1
|
Li X, Lin X, Zhang Z, Zhuang Z, Li Y, Luo Y, Pan Y, Luo Q, Chen X. Neurotoxicity and aggressive behavior induced by anesthetic etomidate exposure in zebrafish: Insights from multi-omics and machine learning. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2025; 282:107321. [PMID: 40068374 DOI: 10.1016/j.aquatox.2025.107321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 03/03/2025] [Accepted: 03/07/2025] [Indexed: 04/05/2025]
Abstract
Etomidate (ETO), widely employed as a surgical anesthetic and more recently recognized as a drug of abuse, has been frequently detected in aquatic environment. However, the toxicity assessment of ETO is insufficient. Adult zebrafish were used to investigate toxicological effects of ETO. Four weeks ETO exposure could induced abnormal behaviors, including reduced anxiety, memory impairment, and heightened aggression. The increased aggression was quantitatively characterized using machine learning, which revealed significantly elevated instantaneous velocity and drastic changes in angular velocity. ETO was predominantly accumulated in the zebrafish brain, where it binds to GABA-A receptors, leading to a significant increase in GABA content. Furthermore, fluorescent staining of reactive oxygen species (ROS) in the brain revealed that ETO exposure significantly increased the oxidative stress level. This oxidative stress resulted in mitochondrial swelling, rupture, and damage to myelinated nerve fibers, ultimately causing cerebral injury in zebrafish. Multi-omics analysis further elucidated that ETO exposure down-regulated the MAPK signaling pathway, hyperactivated motor proteins, and induced metabolic disorders of lipids and amino acids. In summary, this study demonstrates that ETO induces neurotoxicity and behavioral alterations in zebrafish. These findings provide a critical insight into the mechanisms underlying ETO's neurotoxic effects and contribute to a more comprehensive understanding of its environmental and health risks.
Collapse
Affiliation(s)
- Xuewei Li
- Department of Forensic Toxicology, School of Forensic Medicine, Southern Medical University, Guangzhou 510515, China
| | - Xuhui Lin
- The Bartlett School of Sustainable Construction, University College London, London, UK
| | - Zheng Zhang
- Institute of Forensic Science XiangTan City Public Security Bureau, Xiangtan, China
| | - Zile Zhuang
- Department of Forensic Toxicology, School of Forensic Medicine, Southern Medical University, Guangzhou 510515, China
| | - Yihan Li
- Department of Forensic Toxicology, School of Forensic Medicine, Southern Medical University, Guangzhou 510515, China
| | - Yuxuan Luo
- Department of Forensic Toxicology, School of Forensic Medicine, Southern Medical University, Guangzhou 510515, China
| | - Yupeng Pan
- Department of Forensic Toxicology, School of Forensic Medicine, Southern Medical University, Guangzhou 510515, China
| | - Qizhi Luo
- Department of Forensic Toxicology, School of Forensic Medicine, Southern Medical University, Guangzhou 510515, China; Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou 510515, China.
| | - Xuncai Chen
- Department of Forensic Toxicology, School of Forensic Medicine, Southern Medical University, Guangzhou 510515, China; Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou 510515, China.
| |
Collapse
|
2
|
Sahay S, Lundh AE, Sirole RP, McCullumsmith RE, O’Donovan SM. Purinergic System Transcript Changes in the Dorsolateral Prefrontal Cortex in Suicide and Major Depressive Disorder. Int J Mol Sci 2025; 26:1826. [PMID: 40076453 PMCID: PMC11898938 DOI: 10.3390/ijms26051826] [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: 01/22/2025] [Revised: 02/16/2025] [Accepted: 02/17/2025] [Indexed: 03/14/2025] Open
Abstract
Suicide is a major public health priority, and its molecular mechanisms appear to be related to imbalanced purine metabolism in the brain. This exploratory study investigates purinergic gene expression in the postmortem dorsolateral prefrontal cortex (DLPFC) tissue isolated from subjects with major depressive disorder (MDD) who died by suicide (MDD-S, n = 10), MDD subjects who did not die by suicide (MDD-NS, n = 6) and non-psychiatrically ill controls (CTL, n = 9-10). Purinergic system transcripts were assayed by quantitative polymerase chain reactions (qPCR) in superficial and deep gray matter as well as white matter DLPFC cortical layers using laser microdissection (LMD). Across all subjects, regardless of sex, P2RY12 (F(2,23) = 5.40, p = 0.004) and P2RY13 (KW statistic = 11.82, p = 0.001) transcript levels were significantly greater in MDD-S compared to MDD-NS subjects. Several other perturbations were observed in the white matter tissue isolated from females: NT5E (F(2,10) = 13.37, p = 0.001) and P2RY13 (F(2,9) = 3.99, p = 0.011, controlled for age) transcript expression was significantly greater in MDD-S vs. MDD-NS female groups. ENTPD2 (F(2,10) = 5.20, p = 0.03), ENTPD3 (F(2,10) = 28.99, p < 0.0001), and NT5E (F(2,10) = 13.37, p = 0.001) were among the transcripts whose expression was significantly elevated in MDD-S vs. CTL female groups. Transcripts that exhibited significantly altered expression in the superficial and deep gray matter included ENTPD2, NT5E, PANX1, and P2RY13 (p ≤ 0.05). Our medication analysis revealed that the expression of these transcripts was not significantly altered by antidepressants. This is the first study to holistically quantify the purinergic metabolic pathway transcripts in suicide and MDD utilizing human postmortem brain tissue. Our preliminary findings support evidence implicating changes in purinergic P2 receptors in the brain in suicide and provide support for broader purinergic system dysregulation in mood disorders.
Collapse
Affiliation(s)
- Smita Sahay
- Department of Neurosciences and Psychiatry, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA; (S.S.)
| | - Anna E. Lundh
- Department of Neurosciences and Psychiatry, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA; (S.S.)
| | - Roshan P. Sirole
- Department of Neurosciences and Psychiatry, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA; (S.S.)
| | - Robert E. McCullumsmith
- Department of Neurosciences and Psychiatry, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA; (S.S.)
- Neurosciences Institute, ProMedica, Toledo, OH 43606, USA
| | - Sinead M. O’Donovan
- Department of Neurosciences and Psychiatry, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA; (S.S.)
- Department of Biological Sciences, University of Limerick, Castletroy, V94 T9PX Limerick, Ireland
| |
Collapse
|
3
|
Dos Santos B, Piermartiri T, Tasca CI. The impact of purine nucleosides on neuroplasticity in the adult brain. Purinergic Signal 2025; 21:113-131. [PMID: 38367178 PMCID: PMC11958884 DOI: 10.1007/s11302-024-09988-9] [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: 10/02/2023] [Accepted: 01/23/2024] [Indexed: 02/19/2024] Open
Abstract
Neuroplasticity refers to the nervous system's ability to adapt and reorganize its cell structures and neuronal networks in response to internal and external stimuli. In adults, this process involves neurogenesis, synaptogenesis, and synaptic and neurochemical plasticity. Several studies have reported the significant impact of the purinergic system on neuroplasticity modulation. And, there is considerable evidence supporting the role of purine nucleosides, such as adenosine, inosine, and guanosine, in this process. This review presents extensive research on how these nucleosides enhance the neuroplasticity of the adult central nervous system, particularly in response to damage. The mechanisms through which these nucleosides exert their effects involve complex interactions with various receptors and signaling pathways. Adenosine's influence on neurogenesis involves interactions with adenosine receptors, specifically A1R and A2AR. A1R activation appears to inhibit neuronal differentiation and promote astrogliogenesis, while A2AR activation supports neurogenesis, neuritogenesis, and synaptic plasticity. Inosine and guanosine positively impact cell proliferation, neurogenesis, and neuritogenesis. Inosine seems to modulate extracellular adenosine levels, and guanosine might act through interactions between purinergic and glutamatergic systems. Additionally, the review discusses the potential therapeutic implications of purinergic signaling in neurodegenerative and neuropsychiatric diseases, emphasizing the importance of these nucleosides in the neuroplasticity of brain function and recovery.
Collapse
Affiliation(s)
- Beatriz Dos Santos
- Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, SC, 88040-900, Brazil
- Programa de Pós-Graduação Em Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
| | - Tetsade Piermartiri
- Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, SC, 88040-900, Brazil.
- Programa de Pós-Graduação Em Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil.
| | - Carla I Tasca
- Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, SC, 88040-900, Brazil.
- Programa de Pós-Graduação Em Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil.
- Programa de Pós-Graduação Em Neurociências, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil.
| |
Collapse
|
4
|
Leavy A, Phelan J, Jimenez-Mateos EM. Contribution of microglia to the epileptiform activity that results from neonatal hypoxia. Neuropharmacology 2024; 253:109968. [PMID: 38692453 DOI: 10.1016/j.neuropharm.2024.109968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 04/13/2024] [Accepted: 04/22/2024] [Indexed: 05/03/2024]
Abstract
Microglia are described as the immune cells of the brain, their immune properties have been extensively studied since first described, however, their neural functions have only been explored over the last decade. Microglia have an important role in maintaining homeostasis in the central nervous system by surveying their surroundings to detect pathogens or damage cells. While these are the classical functions described for microglia, more recently their neural functions have been defined; they are critical to the maturation of neurons during embryonic and postnatal development, phagocytic microglia remove excess synapses during development, a process called synaptic pruning, which is important to overall neural maturation. Furthermore, microglia can respond to neuronal activity and, together with astrocytes, can regulate neural activity, contributing to the equilibrium between excitation and inhibition through a feedback loop. Hypoxia at birth is a serious neurological condition that disrupts normal brain function resulting in seizures and epilepsy later in life. Evidence has shown that microglia may contribute to this hyperexcitability after neonatal hypoxia. This review will summarize the existing data on the role of microglia in the pathogenesis of neonatal hypoxia and the plausible mechanisms that contribute to the development of hyperexcitability after hypoxia in neonates. This article is part of the Special Issue on "Microglia".
Collapse
Affiliation(s)
- Aisling Leavy
- Discipline of Physiology, School of Medicine, Trinity College Dublin, The University of Dublin, Dublin, Ireland
| | - Jessie Phelan
- Discipline of Physiology, School of Medicine, Trinity College Dublin, The University of Dublin, Dublin, Ireland
| | - Eva M Jimenez-Mateos
- Discipline of Physiology, School of Medicine, Trinity College Dublin, The University of Dublin, Dublin, Ireland.
| |
Collapse
|
5
|
Gao Z, Guan J, Yin S, Liu F. The role of ATP in sleep-wake regulation: In adenosine-dependent and -independent manner. Sleep Med 2024; 119:147-154. [PMID: 38678758 DOI: 10.1016/j.sleep.2024.04.031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 03/31/2024] [Accepted: 04/23/2024] [Indexed: 05/01/2024]
Abstract
ATP plays a crucial role as an energy currency in the body's various physiological functions, including the regulation of the sleep-wake cycle. Evidence from genetics and pharmacology demonstrates a strong association between ATP metabolism and sleep. With the advent of new technologies such as optogenetics, genetically encoded biosensors, and novel ATP detection methods, the dynamic changes in ATP levels between different sleep states have been further uncovered. The classic mechanism for regulating sleep by ATP involves its conversion to adenosine, which increases sleep pressure when accumulated extracellularly. However, emerging evidence suggests that ATP can directly bind to P2 receptors and influence sleep-wake regulation through both adenosine-dependent and independent pathways. The outcome depends on the brain region where ATP acts and the expression type of P2 receptors. This review summarizes the experimental evidence on the relationship between ATP levels and changes in sleep states and outlines the mechanisms by which ATP is involved in regulating the sleep-wake cycle through both adenosine-dependent and independent pathways. Hopefully, this review will provide a comprehensive understanding of the current research basis and progress in this field and promote further investigations into the specific mechanisms of ATP in regulating sleep.
Collapse
Affiliation(s)
- Zhenfei Gao
- Shanghai Key Laboratory of Sleep Disordered Breathing, Department of Otolaryngology-Head and Neck Surgery, Otolaryngology Institute of Shanghai Jiaotong University, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Jian Guan
- Shanghai Key Laboratory of Sleep Disordered Breathing, Department of Otolaryngology-Head and Neck Surgery, Otolaryngology Institute of Shanghai Jiaotong University, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China.
| | - Shankai Yin
- Shanghai Key Laboratory of Sleep Disordered Breathing, Department of Otolaryngology-Head and Neck Surgery, Otolaryngology Institute of Shanghai Jiaotong University, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China.
| | - Feng Liu
- Shanghai Key Laboratory of Sleep Disordered Breathing, Department of Otolaryngology-Head and Neck Surgery, Otolaryngology Institute of Shanghai Jiaotong University, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China.
| |
Collapse
|
6
|
Wikarska A, Roszak K, Roszek K. Mesenchymal Stem Cells and Purinergic Signaling in Autism Spectrum Disorder: Bridging the Gap between Cell-Based Strategies and Neuro-Immune Modulation. Biomedicines 2024; 12:1310. [PMID: 38927517 PMCID: PMC11201695 DOI: 10.3390/biomedicines12061310] [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: 04/28/2024] [Revised: 05/26/2024] [Accepted: 06/11/2024] [Indexed: 06/28/2024] Open
Abstract
The prevalence of autism spectrum disorder (ASD) is still increasing, which means that this neurodevelopmental lifelong pathology requires special scientific attention and efforts focused on developing novel therapeutic approaches. It has become increasingly evident that neuroinflammation and dysregulation of neuro-immune cross-talk are specific hallmarks of ASD, offering the possibility to treat these disorders by factors modulating neuro-immunological interactions. Mesenchymal stem cell-based therapy has already been postulated as one of the therapeutic approaches for ASD; however, less is known about the molecular mechanisms of stem cell influence. One of the possibilities, although still underestimated, is the paracrine purinergic activity of MSCs, by which stem cells ameliorate inflammatory reactions. Modulation of adenosine signaling may help restore neurotransmitter balance, reduce neuroinflammation, and improve overall brain function in individuals with ASD. In our review article, we present a novel insight into purinergic signaling, including but not limited to the adenosinergic pathway and its role in neuroinflammation and neuro-immune cross-talk modulation. We anticipate that by achieving a greater understanding of the purinergic signaling contribution to ASD and related disorders, novel therapeutic strategies may be devised for patients with autism in the near future.
