1
|
Zhong J, Yu X, Lin Z. Phosphodiesterase 4 inhibition as a novel treatment for stroke. PeerJ 2025; 13:e18905. [PMID: 39897494 PMCID: PMC11786714 DOI: 10.7717/peerj.18905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Accepted: 01/06/2025] [Indexed: 02/04/2025] Open
Abstract
The incidence of stroke ranks third among the leading causes of mortality worldwide. It has the characteristics of high morbidity, high disability rate and high recurrence rate. The current risk associated with stroke surgery is exceedingly high. It may potentially outweigh the benefits and fail to ameliorate the cerebral tissue damage following ischemia. Therefore, pharmacological intervention assumes paramount importance. The use of thrombolytic drugs is most common in the treatment of stroke; however, its efficacy is limited due to its time-sensitive nature and propensity for increased bleeding. Over the past few years, the treatment of stroke has witnessed a surge in interest towards neuroprotective drugs that possess the potential to enhance neurological function. The PDE4D gene has been demonstrated to have a positive correlation with the risk of ischemic stroke. Additionally, the utilization of phosphodiesterase 4 inhibitors can enhance synaptic plasticity within the neural circuitry, regulate cellular metabolism, and prevent secondary brain injury caused by impaired blood flow. These mechanisms collectively facilitate the recovery of functional neurons, thereby serving as potential therapeutic interventions. Therefore, the comprehensive investigation of phosphodiesterase 4 as an innovative pharmacological target for stroke injury provides valuable insights into the development of therapeutic interventions in stroke treatment. This review is intended for, but not limited to, pharmacological researchers, drug target researchers, neurologists, neuromedical researchers, and behavioral scientists.
Collapse
Affiliation(s)
- Jiahong Zhong
- Department of Clinical Pharmacy, Meizhou People’s Hospital, Meizhou, Guangdong, China
| | - Xihui Yu
- Department of Pharmacy, The Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Zhuomiao Lin
- Department of Clinical Pharmacy, Meizhou People’s Hospital, Meizhou, Guangdong, China
| |
Collapse
|
2
|
Sun Z, Li L, Zhang L. Apigenin enhancing oxidative resistance and proteostasis to extend lifespan via PTEN-mediated AKT signalling pathway. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167670. [PMID: 39826849 DOI: 10.1016/j.bbadis.2025.167670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 12/27/2024] [Accepted: 01/10/2025] [Indexed: 01/22/2025]
Abstract
Aging is a complicated process, featuring the progressive deterioration of physiological functions and a heightened susceptibility to diseases including neurodegenerative disorders, cardiovascular diseases, and cancer. Apigenin, a flavonoid existing in various plants, has attracted attention due to its potential role in anti-aging. In this investigation, the potential effect of apigenin on extending lifespan in Saccharomyces cerevisiae (yeast) and Drosophila melanogaster (flies) was explored. The results indicate that apigenin significantly extends both replicative and chronological life duration in yeast, as well as longevity in male and female flies. Apigenin treatment also improves resistance to oxidative stress in both organisms, as manifested by enhanced survival, decreased reactive oxygen species (ROS) levels and upregulation of antioxidant enzymes. Furthermore, apigenin activates crucial elements of the proteostasis network (PN), such as upregulation of proteostasis-related enzymes activity and genes expression. Network analysis revealed that apigenin affects aging conserved in the longevity-regulating pathway. Notably, Pten is a hub target in flies. Apigenin regulated DmPten at both mRNA and protein expression level while modulating downstream targets, including the phosphorylation of AKT and associated signalling pathways. In a high-sucrose diet (HSD) model, Apigenin treatment extended lifespan, reduced hemolymph glucose levels, enhanced Pten expression, suppressed AKT phosphorylation, and modulated the phosphorylation status of S6K and expression of DmFoxo. These results demonstrate that apigenin could serve as a longevity research object and potential therapeutic drug for promoting health and longevity through its antioxidant and proteostatic properties.
Collapse
Affiliation(s)
- Zhengqiong Sun
- State Key Laboratory of Subtropical Silviculture, Zhejiang A & F University, Hangzhou 311300, China
| | - Lei Li
- Department of Pharmaceutical Botany, School of Pharmacy, Naval Medical University, Shanghai 200433, China
| | - Lei Zhang
- State Key Laboratory of Subtropical Silviculture, Zhejiang A & F University, Hangzhou 311300, China; Department of Pharmaceutical Botany, School of Pharmacy, Naval Medical University, Shanghai 200433, China.
| |
Collapse
|
3
|
Desouky MA, Michel HE, Elsherbiny DA, George MY. Recent pharmacological insights on abating toxic protein species burden in neurological disorders: Emphasis on 26S proteasome activation. Life Sci 2024; 359:123206. [PMID: 39489397 DOI: 10.1016/j.lfs.2024.123206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Revised: 07/30/2024] [Accepted: 10/30/2024] [Indexed: 11/05/2024]
Abstract
Protein homeostasis (proteostasis) refers to the plethora of mechanisms that safeguard the proper folding of the newly synthesized proteins. It entails various intricately regulated cues that demolish the toxic protein species to prevent their aggregation. The ubiquitin-proteasome system (UPS) is recognized as a salient protein degradation system, with a substantial role in maintaining proteostasis. However, under certain circumstances the protein degradation capacity of the UPS is overwhelmed, leading to the accumulation of misfolded proteins. Several neurodegenerative disorders, such as Alzheimer's disease, Parkinson's disease, Huntington disease, and amyotrophic lateral sclerosis are characterized with the presence of protein aggregates and proteinopathy. Accordingly, enhancing the 26S proteasome degradation activity might delineate a pioneering approach in targeting various proteotoxic disorders. Regrettably, the exact molecular approaches that enhance the proteasomal activity are still not fully understood. Therefore, this review aimed to underscore several signaling cascades that might restore the degradation capacity of this molecular machine. In this review, we discuss the different molecular components of the UPS and how 26S proteasomes are deleteriously affected in many neurodegenerative diseases. Moreover, we summarize different signaling pathways that can be utilized to renovate the 26S proteasome functional capacity, alongside currently known druggable targets in this circuit and various classes of proteasome activators.
Collapse
Affiliation(s)
- Mahmoud A Desouky
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, 11566 Cairo, Egypt
| | - Haidy E Michel
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, 11566 Cairo, Egypt
| | - Doaa A Elsherbiny
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, 11566 Cairo, Egypt
| | - Mina Y George
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, 11566 Cairo, Egypt.
| |
Collapse
|
4
|
Decio A, Agarwal N, Panzeri E, Bassi MT, D'Angelo MG. A new variant confirms GNAI2 as a rare cause of periventricular nodular heterotopia. Neurol Sci 2024; 45:5985-5988. [PMID: 39249692 DOI: 10.1007/s10072-024-07764-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Accepted: 09/04/2024] [Indexed: 09/10/2024]
Affiliation(s)
- Alice Decio
- Unit of Rehabilitation of Rare Diseases of the Central and Peripheral Nervous System, Scientific Institute IRCCS E. Medea, Bosisio Parini (LC), Italy.
| | - Nivedita Agarwal
- Neuroradiology and Radiology Unit, IRCCS Eugenio Medea, Bosisio Parini (LC), Italy
| | - Elena Panzeri
- Molecular Biology Laboratory, IRCCS Eugenio Medea, Bosisio Parini (LC), Italy
| | - Maria Teresa Bassi
- Molecular Biology Laboratory, IRCCS Eugenio Medea, Bosisio Parini (LC), Italy
| | - Maria Grazia D'Angelo
- Unit of Rehabilitation of Rare Diseases of the Central and Peripheral Nervous System, Scientific Institute IRCCS E. Medea, Bosisio Parini (LC), Italy
| |
Collapse
|
5
|
Borsdorf S, Zeug A, Wu Y, Mitroshina E, Vedunova M, Gaitonde SA, Bouvier M, Wehr MC, Labus J, Ponimaskin E. The cell adhesion molecule CD44 acts as a modulator of 5-HT7 receptor functions. Cell Commun Signal 2024; 22:563. [PMID: 39580460 PMCID: PMC11585102 DOI: 10.1186/s12964-024-01931-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 11/06/2024] [Indexed: 11/25/2024] Open
Abstract
BACKGROUND Homo- and heteromerization of G protein-coupled receptors (GPCRs) plays an important role in the regulation of receptor functions. Recently, we demonstrated an interaction between the serotonin receptor 7 (5-HT7R), a class A GPCR, and the cell adhesion molecule CD44. However, the functional consequences of this interaction on 5-HT7R-mediated signaling remained enigmatic. METHODS Using a quantitative FRET (Förster resonance energy transfer) approach, we determined the affinities for the formation of homo- and heteromeric complexes of 5-HT7R and CD44. The impact of heteromerization on 5-HT7R-mediated cAMP signaling was assessed using a cAMP responsive luciferase assay and a FRET-based cAMP biosensor under basal conditions as well as upon pharmacological modulation of the 5-HT7R and/or CD44 with specific ligands. We also investigated receptor-mediated G protein activation using BRET (bioluminescence resonance energy transfer)-based biosensors in both, homo- and heteromeric conditions. Finally, we analyzed expression profiles for 5-HT7R and CD44 in the brain during development. RESULTS We found that homo- and heteromerization of the 5-HT7R and CD44 occur at similar extent. Functionally, heteromerization increased 5-HT7R-mediated cAMP production under basal conditions. In contrast, agonist-mediated cAMP production was decreased in the presence of CD44. Mechanistically, this might be explained by increased Gαs and decreased GαoB activation by 5-HT7R/CD44 heteromers. Unexpectedly, treatment of the heteromeric complex with the CD44 ligand hyaluronic acid boosted constitutive 5-HT7R-mediated cAMP signaling and receptor-mediated transcription, suggesting the existence of a transactivation mechanism. CONCLUSIONS Interaction with the hyaluronan receptor CD44 modulates both the constitutive activity of 5-HT7R as well as its agonist-mediated signaling. Heteromerization also results in the transactivation of 5-HT7R-mediated signaling via CD44 ligand.
