenic variants. We also assessed two gene expression modalities that augmented UBA5 expression to rescue aberrant molecular and cellular phenotypes. Our study provides a novel humanized model that allows further investigations of UBA5 variants in the brain and highlights novel systemic approaches to alleviate cellular aberrations for this rare, developmental disorder.
Collapse
Affiliation(s)
- Helen Chen
- Center for Pediatric Neurological Disease Research, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Yong-Dong Wang
- Department of Cell and Molecular Biology, St. Jude Children’s Research Hospital, Memphis TN, USA
| | - Aidan W. Blan
- Center for Pediatric Neurological Disease Research, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Edith P. Almanza-Fuerte
- Center for Pediatric Neurological Disease Research, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Emily S. Bonkowski
- Center for Pediatric Neurological Disease Research, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Richa Bajpai
- Department of Cell and Molecular Biology, St. Jude Children’s Research Hospital, Memphis TN, USA
- Center for Advanced Genome Engineering, St. Jude Children’s Research Hospital, Memphis TN, USA
| | - Shondra M. Pruett-Miller
- Department of Cell and Molecular Biology, St. Jude Children’s Research Hospital, Memphis TN, USA
- Center for Advanced Genome Engineering, St. Jude Children’s Research Hospital, Memphis TN, USA
| | - Heather C. Mefford
- Center for Pediatric Neurological Disease Research, St. Jude Children’s Research Hospital, Memphis, TN, USA
| |
Collapse
|
20
|
Chen Y, Ji X, Bao Z. Identification of the Shared Gene Signatures Between Alzheimer's Disease and Diabetes-Associated Cognitive Dysfunction by Bioinformatics Analysis Combined with Biological Experiment. J Alzheimers Dis 2024; 101:611-625. [PMID: 39213070 PMCID: PMC11492114 DOI: 10.3233/jad-240353] [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] [Accepted: 07/03/2024] [Indexed: 09/04/2024]
Abstract
Background The connection between diabetes-associated cognitive dysfunction (DACD) and Alzheimer's disease (AD) has been shown in several observational studies. However, it remains controversial as to how the two related. Objective To explore shared genes and pathways between DACD and AD using bioinformatics analysis combined with biological experiment. Methods We analyzed GEO microarray data to identify DEGs in AD and type 2 diabetes mellitus (T2DM) induced-DACD datasets. Weighted gene co-expression network analysis was used to find modules, while R packages identified overlapping genes. A robust protein-protein interaction network was constructed, and hub genes were identified with Gene ontology enrichment and Kyoto Encyclopedia of Genome and Genome pathway analyses. HT22 cells were cultured under high glucose and amyloid-β 25-35 (Aβ25-35) conditions to establish DACD and AD models. Quantitative polymerase chain reaction with reverse transcription verification analysis was then performed on intersection genes. Results Three modules each in AD and T2DM induced-DACD were identified as the most relevant and 10 hub genes were screened, with analysis revealing enrichment in pathways such as synaptic vesicle cycle and GABAergic synapse. Through biological experimentation verification, 6 key genes were identified. Conclusions This study is the first to use bioinformatics tools to uncover the genetic link between AD and DACD. GAD1, UCHL1, GAP43, CARNS1, TAGLN3, and SH3GL2 were identified as key genes connecting AD and DACD. These findings offer new insights into the diseases' pathogenesis and potential diagnostic and therapeutic targets.
Collapse
Affiliation(s)
- Yixin Chen
- Department of Gerontology, Huadong Hospital Affiliated to Fudan University, Shanghai, China
- Research Center on Aging and Medicine, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Clinical Geriatric Medicine, Shanghai, China, Fudan University, Shanghai, China
| | - Xueying Ji
- Department of General Practice, Huadong Hospital Affiliated to Fudan University, Shanghai, China
| | - Zhijun Bao
- Department of Gerontology, Huadong Hospital Affiliated to Fudan University, Shanghai, China
- Research Center on Aging and Medicine, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Clinical Geriatric Medicine, Shanghai, China, Fudan University, Shanghai, China
| |
Collapse
|
21
|
Majumdar M, Badwaik H. Trends on Novel Targets and Nanotechnology-Based Drug Delivery System in the Treatment of Parkinson's disease: Recent Advancement in Drug Development. Curr Drug Targets 2024; 25:987-1011. [PMID: 39313872 DOI: 10.2174/0113894501312703240826070530] [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: 04/03/2024] [Revised: 05/29/2024] [Accepted: 07/24/2024] [Indexed: 09/25/2024]
Abstract
Parkinson's disease (PD) is a progressive neurodegenerative disorder that impacts a significant portion of the population. Despite extensive research, an effective cure for PD remains elusive, and conventional pharmacological treatments often face limitations in efficacy and management of symptoms. There has been a lot of discussion about using nanotechnology to increase the bioavailability of small- molecule drugs to target cells in recent years. It is possible that PD treatment might become far more effective and have fewer side effects if medication delivery mechanisms were to be improved. Potential alternatives to pharmacological therapy for molecular imaging and treatment of PD may lie in abnormal proteins such as parkin, α-synuclein, leucine-rich repeat serine and threonine protein kinase 2. Published research has demonstrated encouraging outcomes when nanomedicine-based approaches are used to address the challenges of PD therapy. So, to address the present difficulties of antiparkinsonian treatment, this review outlines the key issues and limitations of antiparkinsonian medications, new therapeutic strategies, and the breadth of delivery based on nanomedicine. This review covers a wide range of subjects, including drug distribution in the brain, the efficacy of drug-loaded nano-carriers in crossing the blood-brain barrier, and their release profiles. In PD, the nano-carriers are also used. Novel techniques of pharmaceutical delivery are currently made possible by vesicular carriers, which eliminate the requirement to cross the blood-brain barrier (BBB).
Collapse
Affiliation(s)
- Manisha Majumdar
- Department of Pharmacy, Shri Shankaracharya Professional University, Bhilai, Chhattisgarh, India
| | - Hemant Badwaik
- Department of Pharmacy, Shri Shankaracharya Professional University, Bhilai, Chhattisgarh, India
| |
Collapse
|
22
|
Stanisavljević Ilić A, Đorđević S, Inta D, Borgwardt S, Filipović D. Olanzapine Effects on Parvalbumin/GAD67 Cell Numbers in Layers/Subregions of Dorsal Hippocampus of Chronically Socially Isolated Rats. Int J Mol Sci 2023; 24:17181. [PMID: 38139008 PMCID: PMC10743576 DOI: 10.3390/ijms242417181] [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: 09/29/2023] [Revised: 11/08/2023] [Accepted: 11/17/2023] [Indexed: 12/24/2023] Open
Abstract
Depression is linked to changes in GABAergic inhibitory neurons, especially parvalbumin (PV) interneurons, which are susceptible to redox dysregulation. Olanzapine (Olz) is an atypical antipsychotic whose mode of action remains unclear. We determined the effect of Olz on PV-positive (+) and glutamate decarboxylase 67 (GAD67) + cell numbers in the layers of dorsal hippocampus (dHIPP) cornu ammonis (CA1-CA3) and dentate gyrus (DG) subregions in rats exposed to chronic social isolation (CSIS), which is an animal model of depression. Antioxidative enzymes and proinflammatory cytokine levels were also examined. CSIS decreased the PV+ cell numbers in the Stratum Oriens (SO) and Stratum Pyramidale (SP) of dCA1 and dDG. It increased interleukin-6 (IL-6), suppressor of cytokine signaling 3 (SOCS3), and copper-zinc superoxide dismutase (CuZnSOD) levels, and it decreased catalase (CAT) protein levels. Olz in CSIS increased the number of GAD67+ cells in the SO and SP layers of dCA1 with no effect on PV+ cells. It reduced the PV+ and GAD67+ cell numbers in the Stratum Radiatum of dCA3 in CSIS. Olz antagonizes the CSIS-induced increase in CuZnSOD, CAT and SOCS3 protein levels with no effect on IL-6. Data suggest that the protective Olz effects in CSIS may be mediated by altering the number of PV+ and GAD67+ cells in dHIPP subregional layers.
Collapse
Affiliation(s)
- Andrijana Stanisavljević Ilić
- Department of Molecular Biology and Endocrinology, “VINČA” Institute of Nuclear Sciences, National Institute of the Republic of Serbia, University of Belgrade, 11000 Belgrade, Serbia;
| | - Snežana Đorđević
- Poisoning Control Centre, Military Medical Academy, 11000 Belgrade, Serbia;
| | - Dragoš Inta
- Department for Community Health Faculty of Natural Sciences, Medicine, University of Fribourg, 1700 Fribourg, Switzerland;
- Department of Biomedicine, University of Basel, 4001 Basel, Switzerland
| | - Stefan Borgwardt
- Department of Psychiatry and Psychotherapy, Center of Brain Behavior and Metabolism, University of Lübeck, 23562 Lübeck, Germany;
| | - Dragana Filipović
- Department of Molecular Biology and Endocrinology, “VINČA” Institute of Nuclear Sciences, National Institute of the Republic of Serbia, University of Belgrade, 11000 Belgrade, Serbia;
| |
Collapse
|
23
|
Pandey K, Bessières B, Sheng SL, Taranda J, Osten P, Sandovici I, Constancia M, Alberini CM. Neuronal activity drives IGF2 expression from pericytes to form long-term memory. Neuron 2023; 111:3819-3836.e8. [PMID: 37788670 PMCID: PMC10843759 DOI: 10.1016/j.neuron.2023.08.030] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 08/03/2023] [Accepted: 08/31/2023] [Indexed: 10/05/2023]
Abstract
Investigations of memory mechanisms have been, thus far, neuron centric, despite the brain comprising diverse cell types. Using rats and mice, we assessed the cell-type-specific contribution of hippocampal insulin-like growth factor 2 (IGF2), a polypeptide regulated by learning and required for long-term memory formation. The highest level of hippocampal IGF2 was detected in pericytes, the multi-functional mural cells of the microvessels that regulate blood flow, vessel formation, the blood-brain barrier, and immune cell entry into the central nervous system. Learning significantly increased pericytic Igf2 expression in the hippocampus, particularly in the highly vascularized stratum lacunosum moleculare and stratum moleculare layers of the dentate gyrus. Igf2 increases required neuronal activity. Regulated hippocampal Igf2 knockout in pericytes, but not in fibroblasts or neurons, impaired long-term memories and blunted the learning-dependent increase of neuronal immediate early genes (IEGs). Thus, neuronal activity-driven signaling from pericytes to neurons via IGF2 is essential for long-term memory.