Collapse
Affiliation(s)
| | | | - Katarzyna Roszek
- Department of Biochemistry, Faculty of Biological and Veterinary Sciences, Nicolaus Copernicus University in Torun, Lwowska 1, 87-100 Torun, Poland; (A.W.); (K.R.)
| |
Collapse
|
7
|
Pereira ADS, Bottari NB, Nauderer JN, Assmann CE, Copetti PM, Reichert KP, Mostardeiro VB, da Silveira MV, Morsch VMM, Schetinger MRC. Purinergic signaling influences the neuroinflammatory outcomes of a testosterone-derived synthetic in female rats: Resistance training protective effects on brain health. Steroids 2024; 203:109352. [PMID: 38128896 DOI: 10.1016/j.steroids.2023.109352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 12/06/2023] [Accepted: 12/18/2023] [Indexed: 12/23/2023]
Abstract
Physical exercise is recognized as a non-pharmacological approach to treat and protect against several neuroinflammatory conditions and thus to prevent brain disorders. However, the interest in ergogenic resources by athletes and bodybuilding practitioners is widespread and on the rise. These substances shorten the process of performance gain and improve aesthetics, having led to the prominent use and abuse of hormones in the past years. Recent evidence has shown that the purinergic system, composed of adenine nucleotides, nucleosides, enzymes, and receptors, participates in a wide range of processes within the brain, such as neuroinflammation, neuromodulation, and cellular communication. Here, we investigated the effects of the anabolic androgenic steroid (AAS) testosterone (TES) at a dose of 70 mg/kg/week in female rats and the neuroprotective effect of resistance exercise related to the purinergic system and oxidative stress parameters. Our findings showed a decrease in ATP and ADO hydrolysis in treated and trained animals. Furthermore, there was an increase in the density of purinoceptors (P2X7 and A2A) and inflammatory markers (IBA-1, NRLP3, CASP-1, IL-1β, and IL-6) in the cerebral cortex of animals that received AAS. On the other hand, exercise reversed neuroinflammatory parameters such as IBA-1, NLRP3, CASP-1, and IL-1β and improved antioxidant response and anti-inflammatory IL-10 cytokine levels. Overall, this study shows that the use of TES without indication or prescription disrupts brain homeostasis, as demonstrated by the increase in neuroinflammation, and that the practice of exercise can protect brain health.
Collapse
Affiliation(s)
- Aline da Silva Pereira
- Graduate Program in Toxicological Biochemistry, Department of Biochemistry and Molecular Biology, Federal University of Santa Maria (UFSM), Santa Maria, RS, Brazil.
| | - Nathieli Bianchin Bottari
- Graduate Program in Toxicological Biochemistry, Department of Biochemistry and Molecular Biology, Federal University of Santa Maria (UFSM), Santa Maria, RS, Brazil
| | - Jelson Norberto Nauderer
- Graduate Program in Toxicological Biochemistry, Department of Biochemistry and Molecular Biology, Federal University of Santa Maria (UFSM), Santa Maria, RS, Brazil
| | - Charles Elias Assmann
- Graduate Program in Toxicological Biochemistry, Department of Biochemistry and Molecular Biology, Federal University of Santa Maria (UFSM), Santa Maria, RS, Brazil
| | - Priscila Marquezan Copetti
- Graduate Program in Toxicological Biochemistry, Department of Biochemistry and Molecular Biology, Federal University of Santa Maria (UFSM), Santa Maria, RS, Brazil
| | - Karine Paula Reichert
- Graduate Program in Toxicological Biochemistry, Department of Biochemistry and Molecular Biology, Federal University of Santa Maria (UFSM), Santa Maria, RS, Brazil
| | - Vitor Bastianello Mostardeiro
- Graduate Program in Toxicological Biochemistry, Department of Biochemistry and Molecular Biology, Federal University of Santa Maria (UFSM), Santa Maria, RS, Brazil
| | - Marcylene Vieira da Silveira
- Graduate Program in Toxicological Biochemistry, Department of Biochemistry and Molecular Biology, Federal University of Santa Maria (UFSM), Santa Maria, RS, Brazil
| | - Vera Maria Melchiors Morsch
- Graduate Program in Toxicological Biochemistry, Department of Biochemistry and Molecular Biology, Federal University of Santa Maria (UFSM), Santa Maria, RS, Brazil
| | - Maria Rosa Chitolina Schetinger
- Graduate Program in Toxicological Biochemistry, Department of Biochemistry and Molecular Biology, Federal University of Santa Maria (UFSM), Santa Maria, RS, Brazil.
| |
Collapse
|
8
|
Ramakrishnan P, Joshi A, Fazil M, Yadav P. A comprehensive review on therapeutic potentials of photobiomodulation for neurodegenerative disorders. Life Sci 2024; 336:122334. [PMID: 38061535 DOI: 10.1016/j.lfs.2023.122334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Revised: 12/02/2023] [Accepted: 12/04/2023] [Indexed: 12/24/2023]
Abstract
A series of experimental trials over the past two centuries has put forth Photobiomodulation (PBM) as a treatment modality that utilizes colored lights for various conditions. While in its cradle, PBM was used for treating simple conditions such as burns and wounds, advancements in recent years have extended the use of PBM for treating complex neurodegenerative diseases (NDDs). PBM has exhibited the potential to curb several symptoms and signs associated with NDDs. While several of the currently used therapeutics cause adverse side effects alongside being highly invasive, PBM on the contrary, seems to be broad-acting, less toxic, and non-invasive. Despite being projected as an ideal therapeutic for NDDs, PBM still isn't considered a mainstream treatment modality due to some of the challenges and knowledge gaps associated with it. Here, we review the advantages of PBM summarized above with an emphasis on the common mechanisms that underlie major NDDs and how PBM helps tackle them. We also discuss important questions such as whether PBM should be considered a mainstay treatment modality for these conditions and if PBM's properties can be harnessed to develop prophylactic therapies for high-risk individuals and also highlight important animal studies that underscore the importance of PBM and the challenges associated with it. Overall, this review is intended to bring the major advances made in the field to the spotlight alongside addressing the practicalities and caveats to develop PBM as a major therapeutic for NDDs.
Collapse
Affiliation(s)
- Pooja Ramakrishnan
- Fly Laboratory # 210, Anusandhan Kendra-II, School of Chemical & Biotechnology, SASTRA Deemed to be University, Thanjavur 613401, Tamil Nadu, India.
| | - Aradhana Joshi
- Fly Laboratory # 210, Anusandhan Kendra-II, School of Chemical & Biotechnology, SASTRA Deemed to be University, Thanjavur 613401, Tamil Nadu, India.
| | - Mohamed Fazil
- Fly Laboratory # 210, Anusandhan Kendra-II, School of Chemical & Biotechnology, SASTRA Deemed to be University, Thanjavur 613401, Tamil Nadu, India; School of Chemical & Biotechnology, SASTRA Deemed to be University, Thanjavur 613401, Tamil Nadu, India
| | - Pankaj Yadav
- Fly Laboratory # 210, Anusandhan Kendra-II, School of Chemical & Biotechnology, SASTRA Deemed to be University, Thanjavur 613401, Tamil Nadu, India.
| |
Collapse
|
9
|
Zaib S, Areeba, Khan I. Purinergic Signaling and its Role in the Stem Cell Differentiation. Mini Rev Med Chem 2024; 24:863-883. [PMID: 37828668 DOI: 10.2174/0113895575261206231003151416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Revised: 07/30/2023] [Accepted: 08/30/2023] [Indexed: 10/14/2023]
Abstract
Purinergic signaling is a mechanism in which extracellular purines and pyrimidines interact with specialized cell surface receptors known as purinergic receptors. These receptors are divided into two families of P1 and P2 receptors, each responding to different nucleosides and nucleotides. P1 receptors are activated by adenosine, while P2 receptors are activated by pyrimidine and purines. P2X receptors are ligand-gated ion channels, including seven subunits (P2X1-7). However, P2Y receptors are the G-protein coupled receptors comprising eight subtypes (P2Y1/2/4/6/11/12/13/14). The disorder in purinergic signaling leads to various health-related issues and diseases. In various aspects, it influences the activity of non-neuronal cells and neurons. The molecular mechanism of purinergic signaling provides insight into treating various human diseases. On the contrary, stem cells have been investigated for therapeutic applications. Purinergic signaling has shown promising effect in stem cell engraftment. The immune system promotes the autocrine and paracrine mechanisms and releases the significant factors essential for successful stem cell therapy. Each subtype of purinergic receptor exerts a beneficial effect on the damaged tissue. The most common effect caused by purinergic signaling is the proliferation and differentiation that treat different health-related conditions.
Collapse
Affiliation(s)
- Sumera Zaib
- Department of Basic and Applied Chemistry, Faculty of Science and Technology, University of Central Punjab, Lahore, 54590, Pakistan
| | - Areeba
- Department of Basic and Applied Chemistry, Faculty of Science and Technology, University of Central Punjab, Lahore, 54590, Pakistan
| | - Imtiaz Khan
- Manchester Institute of Biotechnology, The University of Manchester, 131 Princess Street, Manchester, M1 7DN, United Kingdom
| |
Collapse
|
10
|
Mi X, Ni C, Zhao J, Amin N, Jiao D, Fang M, Ye X. P2Y12 receptor mediates apoptosis and demyelination to affect functional recovery in mice with spinal cord injury. Neurochem Int 2023; 171:105641. [PMID: 37952830 DOI: 10.1016/j.neuint.2023.105641] [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: 03/10/2023] [Revised: 09/28/2023] [Accepted: 11/06/2023] [Indexed: 11/14/2023]
Abstract
Among diseases of the central nervous system (CNS), spinal cord injury (SCI) has a high fatality rate. It has been proven that P2Y G protein-coupled purinergic receptors have a neuroprotective role in apoptosis and regeneration inside the damaged spinal cord. The P2Y12 receptor (P2Y12R) has recently been linked to peripheral neuropathy and stroke. However, the role of P2Y12R after SCI remains unclear. Our study randomly divided C57BL/6J female mice into 3 groups: Sham+DMSO, SCI+DMSO, and SCI+MRS2395. MRS2395 as a P2Y12R inhibitor was intraperitoneally injected at a dose of 1.5 mg/kg once daily for 7 days. We showed that the P2Y12R was markedly activated after injury, and it was double labeled with the microglial and neuron. Behavioral tests were employed to assess motor function recovery. By using immunofluorescence staining, the NeuN expression level was detected. The morphology of neurons was observed by hematoxylin-eosin and Nissl staining. P2Y12R, Bax, GFAP, PCNA and calbindin expression levels were detected using Western blot. Meanwhile, mitochondria and myelin sheath were observed by transmission electron microscopy (TEM). Our findings demonstrated that MRS2395 significantly enhanced motor function induced by SCI and that was used to alleviate apoptosis and astrocyte scarring. NeuN positive cells in the SCI group were lower than in the therapy group, although Bax, GFAP, PCNA and calbindin expression levels were considerably higher. Moreover, following MRS2395 therapy, the histological damage was reversed. A notable improvement in myelin sheath and mitochondrial morphology was seen in the therapy group. Together, our findings indicate that activation of P2Y12R in damaged spinal cord may be a critical event and suggest that inhibition of P2Y12R might be a feasible therapeutic strategy for treating SCI.