Collapse
Affiliation(s)
- Saskia Borsdorf
- Cellular Neurophysiology, Hannover Medical School, Hannover, Germany
| | - Andre Zeug
- Cellular Neurophysiology, Hannover Medical School, Hannover, Germany
| | - Yuxin Wu
- Research Group Cell Signalling, Department of Psychiatry and Psychotherapy, LMU University Hospital, LMU Munich, Munich, Germany
| | - Elena Mitroshina
- Department of Neurotechnology, Institute of Biology and Biomedicine, Lobachevsky University of Nizhni Novgorod, Nizhny Novgorod, Russia
| | - Maria Vedunova
- Department of Neurotechnology, Institute of Biology and Biomedicine, Lobachevsky University of Nizhni Novgorod, Nizhny Novgorod, Russia
| | - Supriya A Gaitonde
- Department of Biochemistry and Molecular Medicine, Institute for Research in Immunology and Cancer (IRIC), Université de Montréal, Montréal, QC, Canada
| | - Michel Bouvier
- Department of Biochemistry and Molecular Medicine, Institute for Research in Immunology and Cancer (IRIC), Université de Montréal, Montréal, QC, Canada
| | - Michael C Wehr
- Research Group Cell Signalling, Department of Psychiatry and Psychotherapy, LMU University Hospital, LMU Munich, Munich, Germany
- Systasy Bioscience GmbH, Planegg-Martinsried, Germany
| | - Josephine Labus
- Cellular Neurophysiology, Hannover Medical School, Hannover, Germany
| | - Evgeni Ponimaskin
- Cellular Neurophysiology, Hannover Medical School, Hannover, Germany.
| |
Collapse
|
6
|
Caneus J, Autar K, Akanda N, Grillo M, Long CJ, Jackson M, Lindquist S, Guo X, Morgan D, Hickman JJ. Validation of a functional human AD model with four AD therapeutics utilizing patterned ipsc-derived cortical neurons integrated with microelectrode arrays. Sci Rep 2024; 14:24875. [PMID: 39438515 PMCID: PMC11496884 DOI: 10.1038/s41598-024-73869-9] [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: 04/23/2024] [Accepted: 09/23/2024] [Indexed: 10/25/2024] Open
Abstract
Preclinical methods are needed for screening potential Alzheimer's disease (AD) therapeutics that recapitulate phenotypes found in the Mild Cognitive Impairment (MCI) stage or even before this stage of the disease. This would require a phenotypic system that reproduces cognitive deficits without significant neuronal cell death to mimic the clinical manifestations of AD during these stages. Long-term potentiation (LTP), which is a correlate of learning and memory, was induced in mature human iPSC-derived cortical neurons cultured on microelectrode arrays utilizing circuit patterns connecting two adjacent electrodes. We demonstrated an LTP system that modeled the MCI and pre-MCI stages of Alzheimer's and validated this functional system utilizing four AD therapeutics, which was also verified utilizing patch-clamp electrophysiology. LTP was induced by tetanic electrical stimulation, and LTP maintenance was significantly reduced in the presence of Amyloid-Beta 42 (Aβ42) oligomers compared to the controls, however, co-treatment with AD therapeutics (Donepezil, Memantine, Rolipram and Saracatinib) corrected Aβ42-induced LTP impairment. The results illustrate the utility of the system as a validated platform to model MCI AD pathology, and potentially for the pre-MCI phase before significant neuronal death. This system also has the potential to become an ideal platform for high-content therapeutic screening for other neurodegenerative diseases.
Collapse
Affiliation(s)
- Julbert Caneus
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway, Suite 400, Orlando, FL, 32826, USA.
| | - Kaveena Autar
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway, Suite 400, Orlando, FL, 32826, USA
| | - Nesar Akanda
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway, Suite 400, Orlando, FL, 32826, USA
| | - Marcella Grillo
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway, Suite 400, Orlando, FL, 32826, USA
| | | | - Max Jackson
- Hesperos Inc., 12501 Research Pkwy #100, Orlando, FL, USA
| | | | - Xiufang Guo
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway, Suite 400, Orlando, FL, 32826, USA
| | - Dave Morgan
- Department of Translational Neuroscience, Michigan State University College of Human Medicine, Grand Rapids, MI, USA
| | - James J Hickman
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway, Suite 400, Orlando, FL, 32826, USA
- Hesperos Inc., 12501 Research Pkwy #100, Orlando, FL, USA
| |
Collapse
|
7
|
Bale R, Doshi G. Deciphering the role of siRNA in anxiety and depression. Eur J Pharmacol 2024; 981:176868. [PMID: 39128805 DOI: 10.1016/j.ejphar.2024.176868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 07/02/2024] [Accepted: 08/05/2024] [Indexed: 08/13/2024]
Abstract
Anxiety and depression are central nervous system illnesses that are among the most prevalent medical concerns of the twenty-first century. Patients with this condition and their families bear psychological, financial, and societal hardship. There are currently restrictions when utilizing the conventional course of treatment. RNA interference is expected to become an essential approach in anxiety and depression due to its potent and targeted gene silencing. Silencing of genes by post-transcriptional modification is the mechanism of action of small interfering RNA (siRNA). The suppression of genes linked to disease is typically accomplished by siRNA molecules in an efficient and targeted manner. Unfavourable immune responses, off-target effects, naked siRNA instability, nuclease vulnerability, and the requirement to create an appropriate delivery method are some of the challenges facing the clinical application of siRNA. This review focuses on the use of siRNA in the treatment of anxiety and depression.
Collapse
Affiliation(s)
- Rajeshwari Bale
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, V L M Road, Vile Parle (w), Mumbai, 400056, India
| | - Gaurav Doshi
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, V L M Road, Vile Parle (w), Mumbai, 400056, India.
| |
Collapse
|
8
|
Narain P, Petković A, Šušić M, Haniffa S, Anwar M, Arnoux M, Drou N, Antonio-Saldi G, Chaudhury D. Nighttime-specific differential gene expression in suprachiasmatic nucleus and habenula is associated with resilience to chronic social stress. Transl Psychiatry 2024; 14:407. [PMID: 39358331 PMCID: PMC11447250 DOI: 10.1038/s41398-024-03100-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 09/10/2024] [Accepted: 09/17/2024] [Indexed: 10/04/2024] Open
Abstract
The molecular mechanisms that link stress and biological rhythms still remain unclear. The habenula (Hb) is a key brain region involved in regulating diverse types of emotion-related behaviours while the suprachiasmatic nucleus (SCN) is the body's central clock. To investigate the effects of chronic social stress on transcription patterns, we performed gene expression analysis in the Hb and SCN of stress-naïve and stress-exposed mice. Our analysis revealed a large number of differentially expressed genes and enrichment of synaptic and cell signalling pathways between resilient and stress-naïve mice at zeitgeber 16 (ZT16) in both the Hb and SCN. This transcriptomic signature was nighttime-specific and observed only in stress-resilient mice. In contrast, there were relatively few differences between the stress-susceptible and stress-naïve groups across time points. Our results reinforce the functional link between circadian gene expression patterns and differential responses to stress, thereby highlighting the importance of temporal expression patterns in homoeostatic stress responses.
Collapse
Affiliation(s)
- Priyam Narain
- Center for Genomics and Systems Biology, New York University Abu Dhabi, Abu Dhabi, UAE
| | - Aleksa Petković
- Department of Biology, New York University Abu Dhabi, Abu Dhabi, UAE
| | - Marko Šušić
- Department of Biology, New York University Abu Dhabi, Abu Dhabi, UAE
| | - Salma Haniffa
- Department of Biology, New York University Abu Dhabi, Abu Dhabi, UAE
| | - Mariam Anwar
- Department of Biology, New York University Abu Dhabi, Abu Dhabi, UAE
| | - Marc Arnoux
- Core Technology Platforms, New York University Abu Dhabi, Abu Dhabi, UAE
| | - Nizar Drou
- Center for Genomics and Systems Biology, New York University Abu Dhabi, Abu Dhabi, UAE
| | | | - Dipesh Chaudhury
- Center for Genomics and Systems Biology, New York University Abu Dhabi, Abu Dhabi, UAE.