Collapse
Affiliation(s)
- Kiran Pandey
- Center for Neural Science, New York University, New York, NY 10003, USA
| | | | - Susan L Sheng
- Center for Neural Science, New York University, New York, NY 10003, USA
| | - Julian Taranda
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Pavel Osten
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Ionel Sandovici
- University of Cambridge Metabolic Research Laboratories and MRC Metabolic Diseases Unit, Institute of Metabolic Science, Addenbrookes Hospital, Cambridge, UK; Department of Obstetrics and Gynaecology and National Institute for Health Research Cambridge Biomedical Research Centre, Cambridge, UK; Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Miguel Constancia
- University of Cambridge Metabolic Research Laboratories and MRC Metabolic Diseases Unit, Institute of Metabolic Science, Addenbrookes Hospital, Cambridge, UK; Department of Obstetrics and Gynaecology and National Institute for Health Research Cambridge Biomedical Research Centre, Cambridge, UK; Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | | |
Collapse
|
24
|
Naumova AA, Oleynik EA, Grigorieva YS, Nikolaeva SD, Chernigovskaya EV, Glazova MV. In search of stress: analysis of stress-related markers in mice after hindlimb unloading and social isolation. Neurol Res 2023; 45:957-968. [PMID: 37642364 DOI: 10.1080/01616412.2023.2252280] [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: 03/14/2023] [Accepted: 08/20/2023] [Indexed: 08/31/2023]
Abstract
OBJECTIVES Hindlimb unloading (HU), widely used to simulate microgravity effects, is known to induce a stress response. However, as single-housed animals are usually used in such experiments, social isolation (SI) stress can affect experimental results. In the present study, we aimed to delineate stressful effects of 3-day HU and SI in mice. METHODS Three animal groups, HU, SI, and group-housed (GH) control mice, were recruited. A comprehensive analysis of stress-related markers was performed using ELISA, western blotting, and immunohistochemistry. RESULTS Our results showed that blood corticosterone and activity of glucocorticoid receptors and cAMP response element-binding protein (CREB) in the hippocampus of SI and HU animals did not differ from GH control. However, SI mice demonstrated upregulation of the hippocampal corticotropin-releasing hormone (CRH), inducible NO synthase (iNOS), vesicular glutamate transporter 1 (VGLUT1), and glutamate decarboxylases 65/67 (GAD65/67) along with activation of Fos-related antigen 1 (Fra-1) in the amygdala confirming the expression of stress. In HU mice, the same increase in GAD65/67 and Fra-1 indicated the contribution of SI. The special HU effect was expressed only in neurogenesis attenuation. DISCUSSION Thus, our data indicated that 3-day HU could not be characterized as physiological stress, but SI stress contributed to the negative effects of HU.
Collapse
Affiliation(s)
- Alexandra A Naumova
- Sechenov Institute of Evolutionary Physiology and Biochemistry, The Russian Academy of Sciences, St. Petersburg, Russia
| | - Ekaterina A Oleynik
- Sechenov Institute of Evolutionary Physiology and Biochemistry, The Russian Academy of Sciences, St. Petersburg, Russia
- Institute of Lightweight Design and Structural Biomechanics, TU Wien, Vienna, Austria
| | - Yulia S Grigorieva
- Sechenov Institute of Evolutionary Physiology and Biochemistry, The Russian Academy of Sciences, St. Petersburg, Russia
| | - Svetlana D Nikolaeva
- Sechenov Institute of Evolutionary Physiology and Biochemistry, The Russian Academy of Sciences, St. Petersburg, Russia
| | - Elena V Chernigovskaya
- Sechenov Institute of Evolutionary Physiology and Biochemistry, The Russian Academy of Sciences, St. Petersburg, Russia
| | - Margarita V Glazova
- Sechenov Institute of Evolutionary Physiology and Biochemistry, The Russian Academy of Sciences, St. Petersburg, Russia
| |
Collapse
|
25
|
Heneberg P. Diabetes in stiff-person syndrome. Trends Endocrinol Metab 2023; 34:640-651. [PMID: 37586963 DOI: 10.1016/j.tem.2023.07.005] [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: 02/28/2023] [Revised: 07/08/2023] [Accepted: 07/24/2023] [Indexed: 08/18/2023]
Abstract
Anti-glutamic acid decarboxylase (GAD) autoantibodies are a hallmark of stiff-person syndrome (SPS) and insulin-dependent diabetes mellitus (IDDM). However, patients with concurrent IDDM and SPS often manifest insulin resistance, and SPS-associated IDDM probably has heterogeneous causes. Some patients manifest IDDM associated only with high titers of anti-GAD65 caused by SPS. By contrast, other patients develop IDDM only after being treated with high-dose corticosteroids or they progress to insulin dependency following their treatment with high-dose corticosteroids. The profile of autoantibodies differs markedly between type 1 diabetes mellitus (T1DM), late-onset diabetes mellitus, and SPS-associated IDDM. Therefore, as with new-onset diabetes after transplantation (NODAT), SPS-associated IDDM should be classified as a specific diabetes entity, the pathophysiology of which requires increased attention.
Collapse
Affiliation(s)
- Petr Heneberg
- Charles University, Third Faculty of Medicine, Prague, Czech Republic.
| |
Collapse
|
26
|
Smith SK, Frazel PW, Khodadadi-Jamayran A, Zappile P, Marier C, Okhovat M, Brown S, Long MA, Heguy A, Phelps SM. De novo assembly and annotation of the singing mouse genome. BMC Genomics 2023; 24:569. [PMID: 37749493 PMCID: PMC10521431 DOI: 10.1186/s12864-023-09678-7] [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: 05/24/2023] [Accepted: 09/14/2023] [Indexed: 09/27/2023] Open
Abstract
BACKGROUND Developing genomic resources for a diverse range of species is an important step towards understanding the mechanisms underlying complex traits. Specifically, organisms that exhibit unique and accessible phenotypes-of-interest allow researchers to address questions that may be ill-suited to traditional model organisms. We sequenced the genome and transcriptome of Alston's singing mouse (Scotinomys teguina), an emerging model for social cognition and vocal communication. In addition to producing advertisement songs used for mate attraction and male-male competition, these rodents are diurnal, live at high-altitudes, and are obligate insectivores, providing opportunities to explore diverse physiological, ecological, and evolutionary questions. RESULTS Using PromethION, Illumina, and PacBio sequencing, we produced an annotated genome and transcriptome, which were validated using gene expression and functional enrichment analyses. To assess the usefulness of our assemblies, we performed single nuclei sequencing on cells of the orofacial motor cortex, a brain region implicated in song coordination, identifying 12 cell types. CONCLUSIONS These resources will provide the opportunity to identify the molecular basis of complex traits in singing mice as well as to contribute data that can be used for large-scale comparative analyses.
Collapse
Affiliation(s)
- Samantha K Smith
- Department of Integrative Biology, University of Texas at Austin, Austin, TX, 78712, USA.
| | - Paul W Frazel
- Department of Neuroscience and Physiology, New York University Grossman School of Medicine, New York, NY, 10016, USA
| | - Alireza Khodadadi-Jamayran
- Applied Bioinformatics Laboratory, New York University Grossman School of Medicine, New York, NY, 10016, USA
| | - Paul Zappile
- Genome Technology Center, New York University Grossman School of Medicine, New York, NY, 10016, USA
| | - Christian Marier
- Genome Technology Center, New York University Grossman School of Medicine, New York, NY, 10016, USA
| | - Mariam Okhovat
- Department of Integrative Biology, University of Texas at Austin, Austin, TX, 78712, USA
- Present Address: Oregon Health & Science University, Portland, OR, USA
| | - Stuart Brown
- NYU Center for Health Informatics and Bioinformatics, New York University Grossman School of Medicine, New York, NY, 10016, USA
- Present Address: Exxon Mobil Corporate, Houston, TX, USA
| | - Michael A Long
- Department of Neuroscience and Physiology, New York University Grossman School of Medicine, New York, NY, 10016, USA
| | - Adriana Heguy
- Genome Technology Center, New York University Grossman School of Medicine, New York, NY, 10016, USA
| | - Steven M Phelps
- Department of Integrative Biology, University of Texas at Austin, Austin, TX, 78712, USA
| |
Collapse
|
27
|
Chang Z, Skach J, Kam K. Inhibitory subpopulations in preBötzinger Complex play distinct roles in modulating inspiratory rhythm and pattern. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.07.552303. [PMID: 37609332 PMCID: PMC10441369 DOI: 10.1101/2023.08.07.552303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/24/2023]
Abstract
Inhibitory neurons embedded within mammalian neural circuits shape breathing, walking, chewing, and other rhythmic motor behaviors. At the core of the neural circuit controlling breathing is the preBötzinger Complex (preBötC), a nucleus in the ventrolateral medulla necessary for generation of inspiratory rhythm. In the preBötC, a recurrently connected network of glutamatergic Dbx1-derived (Dbx1 + ) neurons generates rhythmic inspiratory drive. Functionally and anatomically intercalated among Dbx1 + preBötC neurons are GABAergic (GAD1/2 + ) and glycinergic (GlyT2 + ) neurons, whose roles in breathing remain unclear. To elucidate the inhibitory microcircuits within preBötC, we first characterized the spatial distribution of molecularly-defined inhibitory preBötC subpopulations in double reporter mice expressing either the red fluorescent protein tdTomato or EGFP in GlyT2 + , GAD1 + , or GAD2 + neurons. We found that, in postnatal mice, the majority of inhibitory preBötC neurons expressed a combination of GlyT2 and GAD2 while a much smaller subpopulation also expressed GAD1. To determine the functional role of these subpopulations, we used holographic photostimulation, a patterned illumination technique with high spatiotemporal resolution, in rhythmically active medullary slices from neonatal Dbx1 tdTomato ;GlyT2 EGFP and Dbx1 tdTomato ;GAD1 EGFP double reporter mice. Stimulation of 4 or 8 preBötC GlyT2 + neurons during endogenous rhythm prolonged the interburst interval in a phase-dependent manner and increased the latency to burst initiation when bursts were evoked by stimulation of Dbx1 + neurons. In contrast, stimulation of 4 or 8 preBötC GAD1 + neurons did not affect interburst interval or latency to burst initiation. Instead, photoactivation of GAD1 + neurons during the inspiratory burst prolonged endogenous and evoked burst duration and decreased evoked burst amplitude. We conclude that the majority of preBötC inhibitory neurons express both GlyT2 and GAD2 and modulate breathing rhythm by delaying burst initiation while a smaller GAD1 + subpopulation shapes inspiratory patterning by altering burst duration and amplitude.