Collapse
Affiliation(s)
- Xiaodan Mi
- Center for Rehabilitation Medicine, Rehabilitation & Sports Medicine Research Institute of Zhejiang Province, Department of Rehabilitation Medicine, Zhejiang Provincial People's Hospital Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Chengtao Ni
- Graduate School, Bengbu Medical College, Bengbu, Anhui, China
| | - Jingting Zhao
- Center for Rehabilitation Medicine, Rehabilitation & Sports Medicine Research Institute of Zhejiang Province, Department of Rehabilitation Medicine, Zhejiang Provincial People's Hospital Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Nashwa Amin
- Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China; Institute of System Medicine, Zhejiang University School of Medicine, Hangzhou, China; Department of Zoology, Faculty of Science, Aswan University, Egypt
| | - Dian Jiao
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, China
| | - Marong Fang
- Children's Hospital of Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China.
| | - Xiangming Ye
- Center for Rehabilitation Medicine, Rehabilitation & Sports Medicine Research Institute of Zhejiang Province, Department of Rehabilitation Medicine, Zhejiang Provincial People's Hospital Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China.
| |
Collapse
|
11
|
Couto GT, Caetano HA, da Silva GP, Rockenbach L, Silva JSD, Vianna MR, Da Silva RS. Functioning and Gene Expression of Adenosine A 1 Receptor During Zebrafish ( Danio rerio) Development. Zebrafish 2023; 20:210-220. [PMID: 37856674 DOI: 10.1089/zeb.2023.0002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2023] Open
Abstract
The A1 adenosine receptor is the most widely expressed P1 receptor in vertebrates, performing inhibitory tone of the nervous system. Increased levels of adenosine are crucial to promote tissue protection in threatening situations, such as convulsion and hypoxia. Zebrafish is an established model organism for studies on health and disease. In this study, we evaluated the functionality of A1 adenosine receptor through development of zebrafish (6-7-day-, 3-, 8-, and 24-month-old), assessing: (I) the effects of the agonist N6-cyclopenthyladenosine (CPA) over locomotor parameters, (II) the anticonvulsant properties of CPA and adenosine per se in the pentylenetetrazol-induced seizure, and (III) the gene expression of adora1b through development. CPA promoted decreased distance traveled in the highest concentrations/doses tested (larvae: 75 to 500 μM; adults: 20 mg.kg-1), altered mean velocity (larvae: 50-500 μM; adults: 20 mg.kg-1) and time in the bottom zone of apparatus (adults: decrease in 20 mg.kg-1). Adenosine increased the latency of the larvae to reach stage II at 5 and 10 μM. CPA anticonvulsant effect against convulsive stage II was reached at 75 μM, although it decreased basal locomotor activity in larvae. For adults, CPA 10 mg.kg-1 was effective as anticonvulsant without locomotory effects. Adenosine had minor anticonvulsant effects in the concentration tested (larvae: 5 and 10 μM). The level of gene expression of adora1b was stable in brain from adult animals (8- and 24-month-old animals). These results suggest that zebrafish has similar responses to CPA as mammals. To avoid confounding factors, such as locomotor effects, during any brain function investigation using A1 adenosine receptor as a target, the concentration below 75 μM or below the dose of 20 mg.kg-1 of CPA is ideal for zebrafish at larval and adult stages, respectively.
Collapse
Affiliation(s)
- Giovanna Trevisan Couto
- Laboratório de Neuroquímica e Psicofarmacologia, Escola de Ciências da Saúde e da Vida, Pontificia Universidade, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Brazil
| | - Higor Arruda Caetano
- Laboratório de Neuroquímica e Psicofarmacologia, Escola de Ciências da Saúde e da Vida, Pontificia Universidade, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Brazil
| | - Guilherme Pietro da Silva
- Laboratório de Neuroquímica e Psicofarmacologia, Escola de Ciências da Saúde e da Vida, Pontificia Universidade, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Brazil
| | - Liliana Rockenbach
- Programa de Pós-Graduação me Medician e Ciências da Saúde, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Brazil
| | - Jéssica Scheid da Silva
- Laboratório de Neuroquímica e Psicofarmacologia, Escola de Ciências da Saúde e da Vida, Pontificia Universidade, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Brazil
| | - Monica Ryff Vianna
- Laboratório de Biologia do Desenvolvimento do Sistema Nervoso, Escola de Ciências da Saúde e da Vida, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Brazil
| | - Rosane Souza Da Silva
- Programa de Pós-Graduação em Neurociências, Universidade Federal Fluminense, Niterói, Rio de Janeiro, Brazil
| |
Collapse
|
12
|
Severs LJ, Bush NE, Quina LA, Hidalgo-Andrade S, Burgraff NJ, Dashevskiy T, Shih AY, Baertsch NA, Ramirez JM. Purinergic signaling mediates neuroglial interactions to modulate sighs. Nat Commun 2023; 14:5300. [PMID: 37652903 PMCID: PMC10471608 DOI: 10.1038/s41467-023-40812-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 08/10/2023] [Indexed: 09/02/2023] Open
Abstract
Sighs prevent the collapse of alveoli in the lungs, initiate arousal under hypoxic conditions, and are an expression of sadness and relief. Sighs are periodically superimposed on normal breaths, known as eupnea. Implicated in the generation of these rhythmic behaviors is the preBötzinger complex (preBötC). Our experimental evidence suggests that purinergic signaling is necessary to generate spontaneous and hypoxia-induced sighs in a mouse model. Our results demonstrate that driving calcium increases in astrocytes through pharmacological methods robustly increases sigh, but not eupnea, frequency. Calcium imaging of preBötC slices corroborates this finding with an increase in astrocytic calcium upon application of sigh modulators, increasing intracellular calcium through g-protein signaling. Moreover, photo-activation of preBötC astrocytes is sufficient to elicit sigh activity, and this response is blocked with purinergic antagonists. We conclude that sighs are modulated through neuron-glia coupling in the preBötC network, where the distinct modulatory responses of neurons and glia allow for both rhythms to be independently regulated.
Collapse
Affiliation(s)
- Liza J Severs
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, 98101, USA.
- Department of Physiology and Biophysics, University of Washington, Seattle, WA, 98195, USA.
| | - Nicholas E Bush
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, 98101, USA
| | - Lely A Quina
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, 98101, USA
| | - Skyler Hidalgo-Andrade
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, 98101, USA
| | - Nicholas J Burgraff
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, 98101, USA
| | - Tatiana Dashevskiy
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, 98101, USA
| | - Andy Y Shih
- Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, Seattle, WA, 98101, USA
- Department of Pediatrics, University of Washington School of Medicine, Seattle, WA, 98195, USA
- Department of Bioengineering, University of Washington, Seattle, WA, 98195, USA
| | - Nathan A Baertsch
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, 98101, USA
- Department of Pediatrics, University of Washington School of Medicine, Seattle, WA, 98195, USA
| | - Jan-Marino Ramirez
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, 98101, USA.
- Department of Physiology and Biophysics, University of Washington, Seattle, WA, 98195, USA.
- Department of Pediatrics, University of Washington School of Medicine, Seattle, WA, 98195, USA.
- Department of Neurological Surgery, University of Washington School of Medicine, Seattle, WA, 98195, USA.
| |
Collapse
|
13
|
Oliveros A, Poleschuk M, Cole PD, Boison D, Jang MH. Chemobrain: An accelerated aging process linking adenosine A 2A receptor signaling in cancer survivors. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2023; 170:267-305. [PMID: 37741694 PMCID: PMC10947554 DOI: 10.1016/bs.irn.2023.08.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/25/2023]
Abstract
Chemotherapy has a significant positive impact in cancer treatment outcomes, reducing recurrence and mortality. However, many cancer surviving children and adults suffer from aberrant chemotherapy neurotoxic effects on learning, memory, attention, executive functioning, and processing speed. This chemotherapy-induced cognitive impairment (CICI) is referred to as "chemobrain" or "chemofog". While the underlying mechanisms mediating CICI are still unclear, there is strong evidence that chemotherapy accelerates the biological aging process, manifesting as effects which include telomere shortening, epigenetic dysregulation, oxidative stress, mitochondrial defects, impaired neurogenesis, and neuroinflammation, all of which are known to contribute to increased anxiety and neurocognitive decline. Despite the increased prevalence of CICI, there exists a lack of mechanistic understanding by which chemotherapy detrimentally affects cognition in cancer survivors. Moreover, there are no approved therapeutic interventions for this condition. To address this gap in knowledge, this review attempts to identify how adenosine signaling, particularly through the adenosine A2A receptor, can be an essential tool to attenuate accelerated aging phenotypes. Importantly, the adenosine A2A receptor uniquely stands at the crossroads of cancer treatment and improved cognition, given that it is widely known to control tumor induced immunosuppression in the tumor microenvironment, while also posited to be an essential regulator of cognition in neurodegenerative disease. Consequently, we propose that the adenosine A2A receptor may provide a multifaceted therapeutic strategy to enhance anticancer activity, while combating chemotherapy induced cognitive deficits, both which are essential to provide novel therapeutic interventions against accelerated aging in cancer survivors.
Collapse
Affiliation(s)
- Alfredo Oliveros
- Department of Neurosurgery, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, Piscataway, NJ, United States
| | - Michael Poleschuk
- Department of Neurosurgery, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, Piscataway, NJ, United States
| | - Peter D Cole
- Division of Pediatric Hematology/Oncology, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, USA
| | - Detlev Boison
- Department of Neurosurgery, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, Piscataway, NJ, United States.
| | - Mi-Hyeon Jang
- Department of Neurosurgery, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, Piscataway, NJ, United States.
| |
Collapse
|
14
|
Rimbert S, Moreira JB, Xapelli S, Lévi S. Role of purines in brain development, from neuronal proliferation to synaptic refinement. Neuropharmacology 2023:109640. [PMID: 37348675 DOI: 10.1016/j.neuropharm.2023.109640] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Revised: 06/15/2023] [Accepted: 06/19/2023] [Indexed: 06/24/2023]
Abstract
The purinergic system includes P1 and P2 receptors, which are activated by ATP and its metabolites. They are expressed in adult neuronal and glial cells and are crucial in brain function, including neuromodulation and neuronal signaling. As P1 and P2 receptors are expressed throughout embryogenesis and development, purinergic signaling also has an important role in the development of the peripheral and central nervous system. In this review, we present the expression pattern and activity of purinergic receptors and of their signaling pathways during embryonic and postnatal development of the nervous system. In particular, we review the involvement of the purinergic signaling in all the crucial steps of brain development i.e. in neural stem cell proliferation, neuronal differentiation and migration as well as in astrogliogenesis and oligodendrogenesis. Then, we review data showing a crucial role of the ATP and adenosine signaling pathways in the formation of the peripheral neuromuscular junction and of central GABAergic and glutamatergic synapses. Finally, we examine the consequences of deregulation of the purinergic system during development and discuss the therapeutic potential of targeting it at adult stage in diseases with reactivation of the ATP and adenosine pathway.
Collapse
Affiliation(s)
- Solen Rimbert
- INSERM UMR-S 1270, Sorbonne Université, Institut du Fer à Moulin, 75005, Paris, France
| | - João B Moreira
- INSERM UMR-S 1270, Sorbonne Université, Institut du Fer à Moulin, 75005, Paris, France; Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal; Instituto de Medicina Molecular - João Lobo Antunes (iMM - JLA), Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Sara Xapelli
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal; Instituto de Medicina Molecular - João Lobo Antunes (iMM - JLA), Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Sabine Lévi
- INSERM UMR-S 1270, Sorbonne Université, Institut du Fer à Moulin, 75005, Paris, France.
| |
Collapse
|
15
|
Boccazzi M, Raffaele S, Zanettin T, Abbracchio MP, Fumagalli M. Altered Purinergic Signaling in Neurodevelopmental Disorders: Focus on P2 Receptors. Biomolecules 2023; 13:biom13050856. [PMID: 37238724 DOI: 10.3390/biom13050856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 05/09/2023] [Accepted: 05/16/2023] [Indexed: 05/28/2023] Open
Abstract
With the umbrella term 'neurodevelopmental disorders' (NDDs) we refer to a plethora of congenital pathological conditions generally connected with cognitive, social behavior, and sensory/motor alterations. Among the possible causes, gestational and perinatal insults have been demonstrated to interfere with the physiological processes necessary for the proper development of fetal brain cytoarchitecture and functionality. In recent years, several genetic disorders caused by mutations in key enzymes involved in purine metabolism have been associated with autism-like behavioral outcomes. Further analysis revealed dysregulated purine and pyrimidine levels in the biofluids of subjects with other NDDs. Moreover, the pharmacological blockade of specific purinergic pathways reversed the cognitive and behavioral defects caused by maternal immune activation, a validated and now extensively used rodent model for NDDs. Furthermore, Fragile X and Rett syndrome transgenic animal models as well as models of premature birth, have been successfully utilized to investigate purinergic signaling as a potential pharmacological target for these diseases. In this review, we examine results on the role of the P2 receptor signaling in the etiopathogenesis of NDDs. On this basis, we discuss how this evidence could be exploited to develop more receptor-specific ligands for future therapeutic interventions and novel prognostic markers for the early detection of these conditions.