- Department of Biology, New York University Abu Dhabi, Abu Dhabi, UAE.
- Center for Brain and Health, New York University Abu Dhabi, Abu Dhabi, UAE.
| |
Collapse
|
9
|
Chen S, Abou-Khalil BW, Afawi Z, Ali QZ, Amadori E, Anderson A, Anderson J, Andrade DM, Annesi G, Arslan M, Auce P, Bahlo M, Baker MD, Balagura G, Balestrini S, Banks E, Barba C, Barboza K, Bartolomei F, Bass N, Baum LW, Baumgartner TH, Baykan B, Bebek N, Becker F, Bennett CA, Beydoun A, Bianchini C, Bisulli F, Blackwood D, Blatt I, Borggräfe I, Bosselmann C, Braatz V, Brand H, Brockmann K, Buono RJ, Busch RM, Caglayan SH, Canafoglia L, Canavati C, Castellotti B, Cavalleri GL, Cerrato F, Chassoux F, Cherian C, Cherny SS, Cheung CL, Chou IJ, Chung SK, Churchhouse C, Ciullo V, Clark PO, Cole AJ, Cosico M, Cossette P, Cotsapas C, Cusick C, Daly MJ, Davis LK, Jonghe PD, Delanty N, Dennig D, Depondt C, Derambure P, Devinsky O, Vito LD, Dickerson F, Dlugos DJ, Doccini V, Doherty CP, El-Naggar H, Ellis CA, Epstein L, Evans M, Faucon A, Feng YCA, Ferguson L, Ferraro TN, Silva IFD, Ferri L, Feucht M, Fields MC, Fitzgerald M, Fonferko-Shadrach B, Fortunato F, Franceschetti S, French JA, Freri E, Fu JM, Gabriel S, Gagliardi M, Gambardella A, Gauthier L, Giangregorio T, Gili T, Glauser TA, Goldberg E, Goldman A, Goldstein DB, Granata T, Grant R, Greenberg DA, Guerrini R, Gundogdu-Eken A, Gupta N, Haas K, Hakonarson H, Haryanyan G, Häusler M, Hegde M, Heinzen EL, Helbig I, Hengsbach C, Heyne H, Hirose S, Hirsch E, Ho CJ, Hoeper O, Howrigan DP, Hucks D, Hung PC, Iacomino M, Inoue Y, Inuzuka LM, Ishii A, Jehi L, Johnson MR, Johnstone M, Kälviäinen R, Kanaan M, Kara B, Kariuki SM, Kegele J, Kesim Y, Khoueiry-Zgheib N, Khoury J, King C, Klein KM, Kluger G, Knake S, Kok F, Korczyn AD, Korinthenberg R, Koupparis A, Kousiappa I, Krause R, Krenn M, Krestel H, Krey I, Kunz WS, Kurlemann G, Kuzniecky RI, Kwan P, Vega-Talbott ML, Labate A, Lacey A, Lal D, Laššuthová P, Lauxmann S, Lawthom C, Leech SL, Lehesjoki AE, Lemke JR, Lerche H, Lesca G, Leu C, Lewin N, Lewis-Smith D, Li GHY, Liao C, Licchetta L, Lin CH, Lin KL, Linnankivi T, Lo W, Lowenstein DH, Lowther C, Lubbers L, Lui CHT, Macedo-Souza LI, Madeleyn R, Madia F, Magri S, Maillard L, Marcuse L, Marques P, Marson AG, Matthews AG, May P, Mayer T, McArdle W, McCarroll SM, McGoldrick P, McGraw CM, McIntosh A, McQuillan A, Meador KJ, Mei D, Michel V, Millichap JJ, Minardi R, Montomoli M, Mostacci B, Muccioli L, Muhle H, Müller-Schlüter K, Najm IM, Nasreddine W, Neaves S, Neubauer BA, Newton CRJC, Noebels JL, Northstone K, Novod S, O’Brien TJ, Owusu-Agyei S, Özkara Ç, Palotie A, Papacostas SS, Parrini E, Pato C, Pato M, Pendziwiat M, Pennell PB, Petrovski S, Pickrell WO, Pinsky R, Pinto D, Pippucci T, Piras F, Piras F, Poduri A, Pondrelli F, Posthuma D, Powell RHW, Privitera M, Rademacher A, Ragona F, Ramirez-Hamouz B, Rau S, Raynes HR, Rees MI, Regan BM, Reif A, Reinthaler E, Rheims S, Ring SM, Riva A, Rojas E, Rosenow F, Ryvlin P, Saarela A, Sadleir LG, Salman B, Salmon A, Salpietro V, Sammarra I, Scala M, Schachter S, Schaller A, Schankin CJ, Scheffer IE, Schneider N, Schubert-Bast S, Schulze-Bonhage A, Scudieri P, Sedláčková L, Shain C, Sham PC, Shiedley BR, Siena SA, Sills GJ, Sisodiya SM, Smoller JW, Solomonson M, Spalletta G, Sparks KR, Sperling MR, Stamberger H, Steinhoff BJ, Stephani U, Štěrbová K, Stewart WC, Stipa C, Striano P, Strzelczyk A, Surges R, Suzuki T, Talarico M, Talkowski ME, Taneja RS, Tanteles GA, Timonen O, Timpson NJ, Tinuper P, Todaro M, Topaloglu P, Tsai MH, Tumiene B, Turkdogan D, Uğur-İşeri S, Utkus A, Vaidiswaran P, Valton L, van Baalen A, Vari MS, Vetro A, Vlčková M, von Brauchitsch S, von Spiczak S, Wagner RG, Watts N, Weber YG, Weckhuysen S, Widdess-Walsh P, Wiebe S, Wolf SM, Wolff M, Wolking S, Wong I, von Wrede R, Wu D, Yamakawa K, Yapıcı Z, Yis U, Yolken R, Yücesan E, Zagaglia S, Zahnert F, Zara F, Zimprich F, Zizovic M, Zsurka G, Neale BM, Berkovic SF. Exome sequencing of 20,979 individuals with epilepsy reveals shared and distinct ultra-rare genetic risk across disorder subtypes. Nat Neurosci 2024; 27:1864-1879. [PMID: 39363051 PMCID: PMC11646479 DOI: 10.1038/s41593-024-01747-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 08/01/2024] [Indexed: 10/05/2024]
Abstract
Identifying genetic risk factors for highly heterogeneous disorders such as epilepsy remains challenging. Here we present, to our knowledge, the largest whole-exome sequencing study of epilepsy to date, with more than 54,000 human exomes, comprising 20,979 deeply phenotyped patients from multiple genetic ancestry groups with diverse epilepsy subtypes and 33,444 controls, to investigate rare variants that confer disease risk. These analyses implicate seven individual genes, three gene sets and four copy number variants at exome-wide significance. Genes encoding ion channels show strong association with multiple epilepsy subtypes, including epileptic encephalopathies and generalized and focal epilepsies, whereas most other gene discoveries are subtype specific, highlighting distinct genetic contributions to different epilepsies. Combining results from rare single-nucleotide/short insertion and deletion variants, copy number variants and common variants, we offer an expanded view of the genetic architecture of epilepsy, with growing evidence of convergence among different genetic risk loci on the same genes. Top candidate genes are enriched for roles in synaptic transmission and neuronal excitability, particularly postnatally and in the neocortex. We also identify shared rare variant risk between epilepsy and other neurodevelopmental disorders. Our data can be accessed via an interactive browser, hopefully facilitating diagnostic efforts and accelerating the development of follow-up studies.
Collapse
|
10
|
Gil-Martí B, Isidro-Mézcua J, Poza-Rodriguez A, Asti Tello GS, Treves G, Turiégano E, Beckwith EJ, Martin FA. Socialization causes long-lasting behavioral changes. Sci Rep 2024; 14:22302. [PMID: 39333212 PMCID: PMC11436997 DOI: 10.1038/s41598-024-73218-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Accepted: 09/16/2024] [Indexed: 09/29/2024] Open
Abstract
In modern human societies, social isolation acts as a negative factor for health and life quality. On the other hand, social interaction also has profound effects on animal and human, impacting aggressiveness, feeding and sleep, among many other behaviors. Here, we observe that in the fly Drosophila melanogaster these behavioral changes long-last even after social interaction has ceased, suggesting that the socialization experience triggers behavioral plasticity. These modified behaviors maintain similar levels for 24 h and persist up to 72 h, although showing a progressive decay. We also find that impairing long-term memory mechanisms either genetically or by anesthesia abolishes the expected behavioral changes in response to social interaction. Furthermore, we show that socialization increases CREB-dependent neuronal activity and synaptic plasticity in the mushroom body, the main insect memory center analogous to mammalian hippocampus. We propose that social interaction triggers socialization awareness, understood as long-lasting changes in behavior caused by experience with mechanistic similarities to long-term memory formation.