Collapse
|
28
|
García E. Two putative glutamate decarboxylases of Streptococcus pneumoniae as possible antigens for the production of anti-GAD65 antibodies leading to type 1 diabetes mellitus. Int Microbiol 2023; 26:675-690. [PMID: 37154976 PMCID: PMC10165594 DOI: 10.1007/s10123-023-00364-y] [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: 01/11/2023] [Revised: 04/13/2023] [Accepted: 04/21/2023] [Indexed: 05/10/2023]
Abstract
Type 1 diabetes mellitus (T1DM) has been increasing in prevalence in the last decades and has become a global burden. Autoantibodies against human glutamate decarboxylase (GAD65) are among the first to be detected at the onset of T1DM. Diverse viruses have been proposed to be involved in the triggering of T1DM because of molecular mimicry, i.e., similarity between parts of some viral proteins and one or more epitopes of GAD65. However, the possibility that bacterial proteins might also be responsible for GAD65 mimicry has been seldom investigated. To date, many genomes of Streptococcus pneumoniae (the pneumococcus), a prominent human pathogen particularly prevalent among children and the elderly, have been sequenced. A dataset of more than 9000 pneumococcal genomes was mined and two different (albeit related) genes (gadA and gadB), presumably encoding two glutamate decarboxylases similar to GAD65, were found. The various gadASpn alleles were present only in serotype 3 pneumococci belonging to the global lineage GPSC83, although some homologs have also been discovered in two subspecies of Streptococcus constellatus (pharyngis and viborgensis), an isolate of the group B streptococci, and several strains of Lactobacillus delbrueckii. Besides, gadBSpn alleles are present in > 10% of the isolates in our dataset and represent 16 GPSCs with 123 sequence types and 20 different serotypes. Sequence analyses indicated that gadA- and gadB-like genes have been mobilized among different bacteria either by prophage(s) or by integrative and conjugative element(s), respectively. Substantial similarities appear to exist between the putative pneumococcal glutamate decarboxylases and well-known epitopes of GAD65. In this sense, the use of broader pneumococcal conjugate vaccines such as PCV20 would prevent the majority of serotypes expressing those genes that might potentially contribute to T1DM. These results deserve upcoming studies on the possible involvement of S. pneumoniae in the etiopathogenesis and clinical onset of T1DM.
Collapse
Affiliation(s)
- Ernesto García
- Departamento de Biotecnología Microbiana y de Plantas, Centro de Investigaciones Biológicas Margarita Salas (CSIC), Ramiro de Maeztu 9, 28040, Madrid, Spain.
| |
Collapse
|
29
|
Qin L, Liang X, Qi Y, Luo Y, Xiao Q, Huang D, Zhou C, Jiang L, Zhou M, Zhou Y, Tang J, Tang Y. MPFC PV + interneurons are involved in the antidepressant effects of running exercise but not fluoxetine therapy. Neuropharmacology 2023:109669. [PMID: 37473999 DOI: 10.1016/j.neuropharm.2023.109669] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 07/14/2023] [Accepted: 07/17/2023] [Indexed: 07/22/2023]
Abstract
Depression is a complex psychiatric disorder. Previous studies have shown that running exercise reverses depression-like behavior faster and more effectively than fluoxetine therapy. GABAergic interneurons, including the PV+ interneuron subtype, in the medial prefrontal cortex (MPFC) are involved in pathological changes of depression. It was unknown whether running exercise and fluoxetine therapy reverse depression-like behavior via GABAergic interneurons or the PV+ interneurons subtype in MPFC. To address this issue, we subjected mice with chronic unpredictable stress (CUS) to a 4-week running exercise or fluoxetine therapy. Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis showed that running exercise enriched GABAergic synaptic pathways in the MPFC of CUS-exposed mice. However, the number of PV+ interneurons but not the total number of GABAergic interneurons in the MPFC of mice exposed to CUS reversed by running exercise, not fluoxetine therapy. Running exercise increased the relative gene expression levels of the PV gene in the MPFC of CUS-exposed mice without altering other subtypes of GABAergic interneurons. Moreover, running exercise and fluoxetine therapy both significantly improved the length, area and volume of dendrites and the spine morphology of PV+ interneurons in the MPFC of mice exposed to CUS. However, running exercise but not fluoxetine therapy improved the dendritic complexity level of PV+ interneurons in the MPFC of mice exposed to CUS. In summary, the number and dendritic complexity level of PV+ interneurons may be important therapeutic targets for the mechanism by which running exercise reverses depression-like behavior faster and more effectively than fluoxetine therapy.
Collapse
Affiliation(s)
- Lu Qin
- Department of Histology and Embryology, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, 400016, PR China; Laboratory of Stem Cells and Tissue Engineering, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, 400016, PR China
| | - Xin Liang
- Laboratory of Stem Cells and Tissue Engineering, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, 400016, PR China; Department of Pathology, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, 400016, PR China
| | - Yingqiang Qi
- Institute of Life Science, Chongqing Medical University, Chongqing, 400016, PR China
| | - Yanmin Luo
- Laboratory of Stem Cells and Tissue Engineering, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, 400016, PR China; Department of Physiology, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, 400016, PR China
| | - Qian Xiao
- Laboratory of Stem Cells and Tissue Engineering, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, 400016, PR China; Department of Radioactive Medicine, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, 400016, PR China
| | - Dujuan Huang
- Department of Histology and Embryology, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, 400016, PR China; Laboratory of Stem Cells and Tissue Engineering, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, 400016, PR China
| | - Chunni Zhou
- Department of Histology and Embryology, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, 400016, PR China; Laboratory of Stem Cells and Tissue Engineering, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, 400016, PR China
| | - Lin Jiang
- Laboratory of Stem Cells and Tissue Engineering, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, 400016, PR China; Lab Teaching & Management Center, Chongqing Medical University, Chongqing, 400016, PR China
| | - Mei Zhou
- Department of Histology and Embryology, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, 400016, PR China; Laboratory of Stem Cells and Tissue Engineering, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, 400016, PR China
| | - Yuning Zhou
- Department of Histology and Embryology, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, 400016, PR China; Laboratory of Stem Cells and Tissue Engineering, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, 400016, PR China
| | - Jing Tang
- Department of Histology and Embryology, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, 400016, PR China; Laboratory of Stem Cells and Tissue Engineering, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, 400016, PR China.
| | - Yong Tang
- Department of Histology and Embryology, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, 400016, PR China; Laboratory of Stem Cells and Tissue Engineering, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, 400016, PR China.
| |
Collapse
|
30
|
Fish KN, Rocco BR, Wilson JD, Lewis DA. Laminar-Specific Alterations in Calbindin-Positive Boutons in the Prefrontal Cortex of Subjects With Schizophrenia. Biol Psychiatry 2023; 94:142-152. [PMID: 36868891 PMCID: PMC10247897 DOI: 10.1016/j.biopsych.2022.12.004] [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: 06/07/2022] [Revised: 12/01/2022] [Accepted: 12/02/2022] [Indexed: 12/13/2022]
Abstract
BACKGROUND Cognitive deficits in schizophrenia are associated with altered GABA (gamma-aminobutyric acid) neurotransmission in the prefrontal cortex (PFC). GABA neurotransmission requires GABA synthesis by 2 isoforms of glutamic acid decarboxylase (GAD65 and GAD67) and packaging by the vesicular GABA transporter (vGAT). Current postmortem findings suggest that GAD67 messenger RNA is lower in a subset of the calbindin-expressing (CB+) class of GABA neurons in schizophrenia. Hence, we assessed if CB+ GABA neuron boutons are affected in schizophrenia. METHODS For 20 matched pairs of subjects with schizophrenia and unaffected comparison subjects, PFC tissue sections were immunolabeled for vGAT, CB, GAD67, and GAD65. The density of CB+ GABA boutons and levels of the 4 proteins per bouton were quantified. RESULTS Some CB+ GABA boutons contained both GAD65 and GAD67 (GAD65+/GAD67+), whereas others contained only GAD65 (GAD65+) or GAD67 (GAD67+). In schizophrenia, vGAT+/CB+/GAD65+/GAD67+ bouton density was not altered, vGAT+/CB+/GAD65+ bouton density was 86% higher in layers 2/superficial 3 (L2/3s), and vGAT+/CB+/GAD67+ bouton density was 36% lower in L5-6. Bouton GAD levels were differentially altered across bouton types and layers. In schizophrenia, the sum of GAD65 and GAD67 levels in vGAT+/CB+/GAD65+/GAD67+ boutons was 36% lower in L6, GAD65 levels were 51% higher in vGAT+/CB+/GAD65+ boutons in L2, and GAD67 levels in vGAT+/CB+/GAD67+ boutons were 30% to 46% lower in L2/3s-6. CONCLUSIONS These findings indicate that schizophrenia-associated alterations in the strength of inhibition from CB+ GABA neurons in the PFC differ across cortical layers and bouton classes, suggesting complex contributions to PFC dysfunction and cognitive impairments in schizophrenia.