Collapse
Affiliation(s)
- Marta Boccazzi
- Laboratory of Molecular and Cellular Pharmacology of Purinergic Transmission, Department of Pharmaceutical Sciences, Università Degli Studi di Milano, 20133 Milan, Italy
| | - Stefano Raffaele
- Laboratory of Molecular and Cellular Pharmacology of Purinergic Transmission, Department of Pharmacological and Biomolecular Sciences, Università Degli Studi di Milano, 20133 Milan, Italy
| | - Thomas Zanettin
- Laboratory of Molecular and Cellular Pharmacology of Purinergic Transmission, Department of Pharmacological and Biomolecular Sciences, Università Degli Studi di Milano, 20133 Milan, Italy
| | - Maria P Abbracchio
- Laboratory of Molecular and Cellular Pharmacology of Purinergic Transmission, Department of Pharmaceutical Sciences, Università Degli Studi di Milano, 20133 Milan, Italy
| | - Marta Fumagalli
- Laboratory of Molecular and Cellular Pharmacology of Purinergic Transmission, Department of Pharmacological and Biomolecular Sciences, Università Degli Studi di Milano, 20133 Milan, Italy
| |
Collapse
|
16
|
Ribeiro DE, Petiz LL, Glaser T, Oliveira-Giacomelli Á, Andrejew R, Saab FDAR, Milanis MDS, Campos HC, Sampaio VFA, La Banca S, Longo BM, Lameu C, Tang Y, Resende RR, Ferreira ST, Ulrich H. Purinergic signaling in cognitive impairment and neuropsychiatric symptoms of Alzheimer's disease. Neuropharmacology 2023; 226:109371. [PMID: 36502867 DOI: 10.1016/j.neuropharm.2022.109371] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 12/04/2022] [Accepted: 12/05/2022] [Indexed: 12/13/2022]
Abstract
About 10 million new cases of dementia develop worldwide each year, of which up to 70% are attributable to Alzheimer's disease (AD). In addition to the widely known symptoms of memory loss and cognitive impairment, AD patients frequently develop non-cognitive symptoms, referred to as behavioral and psychological symptoms of dementia (BPSDs). Sleep disorders are often associated with AD, but mood alterations, notably depression and apathy, comprise the most frequent class of BPSDs. BPSDs negatively affect the lives of AD patients and their caregivers, and have a significant impact on public health systems and the economy. Because treatments currently available for AD are not disease-modifying and mainly aim to ameliorate some of the cognitive symptoms, elucidating the mechanisms underlying mood alterations and other BPSDs in AD may reveal novel avenues for progress in AD therapy. Purinergic signaling is implicated in the pathophysiology of several central nervous system (CNS) disorders, such as AD, depression and sleep disorders. Here, we review recent findings indicating that purinergic receptors, mainly the A1, A2A, and P2X7 subtypes, are associated with the development/progression of AD. Current evidence suggests that targeting purinergic signaling may represent a promising therapeutic approach in AD and related conditions. This article is part of the Special Issue on "Purinergic Signaling: 50 years".
Collapse
Affiliation(s)
- Deidiane Elisa Ribeiro
- Department of Biochemistry, Chemistry Institute, University of São Paulo (USP), São Paulo, Brazil.
| | - Lyvia Lintzmaier Petiz
- Department of Biochemistry and Molecular Biology, Universidade Federal do Paraná, Curitiba, Brazil
| | - Talita Glaser
- Department of Biochemistry, Chemistry Institute, University of São Paulo (USP), São Paulo, Brazil
| | | | - Roberta Andrejew
- Department of Biochemistry, Chemistry Institute, University of São Paulo (USP), São Paulo, Brazil
| | | | - Milena da Silva Milanis
- Department of Biochemistry, Chemistry Institute, University of São Paulo (USP), São Paulo, Brazil
| | - Henrique Correia Campos
- Laboratory of Neurophysiology, Department of Physiology, Federal University of São Paulo (UNIFESP), São Paulo, Brazil
| | | | - Sophia La Banca
- Department of Biochemistry, Chemistry Institute, University of São Paulo (USP), São Paulo, Brazil
| | - Beatriz Monteiro Longo
- Laboratory of Neurophysiology, Department of Physiology, Federal University of São Paulo (UNIFESP), São Paulo, Brazil
| | - Claudiana Lameu
- Department of Biochemistry, Chemistry Institute, University of São Paulo (USP), São Paulo, Brazil
| | - Yong Tang
- International Collaborative Centre on Big Science Plan for Purinergic Signalling, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China; Acupuncture and Chronobiology Key Laboratory of Sichuan Province, Chengdu, 610075, China
| | - Rodrigo Ribeiro Resende
- Department of Biochemistry and Immunology, Federal University of Minas Gerais Belo Horizonte, MG, Brazil
| | - Sergio T Ferreira
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil; Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Henning Ulrich
- Department of Biochemistry, Chemistry Institute, University of São Paulo (USP), São Paulo, Brazil; International Collaborative Centre on Big Science Plan for Purinergic Signalling, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China.
| |
Collapse
|
17
|
Luhmann HJ. Malformations-related neocortical circuits in focal seizures. Neurobiol Dis 2023; 178:106018. [PMID: 36706927 DOI: 10.1016/j.nbd.2023.106018] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 01/18/2023] [Accepted: 01/23/2023] [Indexed: 01/25/2023] Open
Abstract
This review article gives an overview on the molecular, cellular and network mechanisms underlying focal seizures in neocortical networks with developmental malformations. Neocortical malformations comprise a large variety of structural abnormalities associated with epilepsy and other neurological and psychiatric disorders. Genetic or acquired disorders of neocortical cell proliferation, neuronal migration and/or programmed cell death may cause pathologies ranging from the expression of dysmorphic neurons and heterotopic cell clusters to abnormal layering and cortical misfolding. After providing a brief overview on the pathogenesis and structure of neocortical malformations in humans, animal models are discussed and how they contributed to our understanding on the mechanisms of neocortical hyperexcitability associated with developmental disorders. State-of-the-art molecular biological and electrophysiological techniques have been also used in humans and on resectioned neocortical tissue of epileptic patients and provide deep insights into the subcellular, cellular and network mechanisms contributing to focal seizures. Finally, a brief outlook is given how novel models and methods can shape translational research in the near future.
Collapse
Affiliation(s)
- Heiko J Luhmann
- Institute of Physiology, University Medical Center of the Johannes Gutenberg University Mainz, Duesbergweg 6, Mainz, Germany.
| |
Collapse
|
18
|
Xu G, Zhang S, Zheng L, Hu Z, Cheng L, Chen L, Li J, Shi Z. In silico identification of A1 agonists and A2a inhibitors in pain based on molecular docking strategies and dynamics simulations. Purinergic Signal 2023; 19:87-97. [PMID: 34677752 PMCID: PMC9984648 DOI: 10.1007/s11302-021-09808-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 07/05/2021] [Indexed: 12/19/2022] Open
Abstract
Most recently, the adenosine is considered as one of the most promising targets for treating pain, with few side effects. It exists in the central nervous system, and plays a key role in nociceptive afferent pathway. It is reported that the A1 receptor (A1R) could inhibit Ca2+ channels to reduce the pain like analgesic mechanism of morphine. And, A2a receptor (A2aR) was reported to enhance the accumulation of AMP (cAMP) and released peptides from sensory neurons, resulting in constitutive activation of pain. Much evidence showed that A1R and A2aR could be served as the interesting targets for the treatment of pain. Herein, virtual screening was utilized to identify the small molecule compounds towards A1R and A2aR, and top six molecules were considered as candidates via amber scores. The molecular dynamic (MD) simulations and molecular mechanics/generalized born surface area (MM/GBSA) were employed to further analyze the affinity and binding stability of the six molecules towards A1R and A2aR. Moreover, energy decomposition analysis showed significant residues in A1R and A2aR, including His1383, Phe1276, and Glu1277. It provided basics for discovery of novel agonists and antagonists. Finally, the agonists of A1R (ZINC19943625, ZINC13555217, and ZINC04698406) and inhibitors of A2aR (ZINC19370372, ZINC20176051, and ZINC57263068) were successfully recognized. Taken together, our discovered small molecules may serve as the promising candidate agents for future pain research.
Collapse
Affiliation(s)
- Guangya Xu
- Clinical Genetics Laboratory, Clinical Medical College & Affiliated Hospital & College of Basic Medicine & College of Food and Biological Engineering, Chengdu University, Chengdu, 610081, China
| | - Shutao Zhang
- Clinical Genetics Laboratory, Clinical Medical College & Affiliated Hospital & College of Basic Medicine & College of Food and Biological Engineering, Chengdu University, Chengdu, 610081, China
| | - Lulu Zheng
- Clinical Genetics Laboratory, Clinical Medical College & Affiliated Hospital & College of Basic Medicine & College of Food and Biological Engineering, Chengdu University, Chengdu, 610081, China
| | - Zhongjiao Hu
- Clinical Genetics Laboratory, Clinical Medical College & Affiliated Hospital & College of Basic Medicine & College of Food and Biological Engineering, Chengdu University, Chengdu, 610081, China.,School of Pharmacy, Zunyi Medical University, Zunyi, 563000, China
| | - Lijia Cheng
- Clinical Genetics Laboratory, Clinical Medical College & Affiliated Hospital & College of Basic Medicine & College of Food and Biological Engineering, Chengdu University, Chengdu, 610081, China
| | - Lvlin Chen
- Clinical Genetics Laboratory, Clinical Medical College & Affiliated Hospital & College of Basic Medicine & College of Food and Biological Engineering, Chengdu University, Chengdu, 610081, China
| | - Jun Li
- Clinical Genetics Laboratory, Clinical Medical College & Affiliated Hospital & College of Basic Medicine & College of Food and Biological Engineering, Chengdu University, Chengdu, 610081, China. .,Sichuan Wuyan Biotech Co. Ltd Company, Chengdu, 610041, China.
| | - Zheng Shi
- Clinical Genetics Laboratory, Clinical Medical College & Affiliated Hospital & College of Basic Medicine & College of Food and Biological Engineering, Chengdu University, Chengdu, 610081, China. .,School of Pharmacy, Zunyi Medical University, Zunyi, 563000, China.
| |
Collapse
|
19
|
Cadoni C, Peana AT. Energy drinks at adolescence: Awareness or unawareness? Front Behav Neurosci 2023; 17:1080963. [PMID: 36891321 PMCID: PMC9986288 DOI: 10.3389/fnbeh.2023.1080963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 02/06/2023] [Indexed: 02/22/2023] Open
Abstract
Energy drinks (EDs) are beverages similar to soft drinks, characterized by high caffeine concentrations with additional ingredients like taurine and vitamins, marketed for boosting energy, reducing tiredness, increasing concentration, and for their ergogenic effect. The majority of consumers are children, adolescents, and young athletes. Although EDs companies claim about the ergogenic and remineralizing properties of their products, there is a serious lack of evidence at preclinical as well as clinical level to validate their benefits. The regular intake and long-term consequences of these caffeinated drinks are not well documented, especially the possible negative effects in adolescents whose brain is still developing. EDs combined with alcohol are also gaining popularity among adolescents and different publications indicate that this combined consumption might increase the risk to develop an alcohol use disorder, as well as produce serious adverse cardiovascular effects. There is an increasing need to disseminate knowledge on EDs damage on health, so that adolescents can be aware about the potential harmful outcomes of consuming these drinks.
Collapse
Affiliation(s)
- Cristina Cadoni
- Department of Biomedical Sciences, Institute of Neuroscience, National Research Council of Italy, Cagliari, Italy
| | | |
Collapse
|
20
|
Cserép C, Schwarcz AD, Pósfai B, László ZI, Kellermayer A, Környei Z, Kisfali M, Nyerges M, Lele Z, Katona I, Ádám Dénes. Microglial control of neuronal development via somatic purinergic junctions. Cell Rep 2022; 40:111369. [PMID: 36130488 PMCID: PMC9513806 DOI: 10.1016/j.celrep.2022.111369] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 06/28/2022] [Accepted: 08/25/2022] [Indexed: 11/30/2022] Open
Abstract
Microglia, the resident immune cells of the brain, play important roles during development. Although bi-directional communication between microglia and neuronal progenitors or immature neurons has been demonstrated, the main sites of interaction and the underlying mechanisms remain elusive. By using advanced methods, here we provide evidence that microglial processes form specialized contacts with the cell bodies of developing neurons throughout embryonic, early postnatal, and adult neurogenesis. These early developmental contacts are highly reminiscent of somatic purinergic junctions that are instrumental for microglia-neuron communication in the adult brain. The formation and maintenance of these junctions is regulated by functional microglial P2Y12 receptors, and deletion of P2Y12Rs disturbs proliferation of neuronal precursors and leads to aberrant cortical cytoarchitecture during development and in adulthood. We propose that early developmental formation of somatic purinergic junctions represents an important interface for microglia to monitor the status of immature neurons and control neurodevelopment.