Collapse
Affiliation(s)
- Beatriz Gil-Martí
- Cajal Institute, Spanish National Research Council (CSIC), Av Dr Arce 37, 28002, Madrid, Spain
- Department of Biology, Autonomous University of Madrid, Madrid, Spain
| | - Julia Isidro-Mézcua
- Cajal Institute, Spanish National Research Council (CSIC), Av Dr Arce 37, 28002, Madrid, Spain
| | - Adriana Poza-Rodriguez
- Cajal Institute, Spanish National Research Council (CSIC), Av Dr Arce 37, 28002, Madrid, Spain
| | - Gerson S Asti Tello
- Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE), UBA-CONICET, Buenos Aires, Argentina
| | - Gaia Treves
- Cajal Institute, Spanish National Research Council (CSIC), Av Dr Arce 37, 28002, Madrid, Spain
| | - Enrique Turiégano
- Department of Biology, Autonomous University of Madrid, Madrid, Spain
| | - Esteban J Beckwith
- Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE), UBA-CONICET, Buenos Aires, Argentina.
| | - Francisco A Martin
- Cajal Institute, Spanish National Research Council (CSIC), Av Dr Arce 37, 28002, Madrid, Spain.
| |
Collapse
|
11
|
Ali MZ, Anushree, Ahsan A, Ola MS, Haque R, Ahsan J. Ionotropic receptors mediate olfactory learning and memory in Drosophila. INSECT SCIENCE 2024; 31:1249-1269. [PMID: 38114448 DOI: 10.1111/1744-7917.13308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 10/16/2023] [Accepted: 10/27/2023] [Indexed: 12/21/2023]
Abstract
Phenylacetaldehyde (PAH), an aromatic compound, is present in a diverse range of fruits including overripe bananas and prickly pear cactus, the two major host fruits for Drosophila melanogaster. PAH acts as a potent ligand for the ionotropic receptor 84a (IR84a) in the adult fruit fly and it is detected by the IR84a/IR8a heterotetrameric complex. Its role in the male courtship behavior through IR84a as an environmental aphrodisiac is of additional importance. In D. melanogaster, two distinct kinds of olfactory receptors, that is, odorant receptors (ORs) and ionotropic receptors (IRs), perceive the odorant stimuli. They display unique structural, molecular, and functional characteristics in addition to having different evolutionary origins. Traditionally, olfactory cues detected by the ORs such as ethyl acetate, 1-butanol, isoamyl acetate, 1-octanol, 4-methylcyclohexanol, etc. classified as aliphatic esters and alcohols have been employed in olfactory classical conditioning using fruit flies. This underlines the participation of OR-activated olfactory pathways in learning and memory formation. Our study elucidates that likewise ethyl acetate (EA) (an OR-responsive odorant), PAH (an IR-responsive aromatic compound) too can form learning and memory when associated with an appetitive gustatory reinforcer. The association of PAH with sucrose (PAH/SUC) led to learning and formation of the long-term memory (LTM). Additionally, the Orco1, Ir84aMI00501, and Ir8a1 mutant flies were used to confirm the exclusive participation of the IR84a/IR8a complex in PAH/SUC olfactory associative conditioning. These results highlight the involvement of IRs via an IR-activated pathway in facilitating robust olfactory behavior.
Collapse
Affiliation(s)
- Md Zeeshan Ali
- Department of Biotechnology, Central University of South Bihar, Gaya, Bihar, India
| | - Anushree
- Department of Biotechnology, Central University of South Bihar, Gaya, Bihar, India
| | - Aarif Ahsan
- Department of Radiation Oncology, University of Michigan, Ann Arbor, Michigan, USA
| | - Mohammad Shamsul Ola
- Department of Biochemistry, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Rizwanul Haque
- Department of Biotechnology, Central University of South Bihar, Gaya, Bihar, India
| | - Jawaid Ahsan
- Department of Biotechnology, Central University of South Bihar, Gaya, Bihar, India
| |
Collapse
|
12
|
Ho DM, Shaban M, Mahmood F, Ganguly P, Todeschini L, Van Vactor D, Artavanis-Tsakonas S. cAMP/PKA signaling regulates TDP-43 aggregation and mislocalization. Proc Natl Acad Sci U S A 2024; 121:e2400732121. [PMID: 38838021 PMCID: PMC11181030 DOI: 10.1073/pnas.2400732121] [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: 01/12/2024] [Accepted: 05/03/2024] [Indexed: 06/07/2024] Open
Abstract
Cytoplasmic mislocalization and aggregation of TDP-43 protein are hallmarks of amyotrophic lateral sclerosis (ALS) and are observed in the vast majority of both familial and sporadic cases. How these two interconnected processes are regulated on a molecular level, however, remains enigmatic. Genome-wide screens for modifiers of the ALS-associated genes TDP-43 and FUS have identified the phospholipase D (Pld) pathway as a key regulator of ALS-related phenotypes in the fruit fly Drosophila melanogaster [M. W. Kankel et al., Genetics 215, 747-766 (2020)]. Here, we report the results of our search for downstream targets of the enzymatic product of Pld, phosphatidic acid. We identify two conserved negative regulators of the cAMP/PKA signaling pathway, the phosphodiesterase dunce and the inhibitory subunit PKA-R2, as modifiers of pathogenic phenotypes resulting from overexpression of the Drosophila TDP-43 ortholog TBPH. We show that knockdown of either of these genes results in a mitigation of both TBPH aggregation and mislocalization in larval motor neuron cell bodies, as well as an amelioration of adult-onset motor defects and shortened lifespan induced by TBPH. We determine that PKA kinase activity is downstream of both TBPH and Pld and that overexpression of the PKA target CrebA can rescue TBPH mislocalization. These findings suggest a model whereby increasing cAMP/PKA signaling can ameliorate the molecular and functional effects of pathological TDP-43.
Collapse
Affiliation(s)
- Diana M. Ho
- Department of Cell Biology, Harvard Medical School, Boston, MA02115
| | - Muhammad Shaban
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA02115
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA02114
- Cancer Data Science Program, Dana-Farber Cancer Institute, Boston, MA02115
- Cancer Program, Broad Institute of Harvard and MIT, Cambridge, MA02142
| | - Faisal Mahmood
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA02115
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA02114
- Cancer Data Science Program, Dana-Farber Cancer Institute, Boston, MA02115
- Cancer Program, Broad Institute of Harvard and MIT, Cambridge, MA02142
| | - Payel Ganguly
- Department of Cell Biology, Harvard Medical School, Boston, MA02115
| | | | - David Van Vactor
- Department of Cell Biology, Harvard Medical School, Boston, MA02115
| | | |
Collapse
|
13
|
Lin M, Gong J, Wu L, Lin X, Zhang Y, Lin W, Huang H, Zhu C. ADCY3: the pivotal gene in classical ketogenic diet for the treatment of epilepsy. Front Cell Neurosci 2024; 18:1305867. [PMID: 38841200 PMCID: PMC11150708 DOI: 10.3389/fncel.2024.1305867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Accepted: 04/15/2024] [Indexed: 06/07/2024] Open
Abstract
Objective Epilepsy is a common neurological disorder characterized by recurrent epilepsy episodes. As a non-pharmacological treatment, the ketogenic diet has been widely applied in treating epilepsy. However, the exact therapeutic mechanism of the ketogenic diet for epilepsy remains unclear. This study investigates the molecular mechanisms of the ketogenic diet in regulating fatty acid metabolism and activating the ADCY3-initiated cAMP signaling pathway to enhance neuronal inhibition and thereby treat epilepsy. Methods and results Meta-analysis reveals that the ketogenic diet is superior to the conventional diet in treating epilepsy. Animal experiments demonstrate that the ketogenic diet is more effective than the conventional diet in treating epilepsy, with the best results achieved using the classic ketogenic diet. Transcriptome sequencing analysis identifies six essential genes, among which ADCY3 shows increased expression in the ketogenic diet. In vivo experiments confirm that the activation of the cAMP-PKA signaling pathway by ADCY3 enhances neuronal inhibition and improves epilepsy control. Conclusion Clinical observations indicate that the ketogenic diet improves patient epilepsy episodes by regulating the ADCY3-initiated cAMP signaling pathway.