Collapse
Affiliation(s)
- Kenneth N Fish
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania.
| | - Brad R Rocco
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - James D Wilson
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - David A Lewis
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; Department Neuroscience, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| |
Collapse
|
31
|
Zhou Y, Tao L, Zhu Y. TempShift Reveals the Sequential Development of Human Neocortex and Skewed Developmental Timing of Down Syndrome Brains. Brain Sci 2023; 13:1070. [PMID: 37509002 PMCID: PMC10377154 DOI: 10.3390/brainsci13071070] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 07/01/2023] [Accepted: 07/11/2023] [Indexed: 07/30/2023] Open
Abstract
Development is a complex process involving precise regulation. Developmental regulation may vary in tissues and individuals, and is often altered in disorders. Currently, the regulation of developmental timing across neocortical areas and developmental changes in Down syndrome (DS) brains remain unclear. The changes in regulation are often accompanied by changes in the gene expression trajectories, which can be divided into two scenarios: (1) changes of gene expression trajectory shape that reflect changes in cell type composition or altered molecular machinery; (2) temporal shift of gene expression trajectories that indicate different regulation of developmental timing. Therefore, we developed an R package TempShift to separates these two scenarios and demonstrated that TempShift can distinguish temporal shift from different shape (DiffShape) of expression trajectories, and can accurately estimate the time difference between multiple trajectories. We applied TempShift to identify sequential gene expression across 11 neocortical areas, which suggested sequential occurrence of synapse formation and axon guidance, as well as reconstructed interneuron migration pathways within neocortex. Comparison between healthy and DS brains revealed increased microglia, shortened neuronal migration process, and delayed synaptogenesis and myelination in DS. These applications also demonstrate the potential of TempShift in understanding gene expression temporal dynamics during different biological processes.
Collapse
Affiliation(s)
- Yuqiu Zhou
- State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science and Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai 200032, China
| | - Li Tao
- State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science and Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai 200032, China
| | - Ying Zhu
- State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science and Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai 200032, China
| |
Collapse
|
32
|
Woods R, Lorusso J, Fletcher J, ElTaher H, McEwan F, Harris I, Kowash H, D'Souza SW, Harte M, Hager R, Glazier JD. Maternal immune activation and role of placenta in the prenatal programming of neurodevelopmental disorders. Neuronal Signal 2023; 7:NS20220064. [PMID: 37332846 PMCID: PMC10273029 DOI: 10.1042/ns20220064] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 05/16/2023] [Accepted: 05/17/2023] [Indexed: 06/20/2023] Open
Abstract
Maternal infection during pregnancy, leading to maternal immune activation (mIA) and cytokine release, increases the offspring risk of developing a variety of neurodevelopmental disorders (NDDs), including schizophrenia. Animal models have provided evidence to support these mechanistic links, with placental inflammatory responses and dysregulation of placental function implicated. This leads to changes in fetal brain cytokine balance and altered epigenetic regulation of key neurodevelopmental pathways. The prenatal timing of such mIA-evoked changes, and the accompanying fetal developmental responses to an altered in utero environment, will determine the scope of the impacts on neurodevelopmental processes. Such dysregulation can impart enduring neuropathological changes, which manifest subsequently in the postnatal period as altered neurodevelopmental behaviours in the offspring. Hence, elucidation of the functional changes that occur at the molecular level in the placenta is vital in improving our understanding of the mechanisms that underlie the pathogenesis of NDDs. This has notable relevance to the recent COVID-19 pandemic, where inflammatory responses in the placenta to SARS-CoV-2 infection during pregnancy and NDDs in early childhood have been reported. This review presents an integrated overview of these collective topics and describes the possible contribution of prenatal programming through placental effects as an underlying mechanism that links to NDD risk, underpinned by altered epigenetic regulation of neurodevelopmental pathways.
Collapse
Affiliation(s)
- Rebecca M. Woods
- Division of Evolution, Infection and Genomics, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PT, U.K
| | - Jarred M. Lorusso
- Division of Evolution, Infection and Genomics, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PT, U.K
| | - Jennifer Fletcher
- Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Medicine, Biology and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PT, U.K
| | - Heidi ElTaher
- Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Medicine, Biology and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PT, U.K
- Department of Physiology, Faculty of Medicine, Alexandria University, Egypt
| | - Francesca McEwan
- Division of Evolution, Infection and Genomics, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PT, U.K
| | - Isabella Harris
- Division of Evolution, Infection and Genomics, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PT, U.K
| | - Hager M. Kowash
- Division of Developmental Biology and Medicine, School of Medical Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9WL, U.K
| | - Stephen W. D'Souza
- Division of Developmental Biology and Medicine, School of Medical Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9WL, U.K
| | - Michael Harte
- Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Medicine, Biology and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PT, U.K
| | - Reinmar Hager
- Division of Evolution, Infection and Genomics, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PT, U.K
| | - Jocelyn D. Glazier
- Division of Evolution, Infection and Genomics, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PT, U.K
| |
Collapse
|
33
|
Ma S, Zhong H, Liu X, Wang L. Spatial Distribution of Neurons Expressing Single, Double, and Triple Molecular Characteristics of Glutamatergic, Dopaminergic, or GABAergic Neurons in the Mouse Ventral Tegmental Area. J Mol Neurosci 2023; 73:345-362. [PMID: 37243808 DOI: 10.1007/s12031-023-02121-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 05/16/2023] [Indexed: 05/29/2023]
Abstract
The ventral tegmental area (VTA) is a heterogeneous midbrain area that plays a significant role in diverse neural processes such as reward, aversion, and motivation. The VTA contains three main neuronal populations, namely, dopamine (DA), γ-aminobutyric acid (GABA), and glutamate neurons, but some neurons exhibit combinatorial molecular characteristics of dopaminergic, GABAergic, or glutamatergic neurons. However, little information is available regarding detailed distribution of neurons with single, double, and triple molecular characteristics of glutamatergic, dopaminergic, or GABAergic neurons in mice. We present a topographical distribution map of three main neuronal populations expressing a single molecular characteristic of dopaminergic, GABAergic, or glutamatergic neurons, and four neuronal populations co-expressing double or triple molecular characteristics in combinatorial manners, in the mouse VTA, following analysis of triple fluorescent in situ hybridization for the simultaneous detection of tyrosine hydroxylase (TH, marker for dopaminergic neurons), vesicular glutamate transporter 2 (VGLUT2, marker for glutamatergic neurons), and glutamic acid decarboxylase 2 (GAD2, marker for GABAergic neurons) mRNA. We found that the vast majority of neurons expressed a single type of mRNA, and these neurons were intermingled with neurons co-expressing double or triple combinations of VGLUT2, TH, or GAD2 in the VTA. These seven neuronal populations were differentially distributed in the VTA sub-nuclei across the rostro-caudal and latero-medial axes. This histochemical study will lead to a deeper understanding of the complexity of neuronal molecular characteristics in different VTA sub-nuclei, and potentially facilitate clarification of diverse functions of the VTA.
Collapse
Affiliation(s)
- Shaohua Ma
- Shenzhen Key Laboratory of Neuropsychiatric Modulation, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
- CAS Key Laboratory of Brain Connectome and Manipulation, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
- Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Hao Zhong
- Shenzhen Key Laboratory of Neuropsychiatric Modulation, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
- CAS Key Laboratory of Brain Connectome and Manipulation, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
- Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Xuemei Liu
- Shenzhen Key Laboratory of Neuropsychiatric Modulation, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
- CAS Key Laboratory of Brain Connectome and Manipulation, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
- Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
| | - Liping Wang
- Shenzhen Key Laboratory of Neuropsychiatric Modulation, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
- CAS Key Laboratory of Brain Connectome and Manipulation, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
- Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
| |
Collapse
|
34
|
Bach KT, Ananth S, Thind I, Zhong N, Lui F. Co-occurrence of Anti-GAD65 Syndrome, Type 1 Diabetes Mellitus, and Focal Seizures With Impaired Awareness. Cureus 2023; 15:e40611. [PMID: 37476109 PMCID: PMC10354376 DOI: 10.7759/cureus.40611] [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: 06/18/2023] [Indexed: 07/22/2023] Open
Abstract
Glutamic acid decarboxylase (GAD) is an intracellular enzyme found in the presynaptic end of nerve terminals that functions to synthesize gamma-aminobutyric acid (GABA) via decarboxylation. Autoantibodies to the GAD65 isoform have been found in high levels in neurological disorders including stiff person syndrome (SPS), autoimmune encephalitis, and refractory epilepsy. Low levels of anti-GAD65 have also been noted in type 1 diabetes mellitus. We present the unusual case of a woman with a longstanding history of focal seizures with impaired awareness and type 1 diabetes mellitus who was found to have extremely high titers of anti-GAD65 and clinical presentation suggestive of stiff person syndrome. This case highlights the increasing significance of autoimmune etiologies within neurologic disorders, as well as the importance of maintaining a high index of suspicion for rare anti-GAD65 syndromes. Although uncommon and with an unclear pathophysiology, we discuss the importance of establishing SPS diagnostic criteria to facilitate timely diagnosis and quickly direct patient management towards immunotherapy.