Collapse
Affiliation(s)
- Csaba Cserép
- "Momentum" Laboratory of Neuroimmunology, Institute of Experimental Medicine, 1083 Budapest, Hungary.
| | - Anett D Schwarcz
- "Momentum" Laboratory of Neuroimmunology, Institute of Experimental Medicine, 1083 Budapest, Hungary
| | - Balázs Pósfai
- "Momentum" Laboratory of Neuroimmunology, Institute of Experimental Medicine, 1083 Budapest, Hungary; Szentágothai János Doctoral School of Neurosciences, Semmelweis University, 1083 Budapest, Hungary
| | - Zsófia I László
- "Momentum" Laboratory of Molecular Neurobiology, Institute of Experimental Medicine, 1083 Budapest, Hungary; University of Dundee, School of Medicine, Dundee DD1 9SY, UK
| | - Anna Kellermayer
- "Momentum" Laboratory of Neuroimmunology, Institute of Experimental Medicine, 1083 Budapest, Hungary
| | - Zsuzsanna Környei
- "Momentum" Laboratory of Neuroimmunology, Institute of Experimental Medicine, 1083 Budapest, Hungary
| | - Máté Kisfali
- "Momentum" Laboratory of Molecular Neurobiology, Institute of Experimental Medicine, 1083 Budapest, Hungary
| | - Miklós Nyerges
- "Momentum" Laboratory of Neuroimmunology, Institute of Experimental Medicine, 1083 Budapest, Hungary
| | - Zsolt Lele
- "Momentum" Laboratory of Molecular Neurobiology, Institute of Experimental Medicine, 1083 Budapest, Hungary
| | - István Katona
- "Momentum" Laboratory of Molecular Neurobiology, Institute of Experimental Medicine, 1083 Budapest, Hungary; Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN 47405, USA
| | - Ádám Dénes
- "Momentum" Laboratory of Neuroimmunology, Institute of Experimental Medicine, 1083 Budapest, Hungary.
| |
Collapse
|
21
|
Manojlovic-Stojanoski M, Lavrnja I, Stevanovic I, Trifunovic S, Ristic N, Nestorovic N, Sévigny J, Nedeljkovic N, Laketa D. Antenatal Dexamethasone Treatment Induces Sex-dependent Upregulation of NTPDase1/CD39 and Ecto-5'-nucleotidase/CD73 in the Rat Fetal Brain. Cell Mol Neurobiol 2022; 42:1965-1981. [PMID: 33761054 PMCID: PMC11421702 DOI: 10.1007/s10571-021-01081-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Accepted: 03/13/2021] [Indexed: 10/21/2022]
Abstract
Dexamethasone (DEX) is frequently used to treat women at risk of preterm delivery, but although indispensable for the completion of organ maturation in the fetus, antenatal DEX treatment may exert adverse sex-dimorphic neurodevelopmental effects. Literature findings implicated oxidative stress in adverse effects of DEX treatment. Purinergic signaling is involved in neurodevelopment and controlled by ectonucleotidases, among which in the brain the most abundant are ectonucleoside triphosphate diphosphohydrolase 1 (NTPDase1/CD39) and ecto-5'-nucleotidase (e5'NT/CD73), which jointly dephosphorylate ATP to adenosine. They are also involved in cell adhesion and migration, processes integral to brain development. Upregulation of CD39 and CD73 after DEX treatment was reported in adult rat hippocampus. We investigated the effects of maternal DEX treatment on CD39 and CD73 expression and enzymatic activity in the rat fetal brain of both sexes, in the context of oxidative status of the brain tissue. Fetuses were obtained at embryonic day (ED) 21, from Wistar rat dams treated with 0.5 mg DEX/kg/day, at ED 16, 17, and 18, and brains were processed and used for further analysis. Sex-specific increase in CD39 and CD73 expression and in the corresponding enzyme activities was induced in the brain of antenatally DEX-treated fetuses, more prominently in males. The oxidative stress induction after antenatal DEX treatment was confirmed in both sexes, although showing a slight bias in males. Due to the involvement of purinergic system in crucial neurodevelopmental processes, future investigations are needed to determine the role of these observed changes in the adverse effects of antenatal DEX treatment.
Collapse
Affiliation(s)
- Milica Manojlovic-Stojanoski
- Institute for Biological Research, "Siniša Stanković"- National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Irena Lavrnja
- Institute for Biological Research, "Siniša Stanković"- National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Ivana Stevanovic
- Medical Faculty of Military Medical Academy, Institute of Medical Research Belgrade, Belgrade, Serbia
| | - Svetlana Trifunovic
- Institute for Biological Research, "Siniša Stanković"- National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Natasa Ristic
- Institute for Biological Research, "Siniša Stanković"- National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Natasa Nestorovic
- Institute for Biological Research, "Siniša Stanković"- National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Jean Sévigny
- Département de Microbiologie-Infectiologie et d'immunologie, Faculté de Médecine, Université Laval, Québec, Canada
- Centre de recherche du CHU de Québec - Université Laval, Québec, Canada
| | - Nadezda Nedeljkovic
- Department for General Physiology and Biophysics, Faculty of Biology, University of Belgrade, Belgrade, Serbia
| | - Danijela Laketa
- Department for General Physiology and Biophysics, Faculty of Biology, University of Belgrade, Belgrade, Serbia.
| |
Collapse
|
22
|
Jacobson KA, Gao ZG, Matricon P, Eddy MT, Carlsson J. Adenosine A 2A receptor antagonists: from caffeine to selective non-xanthines. Br J Pharmacol 2022; 179:3496-3511. [PMID: 32424811 PMCID: PMC9251831 DOI: 10.1111/bph.15103] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Revised: 04/30/2020] [Accepted: 05/03/2020] [Indexed: 12/12/2022] Open
Abstract
A long evolution of knowledge of the psychostimulant caffeine led in the 1960s to another purine natural product, adenosine and its A2A receptor. Adenosine is a short-lived autocrine/paracrine mediator that acts pharmacologically at four different adenosine receptors in a manner opposite to the pan-antagonist caffeine and serves as an endogenous allostatic regulator. Although detrimental in the developing brain, caffeine appears to be cerebroprotective in aging. Moderate caffeine consumption in adults, except in pregnancy, may also provide benefit in pain, diabetes, and kidney and liver disorders. Inhibition of A2A receptors is one of caffeine's principal effects and we now understand this interaction at the atomic level. The A2A receptor has become a prototypical example of utilizing high-resolution structures of GPCRs for the rational design of chemically diverse drug molecules. The previous focus on discovery of selective A2A receptor antagonists for neurodegenerative diseases has expanded to include immunotherapy for cancer, and clinical trials have ensued. LINKED ARTICLES: This article is part of a themed issue on Structure Guided Pharmacology of Membrane Proteins (BJP 75th Anniversary). To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v179.14/issuetoc.
Collapse
Affiliation(s)
- Kenneth A. Jacobson
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Zhan-Guo Gao
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Pierre Matricon
- Department of Cell and Molecular Biology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Matthew T. Eddy
- Department of Chemistry, University of Florida, Gainesville, FL, USA
| | - Jens Carlsson
- Department of Cell and Molecular Biology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| |
Collapse
|
23
|
Luo SS, Zou KX, Zhu H, Cheng Y, Yan YS, Sheng JZ, Huang HF, Ding GL. Integrated Multi-Omics Analysis Reveals the Effect of Maternal Gestational Diabetes on Fetal Mouse Hippocampi. Front Cell Dev Biol 2022; 10:748862. [PMID: 35237591 PMCID: PMC8883435 DOI: 10.3389/fcell.2022.748862] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 01/19/2022] [Indexed: 11/25/2022] Open
Abstract
Growing evidence suggests that adverse intrauterine environments could affect the long-term health of offspring. Recent evidence indicates that gestational diabetes mellitus (GDM) is associated with neurocognitive changes in offspring. However, the mechanism remains unclear. Using a GDM mouse model, we collected hippocampi, the structure critical to cognitive processes, for electron microscopy, methylome and transcriptome analyses. Reduced representation bisulfite sequencing (RRBS) and RNA-seq in the GDM fetal hippocampi showed altered methylated modification and differentially expressed genes enriched in common pathways involved in neural synapse organization and signal transmission. We further collected fetal mice brains for metabolome analysis and found that in GDM fetal brains, the metabolites displayed significant changes, in addition to directly inducing cognitive dysfunction, some of which are important to methylation status such as betaine, fumaric acid, L-methionine, succinic acid, 5-methyltetrahydrofolic acid, and S-adenosylmethionine (SAM). These results suggest that GDM affects metabolites in fetal mice brains and further affects hippocampal DNA methylation and gene regulation involved in cognition, which is a potential mechanism for the adverse neurocognitive effects of GDM in offspring.
Collapse
Affiliation(s)
- Si-Si Luo
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, China
| | - Ke-Xin Zou
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, China
| | - Hong Zhu
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China.,Research Units of Embryo Original Diseases, Chinese Academy of Medical Sciences, Shanghai, China
| | - Yi Cheng
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China.,Research Units of Embryo Original Diseases, Chinese Academy of Medical Sciences, Shanghai, China
| | - Yi-Shang Yan
- The Key Laboratory of Reproductive Genetics (Zhejiang University), Ministry of Education, Hangzhou, China
| | - Jian-Zhong Sheng
- The Key Laboratory of Reproductive Genetics (Zhejiang University), Ministry of Education, Hangzhou, China
| | - He-Feng Huang
- Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, China.,Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China.,Research Units of Embryo Original Diseases, Chinese Academy of Medical Sciences, Shanghai, China.,The Key Laboratory of Reproductive Genetics (Zhejiang University), Ministry of Education, Hangzhou, China
| | - Guo-Lian Ding
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China.,Research Units of Embryo Original Diseases, Chinese Academy of Medical Sciences, Shanghai, China
| |
Collapse
|
24
|
Beamer E, O’Dea MI, Garvey AA, Smith J, Menéndez-Méndez A, Kelly L, Pavel A, Quinlan S, Alves M, Jimenez-Mateos EM, Tian F, Dempsey E, Dale N, Murray DM, Boylan GB, Molloy EJ, Engel T. Novel Point-of-Care Diagnostic Method for Neonatal Encephalopathy Using Purine Nucleosides. Front Mol Neurosci 2021; 14:732199. [PMID: 34566578 PMCID: PMC8458851 DOI: 10.3389/fnmol.2021.732199] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 08/19/2021] [Indexed: 01/19/2023] Open
Abstract
Background: Evidence suggests that earlier diagnosis and initiation of treatment immediately after birth is critical for improved neurodevelopmental outcomes following neonatal encephalopathy (NE). Current diagnostic tests are, however, mainly restricted to clinical diagnosis with no molecular tests available. Purines including adenosine are released during brain injury such as hypoxia and are also present in biofluids. Whether blood purine changes can be used to diagnose NE has not been investigated to date. Methods: Blood purines were measured in a mouse model of neonatal hypoxia and infants with NE using a novel point-of-care diagnostic technology (SMARTChip) based on the summated electrochemical detection of adenosine and adenosine metabolites in the blood. Results: Blood purine concentrations were ∼2-3-fold elevated following hypoxia in mice [2.77 ± 0.48 μM (Control) vs. 7.57 ± 1.41 μM (post-hypoxia), p = 0.029]. Data in infants with NE had a 2-3-fold elevation when compared to healthy controls [1.63 ± 0.47 μM (Control, N = 5) vs. 4.87 ± 0.92 μM (NE, N = 21), p = 0.0155]. ROC curve analysis demonstrates a high sensitivity (81%) and specificity (80%) for our approach to identify infants with NE. Moreover, blood purine concentrations were higher in infants with NE and seizures [8.13 ± 3.23 μM (with seizures, N = 5) vs. 3.86 ± 0.56 μM (without seizures, N = 16), p = 0.044]. Conclusion: Our data provides the proof-of-concept that measurement of blood purine concentrations via SMARTChip technology may offer a low-volume bedside test to support a rapid diagnosis of NE.