Collapse
Affiliation(s)
- Mingxing Lin
- Department of Pediatrics, Fujian Medical University Union Hospital, Fuzhou, China
| | - Jiayin Gong
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Luyan Wu
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Xin Lin
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Yuying Zhang
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Wanhui Lin
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Huapin Huang
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, China
- Fujian Key Laboratory of Molecular Neurology, Fuzhou, China
- Department of Geriatrics, Fujian Medical University Union Hospital, Fuzhou, China
| | - Chaofeng Zhu
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, China
| |
Collapse
|
14
|
Suárez-Grimalt R, Grunwald Kadow IC, Scheunemann L. An integrative sensor of body states: how the mushroom body modulates behavior depending on physiological context. Learn Mem 2024; 31:a053918. [PMID: 38876486 PMCID: PMC11199956 DOI: 10.1101/lm.053918.124] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 04/08/2024] [Indexed: 06/16/2024]
Abstract
The brain constantly compares past and present experiences to predict the future, thereby enabling instantaneous and future behavioral adjustments. Integration of external information with the animal's current internal needs and behavioral state represents a key challenge of the nervous system. Recent advancements in dissecting the function of the Drosophila mushroom body (MB) at the single-cell level have uncovered its three-layered logic and parallel systems conveying positive and negative values during associative learning. This review explores a lesser-known role of the MB in detecting and integrating body states such as hunger, thirst, and sleep, ultimately modulating motivation and sensory-driven decisions based on the physiological state of the fly. State-dependent signals predominantly affect the activity of modulatory MB input neurons (dopaminergic, serotoninergic, and octopaminergic), but also induce plastic changes directly at the level of the MB intrinsic and output neurons. Thus, the MB emerges as a tightly regulated relay station in the insect brain, orchestrating neuroadaptations due to current internal and behavioral states leading to short- but also long-lasting changes in behavior. While these adaptations are crucial to ensure fitness and survival, recent findings also underscore how circuit motifs in the MB may reflect fundamental design principles that contribute to maladaptive behaviors such as addiction or depression-like symptoms.
Collapse
Affiliation(s)
- Raquel Suárez-Grimalt
- Institute for Biology/Genetics, Freie Universität Berlin, 14195 Berlin, Germany
- Institut für Neurophysiologie and NeuroCure Cluster of Excellence, Charité-Universitätsmedizin Berlin, 10117 Berlin, Germany
| | | | - Lisa Scheunemann
- Institute for Biology/Genetics, Freie Universität Berlin, 14195 Berlin, Germany
- Institut für Neurophysiologie and NeuroCure Cluster of Excellence, Charité-Universitätsmedizin Berlin, 10117 Berlin, Germany
| |
Collapse
|
15
|
Sears JC, Broadie K. Use-Dependent, Untapped Dual Kinase Signaling Localized in Brain Learning Circuitry. J Neurosci 2024; 44:e1126232024. [PMID: 38267256 PMCID: PMC10957217 DOI: 10.1523/jneurosci.1126-23.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 01/12/2024] [Accepted: 01/16/2024] [Indexed: 01/26/2024] Open
Abstract
Imaging brain learning and memory circuit kinase signaling is a monumental challenge. The separation of phases-based activity reporter of kinase (SPARK) biosensors allow circuit-localized studies of multiple interactive kinases in vivo, including protein kinase A (PKA) and extracellular signal-regulated kinase (ERK) signaling. In the precisely-mapped Drosophila brain learning/memory circuit, we find PKA and ERK signaling differentially enriched in distinct Kenyon cell connectivity nodes. We discover that potentiating normal circuit activity induces circuit-localized PKA and ERK signaling, expanding kinase function within new presynaptic and postsynaptic domains. Activity-induced PKA signaling shows extensive overlap with previously selective ERK signaling nodes, while activity-induced ERK signaling arises in new connectivity nodes. We find targeted synaptic transmission blockade in Kenyon cells elevates circuit-localized ERK induction in Kenyon cells with normally high baseline ERK signaling, suggesting lateral and feedback inhibition. We discover overexpression of the pathway-linking Meng-Po (human SBK1) serine/threonine kinase to improve learning acquisition and memory consolidation results in dramatically heightened PKA and ERK signaling in separable Kenyon cell circuit connectivity nodes, revealing both synchronized and untapped signaling potential. Finally, we find that a mechanically-induced epileptic seizure model (easily shocked "bang-sensitive" mutants) has strongly elevated, circuit-localized PKA and ERK signaling. Both sexes were used in all experiments, except for the hemizygous male-only seizure model. Hyperexcitable, learning-enhanced, and epileptic seizure models have comparably elevated interactive kinase signaling, suggesting a common basis of use-dependent induction. We conclude that PKA and ERK signaling modulation is locally coordinated in use-dependent spatial circuit dynamics underlying seizure susceptibility linked to learning/memory potential.
Collapse
Affiliation(s)
- James C Sears
- Vanderbilt Brain Institute, Vanderbilt University and Medical Center, Nashville, Tennessee 37235
- Departments of Biological Sciences, Vanderbilt University and Medical Center, Nashville, Tennessee 37235
| | - Kendal Broadie
- Vanderbilt Brain Institute, Vanderbilt University and Medical Center, Nashville, Tennessee 37235
- Departments of Biological Sciences, Vanderbilt University and Medical Center, Nashville, Tennessee 37235
- Cell and Developmental Biology, Vanderbilt University and Medical Center, Nashville, Tennessee 37235
- Vanderbilt Kennedy Center, Vanderbilt University and Medical Center, Nashville, Tennessee 37235
| |
Collapse
|
16
|
Nguyen QTN, Park J, Kim DY, Tran DT, Han IO. Forskolin rescues hypoxia-induced cognitive dysfunction in zebrafish with potential involvement of O-GlcNAc cycling regulation. Biochem Pharmacol 2024; 221:116032. [PMID: 38281601 DOI: 10.1016/j.bcp.2024.116032] [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: 12/09/2023] [Revised: 12/28/2023] [Accepted: 01/23/2024] [Indexed: 01/30/2024]
Abstract
Repeated sublethal hypoxia exposure induces brain inflammation and affects the initiation and progression of cognitive dysfunction. Experiments from the current study showed that hypoxic exposure downregulates PKA/CREB signaling, which is restored by forskolin (FSK), an adenylate cyclase activator, in both Neuro2a (N2a) cells and zebrafish brain. FSK significantly protected N2a cells from hypoxia-induced cell death and neurite shrinkage. Intraperitoneal administration of FSK for 5 days on zebrafish additionally led to significant recovery from hypoxia-induced social interaction impairment and learning and memory (L/M) deficit. FSK suppressed hypoxia-induced neuroinflammation, as indicated by the observed decrease in NF-κB activation and GFAP expression. We further investigated the potential effect of FSK on O-GlcNAcylation changes induced by hypoxia. Intriguingly FSK induced marked upregulation of the protein level of O-GlcNAc transferase catalyzing addition of the GlcNAc group to target proteins, accompanied by elevated O-GlcNAcylation of nucleocytoplasmic proteins. The hypoxia-induced O-GlcNAcylation decrease in the brain of zebrafish was considerably restored following FSK treatment. Based on the collective results, we propose that FSK rescues hypoxia-induced cognitive dysfunction, potentially through regulation of HBP/O-GlcNAc cycling.
Collapse
Affiliation(s)
- Quynh T N Nguyen
- Department of Biomedical Science, Program in Biomedical Science and Engineering, College of Medicine, Inha University, Incheon, South Korea
| | - Jiwon Park
- Department of Biomedical Science, Program in Biomedical Science and Engineering, College of Medicine, Inha University, Incheon, South Korea
| | - Dong Yeol Kim
- Department of Biomedical Science, Program in Biomedical Science and Engineering, College of Medicine, Inha University, Incheon, South Korea
| | - Duong T Tran
- Department of Biomedical Science, Program in Biomedical Science and Engineering, College of Medicine, Inha University, Incheon, South Korea
| | - Inn Oc Han
- Department of Biomedical Science, Program in Biomedical Science and Engineering, College of Medicine, Inha University, Incheon, South Korea.
| |
Collapse
|
17
|
Zhu Z, Tao X, Dai T, Wu J, Han C, Huang P, Gong W. Cognitive-exercise dual-task attenuates chronic cerebral ischemia-induced cognitive impairment by activating cAMP/PKA pathway through inhibiting EphrinA3/EphA4. Exp Neurol 2024; 372:114617. [PMID: 38007209 DOI: 10.1016/j.expneurol.2023.114617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 11/03/2023] [Accepted: 11/19/2023] [Indexed: 11/27/2023]
Abstract
BACKGROUND The prevalence of vascular cognitive impairment induced by chronic cerebral ischemia (CCI) is increasing year by year. Cognitive-exercise dual-task intervention has shown beneficial effects on improving cognitive performance in ischemic patients. It is well known that the tyrosine kinase ligand-receptor (Ephrin-Eph) system plays an important role in synaptic transmission and that the cAMP/PKA pathway is associated with cognitive function. However, it is unclear whether they are responsible for the dual-task improving cognitive impairment in CCI. METHODS Bilateral common carotid artery occlusion (BCCAO) in SD rats was used to establish the CCI model. The effects of dual-task and single-task on cognitive function and the expressions of EphrinA3, EphA4, cAMP, and PKA in rats were detected by the novel object recognition (NOR) test, immunofluorescence staining, quantitative real-time polymerase chain reaction (qPCR), and Western blotting (WB), respectively. Overexpression or knockdown of EphrinA3 in astrocytes or rats were constructed by lentivirus infection to verify the effects of EphrinA3/EphA4 on the cAMP/PKA pathway. RESULTS After dual-task intervention, the discrimination index of rats increased significantly compared with the rats in the CCI group. The expressions of EphrinA3 and EphA4 were decreased, while the expressions of cAMP and PKA were increased. Furthermore, knockdown of EphrinA3 alleviated the trend of CCI-induced cognitive decline in rats and OGD-stimulated cellular damage. It also increased cAMP/PKA expression in hippocampal neurons. CONCLUSION Cognitive-exercise dual-task can significantly improve the cognitive impairment induced by CCI, and this effect may be better than that of the cognitive or exercise single-task intervention. The improvement may be related to the inhibition of EphrinA3/EphA4, followed by activation of the cAMP/PKA pathway.