Collapse
Affiliation(s)
- Kieu-Tram Bach
- Clinical Sciences, California Northstate University College of Medicine, Sacramento, USA
| | - Shahini Ananth
- Clinical Sciences, California Northstate University College of Medicine, Sacramento, USA
| | - Ikjot Thind
- Clinical Sciences, California Northstate University College of Medicine, Sacramento, USA
| | - Ning Zhong
- Neurology, Kaiser Permanente Sacramento Medical Center, Sacramento, USA
| | - Forshing Lui
- Clinical Sciences, California Northstate University College of Medicine, Elk Grove, USA
| |
Collapse
|
35
|
Ueberbach T, Simacek CA, Tegeder I, Kirischuk S, Mittmann T. Tonic activation of GABA B receptors via GAT-3 mediated GABA release reduces network activity in the developing somatosensory cortex in GAD67-GFP mice. Front Synaptic Neurosci 2023; 15:1198159. [PMID: 37325697 PMCID: PMC10267986 DOI: 10.3389/fnsyn.2023.1198159] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 05/05/2023] [Indexed: 06/17/2023] Open
Abstract
The efficiency of neocortical information processing critically depends on the balance between the glutamatergic (excitatory, E) and GABAergic (inhibitory, I) synaptic transmission. A transient imbalance of the E/I-ratio during early development might lead to neuropsychiatric disorders later in life. The transgenic glutamic acid decarboxylase 67-green fluorescent protein (GAD67-GFP) mouse line (KI) was developed to selectively visualize GABAergic interneurons in the CNS. However, haplodeficiency of the GAD67 enzyme, the main GABA synthetizing enzyme in the brain, temporarily leads to a low GABA level in the developing brain of these animals. However, KI mice did not demonstrate any epileptic activity and only few and mild behavioral deficits. In the present study we investigated how the developing somatosensory cortex of KI-mice compensates the reduced GABA level to prevent brain hyperexcitability. Whole-cell patch clamp recordings from layer 2/3 pyramidal neurons at P14 and at P21 revealed a reduced frequency of miniature inhibitory postsynaptic currents (mIPSCs) in KI mice without any change in amplitude or kinetics. Interestingly, mEPSC frequencies were also decreased, while the E/I-ratio was nevertheless shifted toward excitation. Surprisingly, multi-electrode-recordings (MEA) from acute slices revealed a decreased spontaneous neuronal network activity in KI mice compared to wild-type (WT) littermates, pointing to a compensatory mechanism that prevents hyperexcitability. Blockade of GABAB receptors (GABABRs) with CGP55845 strongly increased the frequency of mEPSCs in KI, but failed to affect mIPSCs in any genotype or age. It also induced a membrane depolarization in P14 KI, but not in P21 KI or WT mice. MEA recordings in presence of CGP55845 revealed comparable levels of network activity in both genotypes, indicating that tonically activated GABABRs balance neuronal activity in P14 KI cortex despite the reduced GABA levels. Blockade of GABA transporter 3 (GAT-3) reproduced the CGP55845 effects suggesting that tonic activation of GABABRs is mediated by ambient GABA released via GAT-3 operating in reverse mode. We conclude that GAT-3-mediated GABA release leads to tonic activation of both pre- and postsynaptic GABABRs and restricts neuronal excitability in the developing cortex to compensate for reduced neuronal GABA synthesis. Since GAT-3 is predominantly located in astrocytes, GAD67 haplodeficiency may potentially stimulate astrocytic GABA synthesis through GAD67-independent pathways.
Collapse
Affiliation(s)
- Timo Ueberbach
- Institute for Physiology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Clara A. Simacek
- Institute for Physiology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Irmgard Tegeder
- Institute of Clinical Pharmacology, Faculty of Medicine, Goethe-University, Frankfurt, Germany
| | - Sergei Kirischuk
- Institute for Physiology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Thomas Mittmann
- Institute for Physiology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| |
Collapse
|
36
|
Matsunaga W, Shinoe T, Hirono M. GAD65 deficient mice are susceptible to ethanol-induced impairment of motor coordination and facilitation of cerebellar neuronal firing. PLoS One 2023; 18:e0286031. [PMID: 37216370 DOI: 10.1371/journal.pone.0286031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2023] [Accepted: 05/06/2023] [Indexed: 05/24/2023] Open
Abstract
γ-aminobutyric acid (GABA) is a major inhibitory neurotransmitter and its concentrations in the brain could be associated with EtOH-induced impairment of motor coordination. GABA is synthesized by two isoforms of glutamate decarboxylase (GAD): GAD65 and GAD67. Mice deficient in GAD65 (GAD65-KO) can grow up to adulthood, and show that GABA concentration in their adult brains was 50-75% that of wild-type C57BL/6 mice (WT). Although a previous study showed that there was no difference in recovery from the motor-incoordination effect of acute intraperitoneally administered injections of 2.0 g/kg EtOH between WT and GAD65-KO, the sensitivity of GAD65-KO to acute EtOH-induced ataxia has not been fully understood. Here, we sought to determine whether motor coordination and spontaneous firing of cerebellar Purkinje cells (PCs) in GAD65-KO are more sensitive to the effect of EtOH than in WT. Motor performance in WT and GAD65-KO was examined by rotarod and open-field tests following acute administration of EtOH at lower-doses, 0.8, 1.2 and 1.6 g/kg. In a rotarod test, there was no significant difference between WT and GAD65-KO in terms of baseline motor coordination. However, only the KO mice showed a significant decrease in rotarod performance of 1.2 g/kg EtOH. In the open-field test, GAD65-KO showed a significant increase in locomotor activity after 1.2 and 1.6 g/kg EtOH injections, but not WT. In in vitro studies of cerebellar slices, the firing rate of PCs was increased by 50 mM EtOH in GAD65-KO compared with WT, whereas no difference was observed in the effect of EtOH at more than 100 mM between the genotypes. Taken together, GAD65-KO are more susceptible to the effect of acute EtOH exposure on motor coordination and PC firing than WT. This different sensitivity could be attributed to the basal low GABA concentration in the brain of GAD65-KO.
Collapse
Affiliation(s)
| | - Toru Shinoe
- RIKEN Brain Science Institute, Wako, Saitama, Japan
| | | |
Collapse
|
37
|
Carrero L, Antequera D, Alcalde I, Megías D, Figueiro-Silva J, Merayo-Lloves J, Municio C, Carro E. Disturbed circadian rhythm and retinal degeneration in a mouse model of Alzheimer's disease. Acta Neuropathol Commun 2023; 11:55. [PMID: 37004084 PMCID: PMC10067208 DOI: 10.1186/s40478-023-01529-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 02/11/2023] [Indexed: 04/03/2023] Open
Abstract
The circadian clock is synchronized to the 24 h day by environmental light which is transmitted from the retina to the suprachiasmatic nucleus (SCN) primarily via the retinohypothalamic tract (RHT). Circadian rhythm abnormalities have been reported in neurodegenerative disorders such as Alzheimer's disease (AD). Whether these AD-related changes are a result of the altered clock gene expression, retina degeneration, including the dysfunction in RHT transmission, loss of retinal ganglion cells and its electrophysiological capabilities, or a combination of all of these pathological mechanisms, is not known. Here, we evaluated transgenic APP/PS1 mouse model of AD and wild-type mice at 6- and 12-month-old, as early and late pathological stage, respectively. We noticed the alteration of circadian clock gene expression not only in the hypothalamus but also in two extra-hypothalamic brain regions, cerebral cortex and hippocampus, in APP/PS1 mice. These alterations were observed in 6-month-old transgenic mice and were exacerbated at 12 months of age. This could be explained by the reduced RHT projections in the SCN of APP/PS1 mice, correlating with downregulation of hypothalamic GABAergic response in APP/PS1 mice in advanced stage of pathology. Importantly, we also report retinal degeneration in APP/PS1 mice, including Aβ deposits and reduced choline acetyltransferase levels, loss of melanopsin retinal ganglion cells and functional integrity mainly of inner retina layers. Our findings support the theory that retinal degeneration constitutes an early pathological event that directly affects the control of circadian rhythm in AD.
Collapse
Affiliation(s)
- Laura Carrero
- Group of Neurodegenerative Diseases, Hospital Universitario 12 de Octubre Research Institute (imas12), 28041, Madrid, Spain
- Network Center for Biomedical Research in Neurodegenerative Diseases (CIBERNED), ISCIII, Madrid, Spain
- Autonoma de Madrid University, Madrid, Spain
| | - Desireé Antequera
- Network Center for Biomedical Research in Neurodegenerative Diseases (CIBERNED), ISCIII, Madrid, Spain
- Neurobiology of Alzheimer's Disease Unit, Functional Unit for Research into Chronic Diseases, Instituto de Salud Carlos III, Madrid, Spain
| | - Ignacio Alcalde
- Instituto Universitario Fernández-Vega, Universidad de Oviedo and Fundación de Investigación Oftalmológica, Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain
| | - Diego Megías
- Advanced Optical Microscopy Unit, Unidades Centrales Científico-Técnicas, Instituto de Salud Carlos III, Madrid, Spain
| | - Joana Figueiro-Silva
- Institute of Medical Genetics, University of Zurich, Zurich, Switzerland
- Department of Molecular Life Science, University of Zurich, Zurich, Switzerland
| | - Jesús Merayo-Lloves
- Instituto Universitario Fernández-Vega, Universidad de Oviedo and Fundación de Investigación Oftalmológica, Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain
| | - Cristina Municio
- Group of Neurodegenerative Diseases, Hospital Universitario 12 de Octubre Research Institute (imas12), 28041, Madrid, Spain.
- Network Center for Biomedical Research in Neurodegenerative Diseases (CIBERNED), ISCIII, Madrid, Spain.
| | - Eva Carro
- Network Center for Biomedical Research in Neurodegenerative Diseases (CIBERNED), ISCIII, Madrid, Spain.