Collapse
Affiliation(s)
- Edward Beamer
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, University of Medicine and Health Sciences, Dublin, Ireland
- Centre for Bioscience, Manchester Metropolitan University, Manchester, United Kingdom
| | - Mary Isabel O’Dea
- Coombe Women and Infants University Hospital, Dublin, Ireland
- National Children’s Research Centre, Crumlin, Dublin, Ireland
- Discipline of Paediatrics, Children’s Health Ireland at Crumlin and Tallaght, Dublin, Ireland
- Trinity Research in Childhood Centre (TRiCC), Trinity College Dublin, The University of Dublin, Dublin, Ireland
| | - Aisling A. Garvey
- INFANT Research Centre, University College Cork, Dublin, Ireland
- Department of Paediatrics and Child Health, University College Cork, Dublin, Ireland
| | - Jonathon Smith
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, University of Medicine and Health Sciences, Dublin, Ireland
- FutureNeuro, Science Foundation Ireland Research Centre for Chronic and Rare Neurological Diseases, Royal College of Surgeons in Ireland, University of Medicine and Health Sciences, Dublin, Ireland
| | - Aida Menéndez-Méndez
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, University of Medicine and Health Sciences, Dublin, Ireland
| | - Lynne Kelly
- Coombe Women and Infants University Hospital, Dublin, Ireland
- Discipline of Paediatrics, Children’s Health Ireland at Crumlin and Tallaght, Dublin, Ireland
| | - Andreea Pavel
- INFANT Research Centre, University College Cork, Dublin, Ireland
- Department of Paediatrics and Child Health, University College Cork, Dublin, Ireland
| | - Sean Quinlan
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, University of Medicine and Health Sciences, Dublin, Ireland
| | - Mariana Alves
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, University of Medicine and Health Sciences, Dublin, Ireland
| | - Eva M. Jimenez-Mateos
- Discipline of Physiology, School of Medicine, Trinity College Dublin, The University of Dublin, Dublin, Ireland
| | - Faming Tian
- School of Life Sciences, University of Warwick, Coventry, United Kingdom
| | - Eugene Dempsey
- INFANT Research Centre, University College Cork, Dublin, Ireland
- Department of Paediatrics and Child Health, University College Cork, Dublin, Ireland
| | - Nicholas Dale
- School of Life Sciences, University of Warwick, Coventry, United Kingdom
| | - Deirdre M. Murray
- INFANT Research Centre, University College Cork, Dublin, Ireland
- Department of Paediatrics and Child Health, University College Cork, Dublin, Ireland
| | - Geraldine B. Boylan
- INFANT Research Centre, University College Cork, Dublin, Ireland
- Department of Paediatrics and Child Health, University College Cork, Dublin, Ireland
| | - Eleanor J. Molloy
- Coombe Women and Infants University Hospital, Dublin, Ireland
- National Children’s Research Centre, Crumlin, Dublin, Ireland
- Discipline of Paediatrics, Children’s Health Ireland at Crumlin and Tallaght, Dublin, Ireland
- Trinity Research in Childhood Centre (TRiCC), Trinity College Dublin, The University of Dublin, Dublin, Ireland
| | - Tobias Engel
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, University of Medicine and Health Sciences, Dublin, Ireland
- FutureNeuro, Science Foundation Ireland Research Centre for Chronic and Rare Neurological Diseases, Royal College of Surgeons in Ireland, University of Medicine and Health Sciences, Dublin, Ireland
| |
Collapse
|
25
|
Mazzarino RC, Baresova V, Zikánová M, Duval N, Wilkinson TG, Patterson D, Vacano GN. Transcriptome and metabolome analysis of crGART, a novel cell model of de novo purine synthesis deficiency: Alterations in CD36 expression and activity. PLoS One 2021; 16:e0247227. [PMID: 34283828 PMCID: PMC8291708 DOI: 10.1371/journal.pone.0247227] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 06/24/2021] [Indexed: 11/18/2022] Open
Abstract
In humans, GART [phosphoribosylglycinamide formyltransferase (EC 2.1.2.2) / phosphoribosylglycinamide synthetase (EC 6.3.4.13) / phosphoribosylaminoimidazole synthetase (EC 6.3.3.1)] is a trifunctional protein which catalyzes the second, third, and fifth reactions of the ten step de novo purine synthesis (DNPS) pathway. The second step of DNPS is conversion of phosphoribosylamine (5-PRA) to glycineamide ribonucleotide (GAR). 5-PRA is extremely unstable under physiological conditions and is unlikely to accumulate in the absence of GART activity. Recently, a HeLa cell line null mutant for GART was constructed via CRISPR-Cas9 mutagenesis. This cell line, crGART, is an important cellular model of DNPS inactivation that does not accumulate DNPS pathway intermediates. In the current study, we characterized the crGART versus HeLa transcriptomes in purine-supplemented and purine-depleted growth conditions. We observed multiple transcriptome changes and discuss pathways and ontologies particularly relevant to Alzheimer disease and Down syndrome. We selected the Cluster of Differentiation (CD36) gene for initial analysis based on its elevated expression in crGART versus HeLa as well as its high basal expression, high log2 value, and minimal P-value.
Collapse
Affiliation(s)
- Randall C. Mazzarino
- Knoebel Institute for Healthy Aging, University of Denver, Denver, Colorado, United States of America
- Eleanor Roosevelt Institute, University of Denver, Denver, Colorado, United States of America
- Department of Biological Sciences, University of Denver, Denver, Colorado, United States of America
- Molecular and Cellular Biophysics Program, University of Denver, Denver, Colorado, United States of America
| | - Veronika Baresova
- Research Unit for Rare Diseases, Department of Pediatrics and Adolescent Medicine, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Marie Zikánová
- Research Unit for Rare Diseases, Department of Pediatrics and Adolescent Medicine, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Nathan Duval
- Knoebel Institute for Healthy Aging, University of Denver, Denver, Colorado, United States of America
- Eleanor Roosevelt Institute, University of Denver, Denver, Colorado, United States of America
- Department of Biological Sciences, University of Denver, Denver, Colorado, United States of America
| | - Terry G. Wilkinson
- Knoebel Institute for Healthy Aging, University of Denver, Denver, Colorado, United States of America
- Eleanor Roosevelt Institute, University of Denver, Denver, Colorado, United States of America
| | - David Patterson
- Knoebel Institute for Healthy Aging, University of Denver, Denver, Colorado, United States of America
- Eleanor Roosevelt Institute, University of Denver, Denver, Colorado, United States of America
- Department of Biological Sciences, University of Denver, Denver, Colorado, United States of America
| | - Guido N. Vacano
- Knoebel Institute for Healthy Aging, University of Denver, Denver, Colorado, United States of America
- Eleanor Roosevelt Institute, University of Denver, Denver, Colorado, United States of America
| |
Collapse
|
26
|
Alçada-Morais S, Gonçalves N, Moreno-Juan V, Andres B, Ferreira S, Marques JM, Magalhães J, Rocha JMM, Xu X, Partidário M, Cunha RA, López-Bendito G, Rodrigues RJ. Adenosine A2A Receptors Contribute to the Radial Migration of Cortical Projection Neurons through the Regulation of Neuronal Polarization and Axon Formation. Cereb Cortex 2021; 31:5652-5663. [PMID: 34184030 DOI: 10.1093/cercor/bhab188] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 06/02/2021] [Accepted: 06/03/2021] [Indexed: 01/02/2023] Open
Abstract
Cortical interneurons born in the subpallium reach the cortex through tangential migration, whereas pyramidal cells reach their final position by radial migration. Purinergic signaling via P2Y1 receptors controls the migration of intermediate precursor cells from the ventricular zone to the subventricular zone. It was also reported that the blockade of A2A receptors (A2AR) controls the tangential migration of somatostatin+ interneurons. Here we found that A2AR control radial migration of cortical projection neurons. In A2AR-knockout (KO) mouse embryos or naïve mouse embryos exposed to an A2AR antagonist, we observed an accumulation of early-born migrating neurons in the lower intermediate zone at late embryogenesis. In utero knockdown of A2AR also caused an accumulation of neurons at the lower intermediate zone before birth. This entails the presently identified ability of A2AR to promote multipolar-bipolar transition and axon formation, critical for the transition of migrating neurons from the intermediate zone to the cortical plate. This effect seems to require extracellular ATP-derived adenosine since a similar accumulation of neurons at the lower intermediate zone was observed in mice lacking ecto-5'-nucleotidase (CD73-KO). These findings frame adenosine as a fine-tune regulator of the wiring of cortical inhibitory and excitatory networks.
Collapse
Affiliation(s)
- Sofia Alçada-Morais
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra 3004-504, Portugal.,Institute for Interdisciplinary Research, University of Coimbra, Coimbra 3030-789, Portugal
| | - Nélio Gonçalves
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra 3004-504, Portugal
| | | | - Belén Andres
- Instituto de Neurociencias, CSIC-UMH, San Juan de Alicante 03550, Spain
| | - Sofia Ferreira
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra 3004-504, Portugal.,Institute for Interdisciplinary Research, University of Coimbra, Coimbra 3030-789, Portugal
| | - Joana M Marques
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra 3004-504, Portugal
| | - Joana Magalhães
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra 3004-504, Portugal
| | - João M M Rocha
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra 3004-504, Portugal
| | - Xinli Xu
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra 3004-504, Portugal.,Institute for Interdisciplinary Research, University of Coimbra, Coimbra 3030-789, Portugal
| | - Matilde Partidário
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra 3004-504, Portugal
| | - Rodrigo A Cunha
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra 3004-504, Portugal.,Faculty of Medicine, University of Coimbra, Coimbra 3004-504, Portugal
| | | | - Ricardo J Rodrigues
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra 3004-504, Portugal.,Institute for Interdisciplinary Research, University of Coimbra, Coimbra 3030-789, Portugal
| |
Collapse
|
27
|
Shekhar S, Hall JE, Klubo-Gwiezdzinska J. The Hypothalamic Pituitary Thyroid Axis and Sleep. CURRENT OPINION IN ENDOCRINE AND METABOLIC RESEARCH 2021; 17:8-14. [PMID: 34322645 PMCID: PMC8315115 DOI: 10.1016/j.coemr.2020.10.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Sleep has a bidirectional relationship with the hypothalamic-pituitary-thyroid (HPT) axis, and both these homeostatic processes are inter-dependent for robust physiological functioning. The quality and quantity of sleep influence the circadian pattern of TSH and thyroid hormone secretion. Short term sleep restriction significantly reduces the amplitude of nocturnal TSH secretion and may modulate active thyroid hormone secretion, likely through an increased sympathetic tone. Conversely, TSH and active thyroid hormone affect the quantity and architecture of sleep. For instance, low TSH values are permissive for slow wave sleep and maintenance of normal sleep architecture, while the hypo- or hyper-secretion of active thyroid hormones adversely affects the quality and quantity of sleep. Structural thyroid disorders may also be associated with an altered circadian clock - a phenomenon warranting further investigation. In this review, we aim to provide readers a comprehensive review on the associations between the HPT axis and sleep patterns.
Collapse
Affiliation(s)
- Skand Shekhar
- Section on Endocrinology & Genetics (SEGEN), Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH), Bethesda, Maryland, USA
- Clinical Research Branch (CRB), National Institute of Environmental Health Sciences (NIEHS), National Institutes of Health (NIH), Research Triangle Park, North Carolina, USA
| | - Janet E. Hall
- Clinical Research Branch (CRB), National Institute of Environmental Health Sciences (NIEHS), National Institutes of Health (NIH), Research Triangle Park, North Carolina, USA
| | - Joanna Klubo-Gwiezdzinska
- Thyroid Tumors and Functional Thyroid Disorders Section, Metabolic Disease Branch, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), National Institutes of Health (NIH), Bethesda, Maryland, USA
| |
Collapse
|
28
|
Purinergic signaling in nervous system health and disease: Focus on pannexin 1. Pharmacol Ther 2021; 225:107840. [PMID: 33753132 DOI: 10.1016/j.pharmthera.2021.107840] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/03/2021] [Indexed: 02/06/2023]
Abstract
Purinergic signaling encompasses the cycle of adenosine 5' triphosphate (ATP) release and its metabolism into nucleotide and nucleoside derivatives, the direct release of nucleosides, and subsequent receptor-triggered downstream intracellular pathways. Since the discovery of nerve terminal and glial ATP release into the neuropil, purinergic signaling has been implicated in the modulation of nervous system development, function, and disease. In this review, we detail our current understanding of the roles of the pannexin 1 (PANX1) ATP-release channel in neuronal development and plasticity, glial signaling, and neuron-glial-immune interactions. We additionally provide an overview of PANX1 structure, activation, and permeability to orientate readers and highlight recent research developments. We identify areas of convergence between PANX1 and purinergic receptor actions. Additional highlights include data on PANX1's participation in the pathophysiology of nervous system developmental, degenerative, and inflammatory disorders. Our aim in combining this knowledge is to facilitate the movement of our current understanding of PANX1 in the context of other nervous system purinergic signaling mechanisms one step closer to clinical translation.
Collapse
|
29
|
Lopes CR, Lourenço VS, Tomé ÂR, Cunha RA, Canas PM. Use of knockout mice to explore CNS effects of adenosine. Biochem Pharmacol 2020; 187:114367. [PMID: 33333075 DOI: 10.1016/j.bcp.2020.114367] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 12/09/2020] [Accepted: 12/09/2020] [Indexed: 12/11/2022]
Abstract
The initial exploration using pharmacological tools of the role of adenosine receptors in the brain, concluded that adenosine released as such acted on A1R to inhibit excitability and glutamate release from principal neurons throughout the brain and that adenosine A2A receptors (A2AR) were striatal-'specific' receptors controlling dopamine D2R. This indicted A1R as potential controllers of neurodegeneration and A2AR of psychiatric conditions. Global knockout of these two receptors questioned the key role of A1R and instead identified extra-striatal A2AR as robust controllers of neurodegeneration. Furthermore, transgenic lines with altered metabolic sources of adenosine revealed a coupling of ATP-derived adenosine to activate A2AR and a role of A1R as a hurdle to initiate neurodegeneration. Additionally, cell-selective knockout of A2AR unveiled the different roles of A2AR in different cell types (neurons/astrocytes) in different portions of the striatal circuits (dorsal versus lateral) and in different brain areas (hippocampus/striatum). Finally, a new transgenic mouse line with deletion of all adenosine receptors seems to indicate a major allostatic rather than homeostatic role of adenosine and may allow isolating P2R-mediated responses to unravel their role in the brain, a goal close to heart of Geoffrey Burnstock, to whom we affectionately dedicate this review.