Collapse
Affiliation(s)
- Ziman Zhu
- Beijing Rehabilitation Hospital, Beijing Rehabilitation Medicine Academy, Capital Medical University, Beijing 100144, China
| | - Xue Tao
- Department of Research, Beijing Rehabilitation Hospital, Capital Medical University, Beijing 100144, China
| | - Tengteng Dai
- The Second Clinical Medical College of Yunnan University of Chinese Medicine, Yunnan 650500, China
| | - Jilin Wu
- Beijing Rehabilitation Hospital, Beijing Rehabilitation Medicine Academy, Capital Medical University, Beijing 100144, China
| | - Conglin Han
- Rehabilitation Medicine Academy, Weifang Medical University, Shandong 261053, China
| | - Peiling Huang
- Department of Neurological Rehabilitation, Beijing Rehabilitation Hospital, Capital Medical University, Beijing 100144, China
| | - Weijun Gong
- Department of Neurological Rehabilitation, Beijing Rehabilitation Hospital, Capital Medical University, Beijing 100144, China.
| |
Collapse
|
18
|
Ismael S, Colvin RA, Lee D. Activation of cyclic AMP signaling pathway in dopaminergic neurons rescues locomotion defects in a Drosophila larval model of Parkinson's disease. Brain Res 2024; 1822:148641. [PMID: 37866407 PMCID: PMC10842101 DOI: 10.1016/j.brainres.2023.148641] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 09/03/2023] [Accepted: 10/19/2023] [Indexed: 10/24/2023]
Abstract
Parkinson's disease (PD) is a neurodegenerative disease showing uncontrollable motor symptoms that are primarily caused by the progressive loss of dopaminergic neurons in the brain. Currently no treatment exists to prevent PD progression. Therefore, discovery of new neuroprotective strategies still has great potential to benefit PD patients. A handful of studies show that activation of cAMP pathways is neuroprotective against PD progression. However, the neuroprotective role of this signaling cascade specifically in DA neurons has not been explored. In this study, fruit fly Drosophila melanogaster was used because of its sophisticated and powerful genetic approaches, especially with related to cAMP signaling pathway. We have investigated molecular mechanisms of neuroprotection in a fly larval model of PD by administering an environmental PD toxin rotenone. Increased cAMP signaling in the dunce mutant fly carrying defects in phosphodiesterase (PDE) gene, is neuroprotective against rotenone-induced locomotion deficits. Furthermore, the neuroprotective role of cAMP signaling specifically in DA neurons has been studied as it has not been explored. By using transgenic flies expressing designer receptors exclusively activated by designer drugs (DREADDs), we have shown that an increase of cAMP levels in DA neurons rescues rotenone-induced locomotion deficits. We also showed that this neuroprotection is mediated by activation of Gαs and PKA-C1 subunits. The results provide novel findings that expand our knowledge of neuroprotective mechanisms in DA neurons affecting PD progression, which could contribute to the development of new therapeutic treatments against PD. An important future study will explore downstream targets of cAMP-PKA signaling.
Collapse
Affiliation(s)
- Sazan Ismael
- Neuroscience Program, Dept. of Biological Sciences, Ohio University, Athens, OH 45701, USA; Dept. of Biology, Faculty of Science and Health, Koya University, Koya KOY45, Kurdistan Region-F.R., Iraq
| | - Robert A Colvin
- Neuroscience Program, Dept. of Biological Sciences, Ohio University, Athens, OH 45701, USA
| | - Daewoo Lee
- Neuroscience Program, Dept. of Biological Sciences, Ohio University, Athens, OH 45701, USA.
| |
Collapse
|
19
|
Walters ET. Exaptation and Evolutionary Adaptation in Nociceptor Mechanisms Driving Persistent Pain. BRAIN, BEHAVIOR AND EVOLUTION 2023; 98:314-330. [PMID: 38035556 PMCID: PMC10922759 DOI: 10.1159/000535552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 11/23/2023] [Indexed: 12/02/2023]
Abstract
BACKGROUND Several evolutionary explanations have been proposed for why chronic pain is a major clinical problem. One is that some mechanisms important for driving chronic pain, while maladaptive for modern humans, were adaptive because they enhanced survival. Evidence is reviewed for persistent nociceptor hyperactivity (PNH), known to promote chronic pain in rodents and humans, being an evolutionarily adaptive response to significant bodily injury, and primitive molecular mechanisms related to cellular injury and stress being exapted (co-opted or repurposed) to drive PNH and consequent pain. SUMMARY PNH in a snail (Aplysia californica), squid (Doryteuthis pealeii), fruit fly (Drosophila melanogaster), mice, rats, and humans has been documented as long-lasting enhancement of action potential discharge evoked by peripheral stimuli, and in some of these species as persistent extrinsically driven ongoing activity and/or intrinsic spontaneous activity (OA and SA, respectively). In mammals, OA and SA are often initiated within the protected nociceptor soma long after an inducing injury. Generation of OA or SA in nociceptor somata may be very rare in invertebrates, but prolonged afterdischarge in nociceptor somata readily occurs in sensitized Aplysia. Evidence for the adaptiveness of injury-induced PNH has come from observations of decreased survival of injured squid exposed to predators when PNH is blocked, from plausible survival benefits of chronic sensitization after severe injuries such as amputation, and from the functional coherence and intricacy of mammalian PNH mechanisms. Major contributions of cAMP-PKA signaling (with associated calcium signaling) to the maintenance of PNH both in mammals and molluscs suggest that this ancient stress signaling system was exapted early during the evolution of nociceptors to drive hyperactivity following bodily injury. Vertebrates have retained core cAMP-PKA signaling modules for PNH while adding new extracellular modulators (e.g., opioids) and cAMP-regulated ion channels (e.g., TRPV1 and Nav1.8 channels). KEY MESSAGES Evidence from multiple phyla indicates that PNH is a physiological adaptation that decreases the risk of attacks on injured animals. Core cAMP-PKA signaling modules make major contributions to the maintenance of PNH in molluscs and mammals. This conserved signaling has been linked to ancient cellular responses to stress, which may have been exapted in early nociceptors to drive protective hyperactivity that can persist while bodily functions recover after significant injury.