- Neurobiology of Alzheimer's Disease Unit, Functional Unit for Research into Chronic Diseases, Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
38
|
van Hooijdonk CFM, van der Pluijm M, Bosch I, van Amelsvoort TAMJ, Booij J, de Haan L, Selten JP, Giessen EVD. The substantia nigra in the pathology of schizophrenia: A review on post-mortem and molecular imaging findings. Eur Neuropsychopharmacol 2023; 68:57-77. [PMID: 36640734 DOI: 10.1016/j.euroneuro.2022.12.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 12/19/2022] [Accepted: 12/21/2022] [Indexed: 01/14/2023]
Abstract
Dysregulation of striatal dopamine is considered to be an important driver of pathophysiological processes in schizophrenia. Despite being one of the main origins of dopaminergic input to the striatum, the (dys)functioning of the substantia nigra (SN) has been relatively understudied in schizophrenia. Hence, this paper aims to review different molecular aspects of nigral functioning in patients with schizophrenia compared to healthy controls by integrating post-mortem and molecular imaging studies. We found evidence for hyperdopaminergic functioning in the SN of patients with schizophrenia (i.e. increased AADC activity in antipsychotic-free/-naïve patients and elevated neuromelanin accumulation). Reduced GABAergic inhibition (i.e. decreased density of GABAergic synapses, lower VGAT mRNA levels and lower mRNA levels for GABAA receptor subunits), excessive glutamatergic excitation (i.e. increased NR1 and Glur5 mRNA levels and a reduced number of astrocytes), and several other disturbances implicating the SN (i.e. immune functioning and copper concentrations) could potentially underlie this nigral hyperactivity and associated striatal hyperdopaminergic functioning in schizophrenia. These results highlight the importance of the SN in schizophrenia pathology and suggest that some aspects of molecular functioning in the SN could potentially be used as treatment targets or biomarkers.
Collapse
Affiliation(s)
- Carmen F M van Hooijdonk
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNs), University of Maastricht, Maastricht, the Netherlands; Rivierduinen, Institute for Mental Health Care, Leiden, the Netherlands.
| | - Marieke van der Pluijm
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, University of Amsterdam, the Netherlands
| | - Iris Bosch
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, University of Amsterdam, the Netherlands
| | - Therese A M J van Amelsvoort
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNs), University of Maastricht, Maastricht, the Netherlands
| | - Jan Booij
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, University of Amsterdam, the Netherlands
| | - Lieuwe de Haan
- Department of Psychiatry, Amsterdam UMC, University of Amsterdam, the Netherlands
| | - Jean-Paul Selten
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNs), University of Maastricht, Maastricht, the Netherlands; Rivierduinen, Institute for Mental Health Care, Leiden, the Netherlands
| | - Elsmarieke van de Giessen
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, University of Amsterdam, the Netherlands
| |
Collapse
|
39
|
A Case of Anti-GAD 65 Autoimmune Encephalitis Associated with Focal Segmental Stiff-Person Syndrome. Brain Sci 2023; 13:brainsci13020369. [PMID: 36831912 PMCID: PMC9954581 DOI: 10.3390/brainsci13020369] [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: 01/06/2023] [Revised: 02/10/2023] [Accepted: 02/16/2023] [Indexed: 02/23/2023] Open
Abstract
Glutamic acid decarboxylase (GAD) antibody-related encephalitis is an autoimmune disease associated with intracellular neuronal antigens. We report on a rare case of GAD antibody-associated encephalitis complicated with focal segmental stiffness-person syndrome (SPS) in a middle-aged woman. The disease course lasted for >10 years, initially presenting with drug-resistant epilepsy, followed by stiffness of the right lower limb, and right upper limb involvement. The patient experienced anxiety and depression symptoms due to long-term illness. During hospitalization, serum and cerebrospinal fluid GAD antibodies were positive and no tumor was found. The symptoms were significantly relieved after corticosteroid therapy and intravenous immunoglobulin immunomodulation therapy. To the best of our knowledge, this case is the first to discuss the early recognition and treatment of chronic epilepsy and focal segmental SPS caused by anti-GAD antibody-related encephalitis.
Collapse
|
40
|
Martin-Lopez E, Vidyadhara DJ, Liberia T, Meller SJ, Harmon LE, Hsu RM, Spence N, Brennan B, Han K, Yücel B, Chandra SS, Greer CA. α-Synuclein Pathology and Reduced Neurogenesis in the Olfactory System Affect Olfaction in a Mouse Model of Parkinson's Disease. J Neurosci 2023; 43:1051-1071. [PMID: 36596700 PMCID: PMC9908323 DOI: 10.1523/jneurosci.1526-22.2022] [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: 08/09/2022] [Revised: 12/09/2022] [Accepted: 12/15/2022] [Indexed: 01/05/2023] Open
Abstract
Parkinson's disease (PD) is characterized by multiple symptoms including olfactory dysfunction, whose underlying mechanisms remain unclear. Here, we explored pathologic changes in the olfactory pathway of transgenic (Tg) mice of both sexes expressing the human A30P mutant α-synuclein (α-syn; α-syn-Tg mice) at 6-7 and 12-14 months of age, representing early and late-stages of motor progression, respectively. α-Syn-Tg mice at late stages exhibited olfactory behavioral deficits, which correlated with severe α-syn pathology in projection neurons (PNs) of the olfactory pathway. In parallel, olfactory bulb (OB) neurogenesis in α-syn-Tg mice was reduced in the OB granule cells at six to seven months and OB periglomerular cells at 12-14 months, respectively, both of which could contribute to olfactory dysfunction. Proteomic analyses showed a disruption in endocytic and exocytic pathways in the OB during the early stages which appeared exacerbated at the synaptic terminals when the mice developed olfactory deficits at 12-14 months. Our data suggest that (1) the α-syn-Tg mice recapitulate the olfactory functional deficits seen in PD; (2) olfactory structures exhibit spatiotemporal disparities for vulnerability to α-syn pathology; (3) α-syn pathology is restricted to projection neurons in the olfactory pathway; (4) neurogenesis in adult α-syn-Tg mice is reduced in the OB; and (5) synaptic endocytosis and exocytosis defects in the OB may further explain olfactory deficits.SIGNIFICANCE STATEMENT Olfactory dysfunction is a characteristic symptom of Parkinson's disease (PD). Using the human A30P mutant α-synuclein (α-syn)-expressing mouse model, we demonstrated the appearance of olfactory deficits at late stages of the disease, which was accompanied by the accumulation of α-syn pathology in projection neurons (PNs) of the olfactory system. This dysfunction included a reduction in olfactory bulb (OB) neurogenesis as well as changes in synaptic vesicular transport affecting synaptic function, both of which are likely contributing to olfactory behavioral deficits.
Collapse
Affiliation(s)
- Eduardo Martin-Lopez
- Department of Neurosurgery, Yale University School of Medicine, New Haven, Connecticut 06510
- Department of Neuroscience, Yale University School of Medicine, New Haven, Connecticut 06510
| | - D J Vidyadhara
- Department of Neurology, Yale University School of Medicine, New Haven, Connecticut 06510
- Department of Neuroscience, Yale University School of Medicine, New Haven, Connecticut 06510
| | - Teresa Liberia
- Department of Neurosurgery, Yale University School of Medicine, New Haven, Connecticut 06510
- Department of Neuroscience, Yale University School of Medicine, New Haven, Connecticut 06510
| | - Sarah J Meller
- Department of Neurosurgery, Yale University School of Medicine, New Haven, Connecticut 06510
- Department of Neuroscience, Yale University School of Medicine, New Haven, Connecticut 06510
| | - Leah E Harmon
- Department of Neurology, Yale University School of Medicine, New Haven, Connecticut 06510
- Department of Neuroscience, Yale University School of Medicine, New Haven, Connecticut 06510
| | - Ryan M Hsu
- Department of Neurosurgery, Yale University School of Medicine, New Haven, Connecticut 06510
- Department of Neuroscience, Yale University School of Medicine, New Haven, Connecticut 06510
| | - Natalie Spence
- Department of Neurosurgery, Yale University School of Medicine, New Haven, Connecticut 06510
- Department of Neuroscience, Yale University School of Medicine, New Haven, Connecticut 06510
| | - Bowen Brennan
- Department of Neurosurgery, Yale University School of Medicine, New Haven, Connecticut 06510
- Department of Neuroscience, Yale University School of Medicine, New Haven, Connecticut 06510
| | - Kimberly Han
- Department of Neurosurgery, Yale University School of Medicine, New Haven, Connecticut 06510
- Department of Neuroscience, Yale University School of Medicine, New Haven, Connecticut 06510
| | - Betül Yücel
- Department of Neurology, Yale University School of Medicine, New Haven, Connecticut 06510
- Department of Neuroscience, Yale University School of Medicine, New Haven, Connecticut 06510
| | - Sreeganga S Chandra
- Department of Neurology, Yale University School of Medicine, New Haven, Connecticut 06510
- Department of Neuroscience, Yale University School of Medicine, New Haven, Connecticut 06510
| | - Charles A Greer
- Department of Neurosurgery, Yale University School of Medicine, New Haven, Connecticut 06510
- Department of Neuroscience, Yale University School of Medicine, New Haven, Connecticut 06510
| |
Collapse
|
41
|
Lam P, Newland J, Faull RLM, Kwakowsky A. Cation-Chloride Cotransporters KCC2 and NKCC1 as Therapeutic Targets in Neurological and Neuropsychiatric Disorders. Molecules 2023; 28:1344. [PMID: 36771011 PMCID: PMC9920462 DOI: 10.3390/molecules28031344] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 01/21/2023] [Accepted: 01/25/2023] [Indexed: 02/04/2023] Open
Abstract
Neurological diseases including Alzheimer's, Huntington's disease, Parkinson's disease, Down syndrome and epilepsy, and neuropsychiatric disorders such as schizophrenia, are conditions that affect not only individuals but societies on a global scale. Current therapies offer a means for small symptomatic relief, but recently there has been increasing demand for therapeutic alternatives. The γ-aminobutyric acid (GABA)ergic signaling system has been investigated for developing new therapies as it has been noted that any dysfunction or changes to this system can contribute to disease progression. Expression of the K-Cl-2 (KCC2) and N-K-C1-1 (NKCC1) cation-chloride cotransporters (CCCs) has recently been linked to the disruption of GABAergic activity by affecting the polarity of GABAA receptor signaling. KCC2 and NKCC1 play a part in multiple neurological and neuropsychiatric disorders, making them a target of interest for potential therapies. This review explores current research suggesting the pathophysiological role and therapeutic importance of KCC2 and NKCC1 in neuropsychiatric and neurological disorders.