Collapse
Affiliation(s)
- Cátia R Lopes
- CNC- Center for Neuroscience and Cell Biology, University of Coimbra, 3004-517 Coimbra, Portugal
| | - Vanessa S Lourenço
- CNC- Center for Neuroscience and Cell Biology, University of Coimbra, 3004-517 Coimbra, Portugal
| | - Ângelo R Tomé
- CNC- Center for Neuroscience and Cell Biology, University of Coimbra, 3004-517 Coimbra, Portugal; Department of Life Sciences, Faculty of Sciences and Technology, University of Coimbra, 3000-456 Coimbra, Portugal
| | - Rodrigo A Cunha
- CNC- Center for Neuroscience and Cell Biology, University of Coimbra, 3004-517 Coimbra, Portugal; Faculty of Medicine, University of Coimbra, 3004-504 Coimbra, Portugal.
| | - Paula M Canas
- CNC- Center for Neuroscience and Cell Biology, University of Coimbra, 3004-517 Coimbra, Portugal
| |
Collapse
|
30
|
Gomes JI, Farinha-Ferreira M, Rei N, Gonçalves-Ribeiro J, Ribeiro JA, Sebastião AM, Vaz SH. Of adenosine and the blues: The adenosinergic system in the pathophysiology and treatment of major depressive disorder. Pharmacol Res 2020; 163:105363. [PMID: 33285234 DOI: 10.1016/j.phrs.2020.105363] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 11/30/2020] [Accepted: 12/01/2020] [Indexed: 12/28/2022]
Abstract
Major depressive disorder (MDD) is the foremost cause of global disability, being responsible for enormous personal, societal, and economical costs. Importantly, existing pharmacological treatments for MDD are partially or totally ineffective in a large segment of patients. As such, the search for novel antidepressant drug targets, anchored on a clear understanding of the etiological and pathophysiological mechanisms underpinning MDD, becomes of the utmost importance. The adenosinergic system, a highly conserved neuromodulatory system, appears as a promising novel target, given both its regulatory actions over many MDD-affected systems and processes. With this goal in mind, we herein review the evidence concerning the role of adenosine as a potential player in pathophysiology and treatment of MDD, combining data from both human and animal studies. Altogether, evidence supports the assertions that the adenosinergic system is altered in both MDD patients and animal models, and that drugs targeting this system have considerable potential as putative antidepressants. Furthermore, evidence also suggests that modifications in adenosine signaling may have a key role in the effects of several pharmacological and non-pharmacological antidepressant treatments with demonstrated efficacy, such as electroconvulsive shock, sleep deprivation, and deep brain stimulation. Lastly, it becomes clear from the available literature that there is yet much to study regarding the role of the adenosinergic system in the pathophysiology and treatment of MDD, and we suggest several avenues of research that are likely to prove fruitful.
Collapse
Affiliation(s)
- Joana I Gomes
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal; Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Miguel Farinha-Ferreira
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal; Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Nádia Rei
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal; Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Joana Gonçalves-Ribeiro
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal; Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Joaquim A Ribeiro
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal; Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Ana M Sebastião
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal; Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Sandra H Vaz
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal; Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal.
| |
Collapse
|
31
|
Song W, Tang Y, Wei L, Zhang C, Song D, Li X, Jiang S. Protective effect of CD73 inhibitor α, β-methylene ADP against amyloid-β-induced cognitive impairment by inhibiting adenosine production in hippocampus. ELECTRON J BIOTECHN 2020. [DOI: 10.1016/j.ejbt.2020.09.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
|
32
|
Menéndez Méndez A, Smith J, Engel T. Neonatal Seizures and Purinergic Signalling. Int J Mol Sci 2020; 21:ijms21217832. [PMID: 33105750 PMCID: PMC7660091 DOI: 10.3390/ijms21217832] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 10/18/2020] [Accepted: 10/20/2020] [Indexed: 02/07/2023] Open
Abstract
Neonatal seizures are one of the most common comorbidities of neonatal encephalopathy, with seizures aggravating acute injury and clinical outcomes. Current treatment can control early life seizures; however, a high level of pharmacoresistance remains among infants, with increasing evidence suggesting current anti-seizure medication potentiating brain damage. This emphasises the need to develop safer therapeutic strategies with a different mechanism of action. The purinergic system, characterised by the use of adenosine triphosphate and its metabolites as signalling molecules, consists of the membrane-bound P1 and P2 purinoreceptors and proteins to modulate extracellular purine nucleotides and nucleoside levels. Targeting this system is proving successful at treating many disorders and diseases of the central nervous system, including epilepsy. Mounting evidence demonstrates that drugs targeting the purinergic system provide both convulsive and anticonvulsive effects. With components of the purinergic signalling system being widely expressed during brain development, emerging evidence suggests that purinergic signalling contributes to neonatal seizures. In this review, we first provide an overview on neonatal seizure pathology and purinergic signalling during brain development. We then describe in detail recent evidence demonstrating a role for purinergic signalling during neonatal seizures and discuss possible purine-based avenues for seizure suppression in neonates.
Collapse
Affiliation(s)
- Aida Menéndez Méndez
- Department of Physiology and Medical Physics, RCSI University of Medicine and Health Sciences, Dublin D02 YN77, Ireland; (A.M.M.); (J.S.)
| | - Jonathon Smith
- Department of Physiology and Medical Physics, RCSI University of Medicine and Health Sciences, Dublin D02 YN77, Ireland; (A.M.M.); (J.S.)
- FutureNeuro, Science Foundation Ireland Research Centre for Chronic and Rare Neurological Diseases, RCSI University of Medicine and Health Sciences, Dublin D02 YN77, Ireland
| | - Tobias Engel
- Department of Physiology and Medical Physics, RCSI University of Medicine and Health Sciences, Dublin D02 YN77, Ireland; (A.M.M.); (J.S.)
- FutureNeuro, Science Foundation Ireland Research Centre for Chronic and Rare Neurological Diseases, RCSI University of Medicine and Health Sciences, Dublin D02 YN77, Ireland
- Correspondence: ; Tel.: +35-314-025-199
| |
Collapse
|
33
|
Mazzarino RC, Baresova V, Zikánová M, Duval N, Wilkinson TG, Patterson D, Vacano GN. The CRISPR-Cas9 crATIC HeLa transcriptome: Characterization of a novel cellular model of ATIC deficiency and ZMP accumulation. Mol Genet Metab Rep 2020; 25:100642. [PMID: 32939338 PMCID: PMC7479443 DOI: 10.1016/j.ymgmr.2020.100642] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Accepted: 08/18/2020] [Indexed: 12/15/2022] Open
Abstract
In de novo purine biosynthesis (DNPS), 5-aminoimidazole-4-carboxamide ribonucleotide formyltransferase (EC 2.1.2.3)/inosine monophosphate cyclohydrolase (EC 3.5.4.10) (ATIC) catalyzes the last two reactions of the pathway: conversion of 5-aminoimidazole-4-carboxamide ribonucleotide [aka Z-nucleotide monophosphate (ZMP)] to 5-formamido-4-imidazolecarboxamide ribonucleotide (FAICAR) then to inosine monophosphate (IMP). Mutations in ATIC cause an untreatable and devastating inborn error of metabolism in humans. ZMP is an adenosine monophosphate (AMP) mimetic and a known activator of AMP-activated protein kinase (AMPK). Recently, a HeLa cell line null mutant for ATIC was constructed via CRISPR-Cas9 mutagenesis. This mutant, crATIC, accumulates ZMP during purine starvation. Given that the mutant can accumulate ZMP in the absence of treatment with exogenous compounds, crATIC is likely an important cellular model of DNPS inactivation and ZMP accumulation. In the current study, we characterize the crATIC transcriptome versus the HeLa transcriptome in purine-supplemented and purine-depleted growth conditions. We report and discuss transcriptome changes with particular relevance to Alzheimer's disease and in genes relevant to lipid and fatty acid synthesis, neurodevelopment, embryogenesis, cell cycle maintenance and progression, extracellular matrix, immune function, TGFβ and other cellular processes.
Collapse
Key Words
- 5-aminoimidazole-4-carboxamide ribonucleoside, (AICAr)
- 5-aminoimidazole-4-carboxamide ribonucleotide formyltransferase/inosine monophosphate cyclohydrolase, (ATIC)
- 5-aminoimidazole-4-carboxamide ribonucleotide, (ZMP)
- 5-formamido-4-imidazolecarboxamide ribonucleotide, (FAICAR)
- AICA-ribosiduria
- AMP-activated protein kinase, (AMPK)
- Alzheimer's disease
- Development
- Purine synthesis
- RNA-seq
- Tuberous Sclerosis Complex 1 and 2, (TSC1 and TSC2)
- adenine phosphoribosyltransferase, (APRT)
- adenosine monophosphate, (AMP)
- adenosine triphosphate, (ATP)
- adenylosuccinate lyase, (ADSL)
- arachidonic acid, (AA)
- cyclooxygenase, (COX)
- cytochrome, P450 (CYP)
- cytosolic phospholipase A2, (cPLA2)
- de novo purine synthesis, (DNPS)
- differentially expressed gene, (DEG)
- false discovery rate, (FDR)
- fatty acid amide hydrolase, (FAAH)
- fetal calf macroserum, (FCM)
- fetal calf serum, (FCS)
- fragments per kilobase of exon per million reads mapped, (FPKM)
- gene ontology, (GO)
- guanosine monophosphate, (GMP)
- inosine monophosphate, (IMP)
- interferon, (INF)
- lipoxygenase, (LOX)
- mammalian Target of Rapamycin, (mTOR)
- minus adenine crATIC to minus adenine WT comparison, (MM)
- phospholipase, (PLA)
- phosphoribosyl pyrophosphate, (PRPP)
- phosphoribosylaminoimidazole carboxylase/phosphoribosylaminoimidazole succinocarboxamide synthetase, (PAICS)
- plus adenine crATIC to plus adenine WT comparison, (PP)
- xanthine monophosphate, (XMP)
Collapse
Affiliation(s)
- Randall C Mazzarino
- Knoebel Institute for Healthy Aging, University of Denver, 2155 E. Wesley Avenue, Denver, CO 80210, USA.,Eleanor Roosevelt Institute, University of Denver, Denver, CO 80210, USA.,Department of Biological Sciences, University of Denver, Denver, CO 80210, USA.,Molecular and Cellular Biophysics Program, University of Denver, Denver, CO 80210, USA
| | - Veronika Baresova
- Research Unit for Rare Diseases, Department of Pediatrics and Adolescent Medicine, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic
| | - Marie Zikánová
- Research Unit for Rare Diseases, Department of Pediatrics and Adolescent Medicine, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic
| | - Nathan Duval
- Knoebel Institute for Healthy Aging, University of Denver, 2155 E. Wesley Avenue, Denver, CO 80210, USA.,Eleanor Roosevelt Institute, University of Denver, Denver, CO 80210, USA.,Department of Biological Sciences, University of Denver, Denver, CO 80210, USA
| | - Terry G Wilkinson
- Knoebel Institute for Healthy Aging, University of Denver, 2155 E. Wesley Avenue, Denver, CO 80210, USA.,Eleanor Roosevelt Institute, University of Denver, Denver, CO 80210, USA
| | - David Patterson
- Knoebel Institute for Healthy Aging, University of Denver, 2155 E. Wesley Avenue, Denver, CO 80210, USA.,Eleanor Roosevelt Institute, University of Denver, Denver, CO 80210, USA.,Molecular and Cellular Biophysics Program, University of Denver, Denver, CO 80210, USA
| | - Guido N Vacano
- Knoebel Institute for Healthy Aging, University of Denver, 2155 E. Wesley Avenue, Denver, CO 80210, USA.,Eleanor Roosevelt Institute, University of Denver, Denver, CO 80210, USA
| |
Collapse
|
34
|
Carvalho K, Faivre E, Pietrowski MJ, Marques X, Gomez-Murcia V, Deleau A, Huin V, Hansen JN, Kozlov S, Danis C, Temido-Ferreira M, Coelho JE, Mériaux C, Eddarkaoui S, Gras SL, Dumoulin M, Cellai L, Landrieu I, Chern Y, Hamdane M, Buée L, Boutillier AL, Levi S, Halle A, Lopes LV, Blum D. Exacerbation of C1q dysregulation, synaptic loss and memory deficits in tau pathology linked to neuronal adenosine A2A receptor. Brain 2020; 142:3636-3654. [PMID: 31599329 PMCID: PMC6821333 DOI: 10.1093/brain/awz288] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 07/24/2019] [Accepted: 07/26/2019] [Indexed: 12/14/2022] Open
Abstract
Accumulating data support the role of tau pathology in cognitive decline in ageing and Alzheimer’s disease, but underlying mechanisms remain ill-defined. Interestingly, ageing and Alzheimer’s disease have been associated with an abnormal upregulation of adenosine A2A receptor (A2AR), a fine tuner of synaptic plasticity. However, the link between A2AR signalling and tau pathology has remained largely unexplored. In the present study, we report for the first time a significant upregulation of A2AR in patients suffering from frontotemporal lobar degeneration with the MAPT P301L mutation. To model these alterations, we induced neuronal A2AR upregulation in a tauopathy mouse model (THY-Tau22) using a new conditional strain allowing forebrain overexpression of the receptor. We found that neuronal A2AR upregulation increases tau hyperphosphorylation, potentiating the onset of tau-induced memory deficits. This detrimental effect was linked to a singular microglial signature as revealed by RNA sequencing analysis. In particular, we found that A2AR overexpression in THY-Tau22 mice led to the hippocampal upregulation of C1q complement protein—also observed in patients with frontotemporal lobar degeneration—and correlated with the loss of glutamatergic synapses, likely underlying the observed memory deficits. These data reveal a key impact of overactive neuronal A2AR in the onset of synaptic loss in tauopathies, paving the way for new therapeutic approaches.