Collapse
Affiliation(s)
- Edgar T Walters
- Department of Integrative Biology and Pharmacology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, Texas, USA
| |
Collapse
|
20
|
Hasib RA, Ali MC, Rahman MH, Ahmed S, Sultana S, Summa SZ, Shimu MSS, Afrin Z, Jamal MAHM. Integrated gene expression profiling and functional enrichment analyses to discover biomarkers and pathways associated with Guillain-Barré syndrome and autism spectrum disorder to identify new therapeutic targets. J Biomol Struct Dyn 2023; 42:11299-11321. [PMID: 37776011 DOI: 10.1080/07391102.2023.2262586] [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: 03/15/2023] [Accepted: 09/17/2023] [Indexed: 10/01/2023]
Abstract
Guillain-Barré syndrome (GBS) is one of the most prominent and acute immune-mediated peripheral neuropathy, while autism spectrum disorders (ASD) are a group of heterogeneous neurodevelopmental disorders. The complete mechanism regarding the neuropathophysiology of these disorders is still ambiguous. Even after recent breakthroughs in molecular biology, the link between GBS and ASD remains a mystery. Therefore, we have implemented well-established bioinformatic techniques to identify potential biomarkers and drug candidates for GBS and ASD. 17 common differentially expressed genes (DEGs) were identified for these two disorders, which later guided the rest of the research. Common genes identified the protein-protein interaction (PPI) network and pathways associated with both disorders. Based on the PPI network, the constructed hub gene and module analysis network determined two common DEGs, namely CXCL9 and CXCL10, which are vital in predicting the top drug candidates. Furthermore, coregulatory networks of TF-gene and TF-miRNA were built to detect the regulatory biomolecules. Among drug candidates, imatinib had the highest docking and MM-GBSA score with the well-known chemokine receptor CXCR3 and remained stable during the 100 ns molecular dynamics simulation validated by the principal component analysis and the dynamic cross-correlation map. This study predicted the gene-based disease network for GBS and ASD and suggested prospective drug candidates. However, more in-depth research is required for clinical validation.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Rizone Al Hasib
- Department of Biotechnology and Genetic Engineering, Islamic University, Kushtia, Bangladesh
- Laboratory of Medical and Environmental Biotechnology Islamic University, Kushtia, Bangladesh
| | - Md Chayan Ali
- Department of Biochemistry, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Md Habibur Rahman
- Department of Computer Science and Engineering, Islamic University, Kushtia, Bangladesh
- Center for Advanced Bioinformatics and Artificial Intelligent Research, Islamic University, Kushtia, Bangladesh
| | - Sabbir Ahmed
- Department of Biotechnology and Genetic Engineering, Islamic University, Kushtia, Bangladesh
| | - Shaharin Sultana
- Department of Biotechnology and Genetic Engineering, Islamic University, Kushtia, Bangladesh
- Laboratory of Medical and Environmental Biotechnology Islamic University, Kushtia, Bangladesh
| | - Sadia Zannat Summa
- Department of Biotechnology and Genetic Engineering, Islamic University, Kushtia, Bangladesh
- Laboratory of Medical and Environmental Biotechnology Islamic University, Kushtia, Bangladesh
| | | | - Zinia Afrin
- Department of Biotechnology and Genetic Engineering, Islamic University, Kushtia, Bangladesh
| | - Mohammad Abu Hena Mostofa Jamal
- Department of Biotechnology and Genetic Engineering, Islamic University, Kushtia, Bangladesh
- Laboratory of Medical and Environmental Biotechnology Islamic University, Kushtia, Bangladesh
| |
Collapse
|
21
|
Mascioli I, Iapadre G, Ingrosso D, Donato GD, Giannini C, Salpietro V, Chiarelli F, Farello G. Brain and eye involvement in McCune-Albright Syndrome: clinical and translational insights. Front Endocrinol (Lausanne) 2023; 14:1092252. [PMID: 37274327 PMCID: PMC10235602 DOI: 10.3389/fendo.2023.1092252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 05/10/2023] [Indexed: 06/06/2023] Open
Abstract
McCune-Albright Syndrome (MAS) is a rare mosaic (post-zygotic) genetic disorder presenting with a broad continuum clinical spectrum. MAS arises from somatic, activating mutations in the GNAS gene, which induces a dysregulated Gsα-protein signaling in several tissues and an increased production of intracellular cyclic adenosine monophosphate (cAMP). Overall, MAS is a rare disorder affecting less than 1/100,000 children and, for this reason, data establishing genotype-phenotype correlations remain limited. Affected individuals clinically present with a variable combination of fibrous dysplasia of bone (FD), extra-skeletal manifestations (including cafeí-au-lait spots) and precocious puberty which might also be associated to broad hyperfunctioning endocrinopathies, and also gastrointestinal and cardiological involvement. Central nervous system (CNS) and eye involvement in MAS are among the less frequently described complications and remain largely uncharacterized. These rare complications mainly include neurodevelopmental abnormalities (e.g., delayed motor development, cognitive and language impairment), CNS anomalies (e.g., Chiari malformation type I) and a wide array of ophthalmological abnormalities often associated with vision loss. The pathophysiological mechanisms underlying abnormal neurological development have not been yet fully elucidated. The proposed mechanisms include a deleterious impact of chronically dysregulated Gsα-protein signaling on neurological function, or a secondary (damaging) effect of (antenatal and/or early postnatal) hypercortisolism on early pre- and post-natal CNS development. In this Review, we summarize the main neurological and ophthalmological features eventually associated with the MAS spectrum, also providing a detailed overview of the potential pathophysiological mechanisms underlying these clinical complications.
Collapse
Affiliation(s)
- Ilaria Mascioli
- Department of Pediatrics, University of Chieti, Chieti, Italy
| | - Giulia Iapadre
- Department of Pediatrics, University of L’Aquila, L’Aquila, Italy
| | | | - Giulio Di Donato
- Department of Pediatrics, University of L’Aquila, L’Aquila, Italy
| | - Cosimo Giannini
- Department of Pediatrics, University of Chieti, Chieti, Italy
| | | | | | - Giovanni Farello
- Department of Pediatrics, University of L’Aquila, L’Aquila, Italy
| |
Collapse
|
22
|
Fabian B, Sachse S. Experience-dependent plasticity in the olfactory system of Drosophila melanogaster and other insects. Front Cell Neurosci 2023; 17:1130091. [PMID: 36923450 PMCID: PMC10010147 DOI: 10.3389/fncel.2023.1130091] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 02/07/2023] [Indexed: 02/24/2023] Open
Abstract
It is long known that the nervous system of vertebrates can be shaped by internal and external factors. On the other hand, the nervous system of insects was long assumed to be stereotypic, although evidence for plasticity effects accumulated for several decades. To cover the topic comprehensively, this review recapitulates the establishment of the term "plasticity" in neuroscience and introduces its original meaning. We describe the basic composition of the insect olfactory system using Drosophila melanogaster as a representative example and outline experience-dependent plasticity effects observed in this part of the brain in a variety of insects, including hymenopterans, lepidopterans, locusts, and flies. In particular, we highlight recent advances in the study of experience-dependent plasticity effects in the olfactory system of D. melanogaster, as it is the most accessible olfactory system of all insect species due to the genetic tools available. The partly contradictory results demonstrate that morphological, physiological and behavioral changes in response to long-term olfactory stimulation are more complex than previously thought. Different molecular mechanisms leading to these changes were unveiled in the past and are likely responsible for this complexity. We discuss common problems in the study of experience-dependent plasticity, ways to overcome them, and future directions in this area of research. In addition, we critically examine the transferability of laboratory data to natural systems to address the topic as holistically as possible. As a mechanism that allows organisms to adapt to new environmental conditions, experience-dependent plasticity contributes to an animal's resilience and is therefore a crucial topic for future research, especially in an era of rapid environmental changes.
Collapse
Affiliation(s)
| | - Silke Sachse
- Research Group Olfactory Coding, Max Planck Institute for Chemical Ecology, Jena, Germany
| |
Collapse
|
23
|
Alharbi M, Alshammari A, Kaur G, Kalra S, Mehan S, Suri M, Chhabra S, Kumar N, Alanazi WA, Alshanwani AR, AL-Ghamdi AH, Narula AS, Kalfin R. Effect of Natural Adenylcyclase/cAMP/CREB Signalling Activator Forskolin against Intra-Striatal 6-OHDA-Lesioned Parkinson's Rats: Preventing Mitochondrial, Motor and Histopathological Defects. Molecules 2022; 27:7951. [PMID: 36432051 PMCID: PMC9695774 DOI: 10.3390/molecules27227951] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 11/07/2022] [Accepted: 11/10/2022] [Indexed: 11/18/2022] Open
Abstract
Parkinson's disease (PD) is characterised by dopaminergic neuronal loss in the brain area. PD is a complex disease that deteriorates patients' motor and non-motor functions. In experimental animals, the neurotoxin 6-OHDA induces neuropathological, behavioural, neurochemical and mitochondrial abnormalities and the formation of free radicals, which is related to Parkinson-like symptoms after inter-striatal 6-OHDA injection. Pathological manifestations of PD disrupt the cAMP/ATP-mediated activity of the transcription factor CREB, resulting in Parkinson's-like symptoms. Forskolin (FSK) is a direct AC/cAMP/CREB activator isolated from Coleus forskohlii with various neuroprotective properties. FSK has already been proven in our laboratory to directly activate the enzyme adenylcyclase (AC) and reverse the neurodegeneration associated with the progression of Autism, Multiple Sclerosis, ALS, and Huntington's disease. Several behavioural paradigms were used to confirm the post-lesion effects, including the rotarod, open field, grip strength, narrow beam walk (NBW) and Morris water maze (MWM) tasks. Our results were supported by examining brain cellular, molecular, mitochondrial and histopathological alterations. The FSK treatment (15, 30 and 45 mg/kg, orally) was found to be effective in restoring behavioural and neurochemical defects in a 6-OHDA-induced experimental rat model of PD. As a result, the current study successfully contributes to the investigation of FSK's neuroprotective role in PD prevention via the activation of the AC/cAMP/PKA-driven CREB pathway and the restoration of mitochondrial ETC-complex enzymes.