Collapse
Affiliation(s)
- Patricia Lam
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1023, New Zealand
| | - Julia Newland
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1023, New Zealand
| | - Richard L. M. Faull
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1023, New Zealand
| | - Andrea Kwakowsky
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1023, New Zealand
- Pharmacology and Therapeutics, School of Medicine, Galway Neuroscience Centre, University of Galway, H91 W5P7 Galway, Ireland
| |
Collapse
|
42
|
The GABA and GABA-Receptor System in Inflammation, Anti-Tumor Immune Responses, and COVID-19. Biomedicines 2023; 11:biomedicines11020254. [PMID: 36830790 PMCID: PMC9953446 DOI: 10.3390/biomedicines11020254] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 01/16/2023] [Accepted: 01/16/2023] [Indexed: 01/20/2023] Open
Abstract
GABA and GABAA-receptors (GABAA-Rs) play major roles in neurodevelopment and neurotransmission in the central nervous system (CNS). There has been a growing appreciation that GABAA-Rs are also present on most immune cells. Studies in the fields of autoimmune disease, cancer, parasitology, and virology have observed that GABA-R ligands have anti-inflammatory actions on T cells and antigen-presenting cells (APCs), while also enhancing regulatory T cell (Treg) responses and shifting APCs toward anti-inflammatory phenotypes. These actions have enabled GABAA-R ligands to ameliorate autoimmune diseases, such as type 1 diabetes (T1D), multiple sclerosis (MS), and rheumatoid arthritis, as well as type 2 diabetes (T2D)-associated inflammation in preclinical models. Conversely, antagonism of GABAA-R activity promotes the pro-inflammatory responses of T cells and APCs, enhancing anti-tumor responses and reducing tumor burden in models of solid tumors. Lung epithelial cells also express GABA-Rs, whose activation helps maintain fluid homeostasis and promote recovery from injury. The ability of GABAA-R agonists to limit both excessive immune responses and lung epithelial cell injury may underlie recent findings that GABAA-R agonists reduce the severity of disease in mice infected with highly lethal coronaviruses (SARS-CoV-2 and MHV-1). These observations suggest that GABAA-R agonists may provide off-the-shelf therapies for COVID-19 caused by new SARS-CoV-2 variants, as well as novel beta-coronaviruses, which evade vaccine-induced immune responses and antiviral medications. We review these findings and further advance the notions that (1) immune cells possess GABAA-Rs to limit inflammation in the CNS, and (2) this natural "braking system" on inflammatory responses may be pharmacologically engaged to slow the progression of autoimmune diseases, reduce the severity of COVID-19, and perhaps limit neuroinflammation associated with long COVID.
Collapse
|
43
|
Investigating the Role of GABA in Neural Development and Disease Using Mice Lacking GAD67 or VGAT Genes. Int J Mol Sci 2022; 23:ijms23147965. [PMID: 35887307 PMCID: PMC9318753 DOI: 10.3390/ijms23147965] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 07/12/2022] [Accepted: 07/16/2022] [Indexed: 11/18/2022] Open
Abstract
Normal development and function of the central nervous system involves a balance between excitatory and inhibitory neurotransmission. Activity of both excitatory and inhibitory neurons is modulated by inhibitory signalling of the GABAergic and glycinergic systems. Mechanisms that regulate formation, maturation, refinement, and maintenance of inhibitory synapses are established in early life. Deviations from ideal excitatory and inhibitory balance, such as down-regulated inhibition, are linked with many neurological diseases, including epilepsy, schizophrenia, anxiety, and autism spectrum disorders. In the mammalian forebrain, GABA is the primary inhibitory neurotransmitter, binding to GABA receptors, opening chloride channels and hyperpolarizing the cell. We review the involvement of down-regulated inhibitory signalling in neurological disorders, possible mechanisms for disease progression, and targets for therapeutic intervention. We conclude that transgenic models of disrupted inhibitory signalling—in GAD67+/− and VGAT−/− mice—are useful for investigating the effects of down-regulated inhibitory signalling in a range of neurological diseases.
Collapse
|
44
|
Mafa-dependent GABAergic activity promotes mouse neonatal apneas. Nat Commun 2022; 13:3284. [PMID: 35672398 PMCID: PMC9174494 DOI: 10.1038/s41467-022-30825-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Accepted: 05/19/2022] [Indexed: 01/17/2023] Open
Abstract
While apneas are associated with multiple pathological and fatal conditions, the underlying molecular mechanisms remain elusive. We report that a mutated form of the transcription factor Mafa (Mafa4A) that prevents phosphorylation of the Mafa protein leads to an abnormally high incidence of breath holding apneas and death in newborn Mafa4A/4A mutant mice. This apneic breathing is phenocopied by restricting the mutation to central GABAergic inhibitory neurons and by activation of inhibitory Mafa neurons while reversed by inhibiting GABAergic transmission centrally. We find that Mafa activates the Gad2 promoter in vitro and that this activation is enhanced by the mutation that likely results in increased inhibitory drives onto target neurons. We also find that Mafa inhibitory neurons are absent from respiratory, sensory (primary and secondary) and pontine structures but are present in the vicinity of the hypoglossal motor nucleus including premotor neurons that innervate the geniohyoid muscle, to control upper airway patency. Altogether, our data reveal a role for Mafa phosphorylation in regulation of GABAergic drives and suggest a mechanism whereby reduced premotor drives to upper airway muscles may cause apneic breathing at birth. Apneas are associated with many pathological conditions. Here, the authors show in a mouse model that stabilization of the transcription factor Mafa in brainstem GABAergic neurons may contribute to apnea, by decreasing motor drive to muscles controlling the airways.
Collapse
|
45
|
Rahaman SM, Chowdhury S, Mukai Y, Ono D, Yamaguchi H, Yamanaka A. Functional Interaction Between GABAergic Neurons in the Ventral Tegmental Area and Serotonergic Neurons in the Dorsal Raphe Nucleus. Front Neurosci 2022; 16:877054. [PMID: 35663550 PMCID: PMC9160575 DOI: 10.3389/fnins.2022.877054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 04/21/2022] [Indexed: 12/02/2022] Open
Abstract
GABAergic neurons in the ventral tegmental area (VTA) have brain-wide projections and are involved in multiple behavioral and physiological functions. Here, we revealed the responsiveness of Gad67+ neurons in VTA (VTAGad67+) to various neurotransmitters involved in the regulation of sleep/wakefulness by slice patch clamp recording. Among the substances tested, a cholinergic agonist activated, but serotonin, dopamine and histamine inhibited these neurons. Dense VTAGad67+ neuronal projections were observed in brain areas regulating sleep/wakefulness, including the central amygdala (CeA), dorsal raphe nucleus (DRN), and locus coeruleus (LC). Using a combination of electrophysiology and optogenetic studies, we showed that VTAGad67+ neurons inhibited all neurons recorded in the DRN, but did not inhibit randomly recorded neurons in the CeA and LC. Further examination revealed that the serotonergic neurons in the DRN (DRN5–HT) were monosynaptically innervated and inhibited by VTAGad67+ neurons. All recorded DRN5–HT neurons received inhibitory input from VTAGad67+ neurons, while only one quarter of them received inhibitory input from local GABAergic neurons. Gad67+ neurons in the DRN (DRNGad67+) also received monosynaptic inhibitory input from VTAGad67+ neurons. Taken together, we found that VTAGad67+ neurons were integrated in many inputs, and their output inhibits DRN5–HT neurons, which may regulate physiological functions including sleep/wakefulness.
Collapse
Affiliation(s)
- Sheikh Mizanur Rahaman
- Department of Neuroscience II, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Japan
- Department of Neural Regulation, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Srikanta Chowdhury
- Department of Neuroscience II, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Japan
- Department of Neural Regulation, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Department of Medicine, Columbia University College of Physicians and Surgeons, New York, NY, United States
| | - Yasutaka Mukai
- Department of Neuroscience II, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Japan
- Department of Neural Regulation, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Daisuke Ono
- Department of Neuroscience II, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Japan
- Department of Neural Regulation, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hiroshi Yamaguchi
- Department of Neuroscience II, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Japan
- Department of Neural Regulation, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Akihiro Yamanaka
- Department of Neuroscience II, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Japan
- Department of Neural Regulation, Nagoya University Graduate School of Medicine, Nagoya, Japan
- *Correspondence: Akihiro Yamanaka,
| |
Collapse
|
46
|
Canzian J, Gonçalves FLS, Müller TE, Franscescon F, Santos LW, Adedara IA, Rosemberg DB. Zebrafish as a potential non-traditional model organism in translational bipolar disorder research: Genetic and behavioral insights. Neurosci Biobehav Rev 2022; 136:104620. [PMID: 35300991 DOI: 10.1016/j.neubiorev.2022.104620] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 02/16/2022] [Accepted: 03/10/2022] [Indexed: 01/14/2023]
Abstract
Bipolar disorder (BD) is a severe and debilitating illness that affects 1-2% of the population worldwide. BD is characterized by recurrent and extreme mood swings, including mania/hypomania and depression. Animal experimental models have been used to elucidate the mechanisms underlying BD and different strategies have been proposed to assess BD-like symptoms. The zebrafish (Danio rerio) has been considered a suitable vertebrate system for modeling BD-like responses, due to the genetic tractability, molecular/physiological conservation, and well-characterized behavioral responses. In this review, we discuss how zebrafish-based models can be successfully used to understand molecular, biochemical, and behavioral alterations paralleling those found in BD. We also outline some advantages and limitations of this aquatic species to examine BD-like phenotypes in translational neurobehavioral research. Overall, we reinforce the use of zebrafish as a promising tool to investigate the neural basis associated with BD-like behaviors, which may foster the discovery of novel pharmacological therapies.