Collapse
Affiliation(s)
- Kevin Carvalho
- University of Lille, Inserm, CHU Lille, UMR-S 1172 - JPArc, LabEx DISTALZ, F Lille, France
| | - Emilie Faivre
- University of Lille, Inserm, CHU Lille, UMR-S 1172 - JPArc, LabEx DISTALZ, F Lille, France
| | | | - Xavier Marques
- Institut du Fer à Moulin, Inserm UMR-S 1270, Sorbonne Université, F, Paris, France
| | - Victoria Gomez-Murcia
- University of Lille, Inserm, CHU Lille, UMR-S 1172 - JPArc, LabEx DISTALZ, F Lille, France
| | - Aude Deleau
- University of Lille, Inserm, CHU Lille, UMR-S 1172 - JPArc, LabEx DISTALZ, F Lille, France
| | - Vincent Huin
- University of Lille, Inserm, CHU Lille, UMR-S 1172 - JPArc, LabEx DISTALZ, F Lille, France
| | - Jan N Hansen
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Stanislav Kozlov
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Clément Danis
- University of Lille, Inserm, CHU Lille, UMR-S 1172 - JPArc, LabEx DISTALZ, F Lille, France.,University of Lille, CNRS UMR8576, Unité de Glycobiologie Structurale et Fonctionnelle, LabEx DISTALZ, Lille, F Lille, France
| | - Mariana Temido-Ferreira
- Instituto de Medicina Molecular, Faculdade de Medicina de Lisboa, Universidade de Lisboa, Lisbon, Portugal
| | - Joana E Coelho
- Instituto de Medicina Molecular, Faculdade de Medicina de Lisboa, Universidade de Lisboa, Lisbon, Portugal
| | - Céline Mériaux
- University of Lille, Inserm, CHU Lille, UMR-S 1172 - JPArc, LabEx DISTALZ, F Lille, France
| | - Sabiha Eddarkaoui
- University of Lille, Inserm, CHU Lille, UMR-S 1172 - JPArc, LabEx DISTALZ, F Lille, France
| | - Stéphanie Le Gras
- CNRS, Inserm, UMR 7104, GenomEast Platform, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Université de Strasbourg, F Illkirch, France
| | | | - Lucrezia Cellai
- University of Lille, Inserm, CHU Lille, UMR-S 1172 - JPArc, LabEx DISTALZ, F Lille, France
| | | | - Isabelle Landrieu
- University of Lille, CNRS UMR8576, Unité de Glycobiologie Structurale et Fonctionnelle, LabEx DISTALZ, Lille, F Lille, France
| | - Yijuang Chern
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Malika Hamdane
- University of Lille, Inserm, CHU Lille, UMR-S 1172 - JPArc, LabEx DISTALZ, F Lille, France
| | - Luc Buée
- University of Lille, Inserm, CHU Lille, UMR-S 1172 - JPArc, LabEx DISTALZ, F Lille, France
| | - Anne-Laurence Boutillier
- Laboratoire de Neuroscience Cognitives et Adaptatives (LNCA), CNRS UMR 7364, Université de Strasbourg, F Strasbourg, France
| | - Sabine Levi
- Institut du Fer à Moulin, Inserm UMR-S 1270, Sorbonne Université, F, Paris, France
| | - Annett Halle
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany.,Institute of Neuropathology, University of Bonn Medical Center, Bonn, Germany
| | - Luisa V Lopes
- Instituto de Medicina Molecular, Faculdade de Medicina de Lisboa, Universidade de Lisboa, Lisbon, Portugal
| | - David Blum
- University of Lille, Inserm, CHU Lille, UMR-S 1172 - JPArc, LabEx DISTALZ, F Lille, France
| |
Collapse
|
35
|
Fabbrizio P, Apolloni S, Bianchi A, Salvatori I, Valle C, Lanzuolo C, Bendotti C, Nardo G, Volonté C. P2X7 activation enhances skeletal muscle metabolism and regeneration in SOD1G93A mouse model of amyotrophic lateral sclerosis. Brain Pathol 2020; 30:272-282. [PMID: 31376190 PMCID: PMC7065186 DOI: 10.1111/bpa.12774] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Accepted: 07/26/2019] [Indexed: 12/11/2022] Open
Abstract
Muscle weakness plays an important role in neuromuscular disorders comprising amyotrophic lateral sclerosis (ALS). However, it is not established whether muscle denervation originates from the motor neurons, the muscles or more likely both. Previous studies have shown that the expression of the SOD1G93A mutation in skeletal muscles causes denervation of the neuromuscular junctions, inability to regenerate and consequent atrophy, all clear symptoms of ALS. In this work, we used SOD1G93A mice, a model that best mimics some pathological features of both familial and sporadic ALS, and we investigated some biological effects induced by the activation of the P2X7 receptor in the skeletal muscles. The P2X7, belonging to the ionotropic family of purinergic receptors for extracellular ATP, is abundantly expressed in the healthy skeletal muscles, where it controls cell duplication, differentiation, regeneration or death. In particular, we evaluated whether an in vivo treatment in SOD1G93A mice with the P2X7 specific agonist 2'(3')-O-(4-Benzoylbenzoyl) adenosine5'-triphosphate (BzATP) just before the onset of a pathological neuromuscular phenotype could exert beneficial effects in the skeletal muscles. Our findings indicate that stimulation of P2X7 improves the innervation and metabolism of myofibers, moreover elicits the proliferation/differentiation of satellite cells, thus preventing the denervation atrophy of skeletal muscles in SOD1G93A mice. Overall, this study suggests that a P2X7-targeted and site-specific modulation might be a strategy to interfere with the complex multifactorial and multisystem nature of ALS.
Collapse
Affiliation(s)
- Paola Fabbrizio
- Laboratory of Molecular Neurobiology, Department of NeuroscienceIstituto di Ricerche Farmacologiche Mario Negri IRCCSMilanItaly
| | | | | | | | - Cristiana Valle
- IRCCS Fondazione Santa LuciaRomeItaly
- National Research Council, Institute of Translational PharmacologyRomeItaly
| | - Chiara Lanzuolo
- IRCCS Fondazione Santa LuciaRomeItaly
- National Research Council, Institute of Biomedical TechnologiesMilanItaly
| | - Caterina Bendotti
- Laboratory of Molecular Neurobiology, Department of NeuroscienceIstituto di Ricerche Farmacologiche Mario Negri IRCCSMilanItaly
| | - Giovanni Nardo
- Laboratory of Molecular Neurobiology, Department of NeuroscienceIstituto di Ricerche Farmacologiche Mario Negri IRCCSMilanItaly
| | - Cinzia Volonté
- IRCCS Fondazione Santa LuciaRomeItaly
- National Research Council, Institute for Systems Analysis and Computer ScienceRomeItaly
| |
Collapse
|
36
|
Blanchard C, Boué-Grabot E, Massé K. Comparative Embryonic Spatio-Temporal Expression Profile Map of the Xenopus P2X Receptor Family. Front Cell Neurosci 2019; 13:340. [PMID: 31402854 PMCID: PMC6676501 DOI: 10.3389/fncel.2019.00340] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Accepted: 07/10/2019] [Indexed: 01/27/2023] Open
Abstract
P2X receptors are ATP-gated cations channels formed by the homo or hetero-trimeric association from the seven cloned subunits (P2X1-7). P2X receptors are widely distributed in different organs and cell types throughout the body including the nervous system and are involved in a large variety of physiological but also pathological processes in adult mammals. However, their expression and function during embryogenesis remain poorly understood. Here, we report the cloning and the comparative expression map establishment of the entire P2X subunit family in the clawed frog Xenopus. Orthologous sequences for 6 mammalian P2X subunits were identified in both X. laevis and X. tropicalis, but not for P2X3 subunit, suggesting a potential loss of this subunit in the Pipidae family. Three of these genes (p2rx1, p2rx2, and p2rx5) exist as homeologs in the pseudoallotetraploid X. laevis, making a total of 9 subunits in this species. Phylogenetic analyses demonstrate the high level of conservation of these receptors between amphibian and other vertebrate species. RT-PCR revealed that all subunits are expressed during the development although zygotic p2rx6 and p2rx7 transcripts are mainly detected at late organogenesis stages. Whole mount in situ hybridization shows that each subunit displays a specific spatio-temporal expression profile and that these subunits can therefore be grouped into two groups, based on their expression or not in the developing nervous system. Overlapping expression in the central and peripheral nervous system and in the sensory organs suggests potential heteromerization and/or redundant functions of P2X subunits in Xenopus embryos. The developmental expression of the p2rx subunit family during early phases of embryogenesis indicates that these subunits may have distinct roles during vertebrate development, especially embryonic neurogenesis.
Collapse
Affiliation(s)
- Camille Blanchard
- Université de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, Bordeaux, France
- CNRS, Institut des Maladies Neurodégénératives, UMR 5293, Bordeaux, France
| | - Eric Boué-Grabot
- Université de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, Bordeaux, France
- CNRS, Institut des Maladies Neurodégénératives, UMR 5293, Bordeaux, France
| | - Karine Massé
- Université de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, Bordeaux, France
- CNRS, Institut des Maladies Neurodégénératives, UMR 5293, Bordeaux, France
| |
Collapse
|
37
|
Xiao C, Liu N, Jacobson KA, Gavrilova O, Reitman ML. Physiology and effects of nucleosides in mice lacking all four adenosine receptors. PLoS Biol 2019; 17:e3000161. [PMID: 30822301 PMCID: PMC6415873 DOI: 10.1371/journal.pbio.3000161] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 03/13/2019] [Accepted: 02/07/2019] [Indexed: 11/29/2022] Open
Abstract
Adenosine is a constituent of many molecules of life; increased free extracellular adenosine indicates cell damage or metabolic stress. The importance of adenosine signaling in basal physiology, as opposed to adaptive responses to danger/damage situations, is unclear. We generated mice lacking all four adenosine receptors (ARs), Adora1−/−;Adora2a−/−;Adora2b−/−;Adora3−/− (quad knockout [QKO]), to enable investigation of the AR dependence of physiologic processes, focusing on body temperature. The QKO mice demonstrate that ARs are not required for growth, metabolism, breeding, and body temperature regulation (diurnal variation, response to stress, and torpor). However, the mice showed decreased survival starting at about 15 weeks of age. While adenosine agonists cause profound hypothermia via each AR, adenosine did not cause hypothermia (or bradycardia or hypotension) in QKO mice, indicating that AR-independent signals do not contribute to adenosine-induced hypothermia. The hypothermia elicited by adenosine kinase inhibition (with A134974), inosine, or uridine also required ARs, as each was abolished in the QKO mice. The proposed mechanism for uridine-induced hypothermia is inhibition of adenosine transport by uridine, increasing local extracellular adenosine levels. In contrast, adenosine 5′-monophosphate (AMP)–induced hypothermia was attenuated in QKO mice, demonstrating roles for both AR-dependent and AR-independent mechanisms in this process. The physiology of the QKO mice appears to be the sum of the individual knockout mice, without clear evidence for synergy, indicating that the actions of the four ARs are generally complementary. The phenotype of the QKO mice suggests that, while extracellular adenosine is a signal of stress, damage, and/or danger, it is less important for baseline regulation of body temperature. A study of mice lacking all four adenosine receptors shows that while they mediate effects of uridine, inosine and adenosine, these receptors are dispensable for growth, metabolism, breeding, and body temperature regulation. This suggests that extracellular adenosine is a damage or danger signal, rather than a major regulator of baseline physiology. Elevated extracellular adenosine generally indicates metabolic stress or cell damage and regulates many aspects of physiology. We studied “QKO” mice lacking all four adenosine receptors. Young QKO mice do not appear obviously ill, but do show decreased survival later in life. QKO mice demonstrate that adenosine receptors are not required for growth, metabolism, breeding, and body temperature regulation. QKO mice are missing the pharmacologic effects of adenosine on body temperature, heart rate, and blood pressure. Therefore, all of these effects are mediated by the four adenosine receptors. We also determined that the hypothermic effects of a pharmacologic adenosine kinase inhibitor (A134974), uridine, or inosine each requires adenosine receptors. The uridine-induced hypothermia is likely due to its inhibition of adenosine uptake into cells. QKO mouse physiology appears to be the sum of the individual knockout mice, without evidence for synergy, indicating that the actions of the four adenosine receptors are generally complementary.
Collapse
Affiliation(s)
- Cuiying Xiao
- Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, Maryland, United States of America
| | - Naili Liu
- Mouse Metabolism Core, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, Maryland, United States of America
| | - Kenneth A. Jacobson
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, Maryland, United States of America
| | - Oksana Gavrilova
- Mouse Metabolism Core, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, Maryland, United States of America
| | - Marc L. Reitman
- Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, Maryland, United States of America
- * E-mail:
| |
Collapse
|