Collapse
Affiliation(s)
- Julia Canzian
- Laboratory of Experimental Neuropsychobiology, Department of Biochemistry and Molecular Biology, Natural and Exact Sciences Center, Federal University of Santa Maria, 1000 Roraima Avenue, Santa Maria, RS 97105-900, Brazil; Graduate Program in Biological Sciences: Toxicological Biochemistry, Federal University of Santa Maria, 1000 Roraima Avenue, Santa Maria, RS 97105-900, Brazil
| | - Falco L S Gonçalves
- Laboratory of Experimental Neuropsychobiology, Department of Biochemistry and Molecular Biology, Natural and Exact Sciences Center, Federal University of Santa Maria, 1000 Roraima Avenue, Santa Maria, RS 97105-900, Brazil
| | - Talise E Müller
- Graduate Program in Biological Sciences: Toxicological Biochemistry, Federal University of Santa Maria, 1000 Roraima Avenue, Santa Maria, RS 97105-900, Brazil
| | - Francini Franscescon
- Laboratory of Experimental Neuropsychobiology, Department of Biochemistry and Molecular Biology, Natural and Exact Sciences Center, Federal University of Santa Maria, 1000 Roraima Avenue, Santa Maria, RS 97105-900, Brazil; Graduate Program in Biological Sciences: Toxicological Biochemistry, Federal University of Santa Maria, 1000 Roraima Avenue, Santa Maria, RS 97105-900, Brazil
| | - Laura W Santos
- Laboratory of Experimental Neuropsychobiology, Department of Biochemistry and Molecular Biology, Natural and Exact Sciences Center, Federal University of Santa Maria, 1000 Roraima Avenue, Santa Maria, RS 97105-900, Brazil
| | - Isaac A Adedara
- Laboratory of Experimental Neuropsychobiology, Department of Biochemistry and Molecular Biology, Natural and Exact Sciences Center, Federal University of Santa Maria, 1000 Roraima Avenue, Santa Maria, RS 97105-900, Brazil; Drug Metabolism and Toxicology Research Laboratories, Department of Biochemistry, College of Medicine, University of Ibadan, Ibadan, Nigeria.
| | - Denis B Rosemberg
- Laboratory of Experimental Neuropsychobiology, Department of Biochemistry and Molecular Biology, Natural and Exact Sciences Center, Federal University of Santa Maria, 1000 Roraima Avenue, Santa Maria, RS 97105-900, Brazil; Graduate Program in Biological Sciences: Toxicological Biochemistry, Federal University of Santa Maria, 1000 Roraima Avenue, Santa Maria, RS 97105-900, Brazil; The International Zebrafish Neuroscience Research Consortium (ZNRC), 309 Palmer Court, Slidell, LA 70458, USA.
| |
Collapse
|
47
|
Leung RF, George AM, Roussel EM, Faux MC, Wigle JT, Eisenstat DD. Genetic Regulation of Vertebrate Forebrain Development by Homeobox Genes. Front Neurosci 2022; 16:843794. [PMID: 35546872 PMCID: PMC9081933 DOI: 10.3389/fnins.2022.843794] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Accepted: 03/14/2022] [Indexed: 01/19/2023] Open
Abstract
Forebrain development in vertebrates is regulated by transcription factors encoded by homeobox, bHLH and forkhead gene families throughout the progressive and overlapping stages of neural induction and patterning, regional specification and generation of neurons and glia from central nervous system (CNS) progenitor cells. Moreover, cell fate decisions, differentiation and migration of these committed CNS progenitors are controlled by the gene regulatory networks that are regulated by various homeodomain-containing transcription factors, including but not limited to those of the Pax (paired), Nkx, Otx (orthodenticle), Gsx/Gsh (genetic screened), and Dlx (distal-less) homeobox gene families. This comprehensive review outlines the integral role of key homeobox transcription factors and their target genes on forebrain development, focused primarily on the telencephalon. Furthermore, links of these transcription factors to human diseases, such as neurodevelopmental disorders and brain tumors are provided.
Collapse
Affiliation(s)
- Ryan F. Leung
- Murdoch Children’s Research Institute, The Royal Children’s Hospital Melbourne, Parkville, VIC, Australia
- Department of Paediatrics, University of Melbourne, Parkville, VIC, Australia
| | - Ankita M. George
- Murdoch Children’s Research Institute, The Royal Children’s Hospital Melbourne, Parkville, VIC, Australia
| | - Enola M. Roussel
- Murdoch Children’s Research Institute, The Royal Children’s Hospital Melbourne, Parkville, VIC, Australia
| | - Maree C. Faux
- Murdoch Children’s Research Institute, The Royal Children’s Hospital Melbourne, Parkville, VIC, Australia
- Department of Paediatrics, University of Melbourne, Parkville, VIC, Australia
- Department of Surgery, Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC, Australia
| | - Jeffrey T. Wigle
- Department of Biochemistry and Medical Genetics, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB, Canada
| | - David D. Eisenstat
- Murdoch Children’s Research Institute, The Royal Children’s Hospital Melbourne, Parkville, VIC, Australia
- Department of Paediatrics, University of Melbourne, Parkville, VIC, Australia
- Department of Medical Genetics, University of Alberta, Edmonton, AB, Canada
- Department of Pediatrics, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
48
|
Mezias C, Torok J, Maia PD, Markley E, Raj A. Matrix Inversion and Subset Selection (MISS): A pipeline for mapping of diverse cell types across the murine brain. Proc Natl Acad Sci U S A 2022; 119:e2111786119. [PMID: 35363567 PMCID: PMC9168512 DOI: 10.1073/pnas.2111786119] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Accepted: 12/14/2021] [Indexed: 11/21/2022] Open
Abstract
The advent of increasingly sophisticated imaging platforms has allowed for the visualization of the murine nervous system at single-cell resolution. However, current experimental approaches have not yet produced whole-brain maps of a comprehensive set of neuronal and nonneuronal types that approaches the cellular diversity of the mammalian cortex. Here, we aim to fill in this gap in knowledge with an open-source computational pipeline, Matrix Inversion and Subset Selection (MISS), that can infer quantitatively validated distributions of diverse collections of neural cell types at 200-μm resolution using a combination of single-cell RNA sequencing (RNAseq) and in situ hybridization datasets. We rigorously demonstrate the accuracy of MISS against literature expectations. Importantly, we show that gene subset selection, a procedure by which we filter out low-information genes prior to performing deconvolution, is a critical preprocessing step that distinguishes MISS from its predecessors and facilitates the production of cell-type maps with significantly higher accuracy. We also show that MISS is generalizable by generating high-quality cell-type maps from a second independently curated single-cell RNAseq dataset. Together, our results illustrate the viability of computational approaches for determining the spatial distributions of a wide variety of cell types from genetic data alone.
Collapse
Affiliation(s)
- Christopher Mezias
- Department of Radiology, University of California, San Francisco, CA 94143
| | - Justin Torok
- Department of Radiology, University of California, San Francisco, CA 94143
- Department of Radiology, Weill Cornell Medicine of Cornell University, New York, NY 10065
| | - Pedro D. Maia
- Department of Mathematics, University of Texas at Arlington, Arlington, TX 76019
| | - Eric Markley
- Department of Electrical Engineering and Computer Sciences, University of California, Berkeley, CA 94720
| | - Ashish Raj
- Department of Radiology, University of California, San Francisco, CA 94143
- Department of Radiology, Weill Cornell Medicine of Cornell University, New York, NY 10065
| |
Collapse
|
49
|
Ciancia M, Rataj-Baniowska M, Zinter N, Baldassarro VA, Fraulob V, Charles AL, Alvarez R, Muramatsu SI, de Lera AR, Geny B, Dollé P, Niewiadomska-Cimicka A, Krezel W. Retinoic acid receptor beta protects striatopallidal medium spiny neurons from mitochondrial dysfunction and neurodegeneration. Prog Neurobiol 2022; 212:102246. [DOI: 10.1016/j.pneurobio.2022.102246] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 12/07/2021] [Accepted: 02/04/2022] [Indexed: 12/26/2022]
|
50
|
Sulfasalazine exposure during pregnancy and lactation induces alterations in reproductive behavior in adult female rat offspring. Life Sci 2022; 293:120303. [PMID: 35051419 DOI: 10.1016/j.lfs.2022.120303] [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: 11/12/2021] [Revised: 12/23/2021] [Accepted: 01/04/2022] [Indexed: 11/21/2022]
Abstract
AIMS Sulfasalazine (SAS) is the first line drug in the treatment of chronic inflammatory bowel diseases in pregnant women. SAS and its metabolites cross the placenta and can be transferred through the milk. However, the long-term consequences to the reproductive system of offspring from dams exposed to SAS have not yet been studied. Thus, our study investigated the effects of SAS treatment during gestational and lactational periods on maternal care in F0 and reproductive outcomes in F1 females. MAIN METHODS Wistar female rats (n = 10/group) received 300 mg/kg/day of SAS dissolved in carboxymethyl cellulose (CMC), by gavage, from gestational day 0 to lactation day 21 and 3 mg/kg/day of folic acid during gestation. The control group received CMC only. On PND 21, the female pups were selected for reproductive evaluation at different time points: infancy and adulthood. The reproductive parameters evaluated were installation of puberty (vaginal opening and first estrus), estrous cyclicity, reproductive organs weight, histological analysis of the ovary follicles and uterus, analysis of oxidative stress in ovarian tissue, reproductive behavior (sexual and maternal), and fertility. KEY FINDINGS SAS treatment decreased the retrieving behavior in F0 females. The F1 females presented an increase in the lordosis score, frequency of lordosis of magnitude 3, and lipid peroxidation of ovarian tissues in both infancy and adult life. SIGNIFICANCE The SAS effects observed in the current study represent a relevant concern for public health, as they demonstrated that treatment with SAS compromised the maternal motivation of dams and induced reproductive alterations in F1 females.
Collapse
|