Collapse
Affiliation(s)
- Metab Alharbi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Post Box 2455, Riyadh 11451, Saudi Arabia
| | - Abdulrahman Alshammari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Post Box 2455, Riyadh 11451, Saudi Arabia
| | - Gurpreet Kaur
- Department of Pharmacology, Rajendra Institute of Technology and Sciences, Hisar Road, 4th Mile Stone, Sirsa, Haryana 125055, India
| | - Sanjeev Kalra
- Department of Pharmacology, Rajendra Institute of Technology and Sciences, Hisar Road, 4th Mile Stone, Sirsa, Haryana 125055, India
| | - Sidharth Mehan
- Department of Pharmacology, Rajendra Institute of Technology and Sciences, Hisar Road, 4th Mile Stone, Sirsa, Haryana 125055, India
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy (An Autonomous College), Moga 142001, Punjab, India
| | - Manisha Suri
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy (An Autonomous College), Moga 142001, Punjab, India
| | - Swesha Chhabra
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy (An Autonomous College), Moga 142001, Punjab, India
| | - Nitish Kumar
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy (An Autonomous College), Moga 142001, Punjab, India
| | - Wael A. Alanazi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Post Box 2455, Riyadh 11451, Saudi Arabia
| | - Aliah R. Alshanwani
- Physiology Department, College of Medicine & King Khalid University Hospital, King Saud University, Riyadh 12372, Saudi Arabia
| | - Abdullah Hamed AL-Ghamdi
- Pharmaceutical Care Department, Namerah General Hospital, Ministry of Health, Namerah 65439, Saudi Arabia
| | | | - Reni Kalfin
- Institute of Neurobiology, Bulgarian Academy of Sciences, Acad. G. Bonchev St., Block 23, 1113 Sofia, Bulgaria
- Department of Healthcare, South-West University “NeofitRilski”, Ivan Mihailov St. 66, 2700 Blagoevgrad, Bulgaria
| |
Collapse
|
24
|
Kapoor T, Mehan S, Suri M, Sharma N, Kumar N, Narula AS, Alshammari A, Alasmari AF, Alharbi M, Assiri MA, Kalfin R. Forskolin, an Adenylcyclase/cAMP/CREB Signaling Activator Restoring Myelin-Associated Oligodendrocyte Destruction in Experimental Ethidium Bromide Model of Multiple Sclerosis. Cells 2022; 11:cells11182771. [PMID: 36139346 PMCID: PMC9497421 DOI: 10.3390/cells11182771] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Revised: 08/28/2022] [Accepted: 08/29/2022] [Indexed: 11/16/2022] Open
Abstract
Multiple sclerosis (MS) is a chronic neurodegenerative disease marked by oligodendrocyte loss, which results in central neuronal demyelination. AC/cAMP/CREB signaling dysregulation is involved in the progression of MS, including mitochondrial dysfunctions, reduction in nerve growth factors, neuronal inflammation, apoptosis, and white matter degeneration. Our previous research has shown that Forskolin (FSK), a naturally occurring direct adenylyl cyclase (AC)/cAMP/CREB activator, has neuroprotective potential to alleviate pathogenic factors linked with numerous neurological abnormalities. The current study intends to explore the neuroprotective potential of FSK at doses of 40 mg/kg and 60 mg/kg alone, as well as in combination with conventional medicines, such as Fingolimod (FNG), Donepezil (DON), Memantine (MEM), and Simvastatin (SIM) in EB-induced demyelinated experimental MS rats. Adult Wistar rats were divided into nine groups, and EB was infused stereotaxically in the rat brain’s intracerebropeduncle (ICP) area. Chronic gliotoxin EB treatment results in demyelination as well as motor and cognitive dysfunctions. FSK, combined with standard medications, improves behavioral dysfunctions, such as neuromuscular and motor deficits and memory and cognitive abnormalities. Following pharmacological treatments improved remyelination by enhancing myelin basic protein and increasing AC, cAMP, and CREB levels in brain homogenates. Furthermore, FSK therapy restored brain mitochondrial-ETC complex enzymes and neurotransmitter levels while decreasing inflammatory cytokines and oxidative stress markers. The Luxol fast blue (LFB) stain results further indicate FSK’s neuroprotective potential in preventing oligodendrocyte death. Therefore, the results of these studies contribute to a better understanding of the possible role that natural phytochemicals FSK could have in preventing motor neuron diseases, such as multiple sclerosis.
Collapse
Affiliation(s)
- Tarun Kapoor
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy (An Autonomous College), Moga 142001, Punjab, India
| | - Sidharth Mehan
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy (An Autonomous College), Moga 142001, Punjab, India
- Correspondence: or ; Tel.: +1-91-8059889909
| | - Manisha Suri
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy (An Autonomous College), Moga 142001, Punjab, India
| | - Nidhi Sharma
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy (An Autonomous College), Moga 142001, Punjab, India
| | - Nitish Kumar
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy (An Autonomous College), Moga 142001, Punjab, India
| | | | - Abdulrahman Alshammari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia
| | - Abdullah F. Alasmari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia
| | - Metab Alharbi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia
| | - Mohammed A. Assiri
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia
| | - Reni Kalfin
- Institute of Neurobiology, Bulgarian Academy of Sciences, Acad. G. Bonchev St., Block 23, 1113 Sofia, Bulgaria
- Department of Healthcare, South-West University “NeofitRilski”, Ivan Mihailov St. 66, 2700 Blagoevgrad, Bulgaria
| |
Collapse
|
25
|
Wang Y, Nong Y, Zhang X, Mai T, Cai J, Liu J, Lai KP, Zhang Z. Comparative plasma metabolomic analysis to identify biomarkers for lead-induced cognitive impairment. Chem Biol Interact 2022; 366:110143. [PMID: 36063854 DOI: 10.1016/j.cbi.2022.110143] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 08/24/2022] [Accepted: 08/29/2022] [Indexed: 11/03/2022]
Abstract
BACKGROUND Lead (Pb), an environmental neurotoxicant, is known to induce cognitive impairment. Neuroinflammation and oxidative stress in the brain tissue are common pathogenetic links to Pb-induced cognitive impairment. There are no existing biomarkers to evaluate Pb-reduced cognition. Plasma metabolites are the readout of the biological functions of the host, making it a potential biomarker for assessing heavy metal-induced cognitive impairment. METHODS The present report aims to identify the plasma metabolite changes under conditions of high plasma Pb levels and low cognition. RESULTS We conducted a comparative plasma metabolomic analysis on two groups of adults those with low plasma Pb level and high cognition vs. those with high plasma Pb level and low cognition and identified 20 dysregulated metabolites. In addition, we found a significant reduction in docosahexaenoic acid, glycoursodeoxycholic acid, and arachidonic acid, and significant induction of p-cresol sulfate and phenylacetyl-l-glutamine. Gene Ontology enrichment analysis highlighted the importance of these plasma metabolites in brain functions and neurodegenerative diseases such as Parkinson's disease. CONCLUSIONS The findings of this report provide novel insights into the use of plasma metabolites to assess metal-induced cognitive impairment.
Collapse
Affiliation(s)
- Yuqin Wang
- Key Laboratory of Environmental Pollution and Integrative Omics, Guilin Medical University, Education Department of Guangxi Zhuang Autonomous Region, Guilin, China
| | - Yuan Nong
- Department of Neurology (Area Two), Guigang City People's Hospital, The Eighth Affiliated Hospital of Guangxi Medical University, Guigang, China
| | - Xing Zhang
- Key Laboratory of Environmental Pollution and Integrative Omics, Guilin Medical University, Education Department of Guangxi Zhuang Autonomous Region, Guilin, China
| | - Tingyu Mai
- Department of Environmental Health and Occupational Medicine, School of Public Health, Guilin Medical University, Guilin, 541004, Guangxi, China
| | - Jiansheng Cai
- Guangxi Key Laboratory of Tumor Immunology and Microenvironmental Regulation, Guilin Medical University, Guilin, China
| | - Jiaqi Liu
- Key Laboratory of Environmental Pollution and Integrative Omics, Guilin Medical University, Education Department of Guangxi Zhuang Autonomous Region, Guilin, China.
| | - Keng Po Lai
- Key Laboratory of Environmental Pollution and Integrative Omics, Guilin Medical University, Education Department of Guangxi Zhuang Autonomous Region, Guilin, China.
| | | |
Collapse
|
26
|
Sidoli M, Chen LC, Lu AJ, Wandless TJ, Talbot WS. A cAMP Sensor Based on Ligand-Dependent Protein Stabilization. ACS Chem Biol 2022; 17:2024-2030. [PMID: 35839076 PMCID: PMC9396618 DOI: 10.1021/acschembio.2c00333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 07/12/2022] [Indexed: 11/28/2022]
Abstract
cAMP is a ubiquitous second messenger with many functions in diverse organisms. Current cAMP sensors, including Föster resonance energy transfer (FRET)-based and single-wavelength-based sensors, allow for real time visualization of this small molecule in cultured cells and in some cases in vivo. Nonetheless the observation of cAMP in living animals is still difficult, typically requiring specialized microscopes and ex vivo tissue processing. Here we used ligand-dependent protein stabilization to create a new cAMP sensor. This sensor allows specific and sensitive detection of cAMP in living zebrafish embryos, which may enable new understanding of the functions of cAMP in living vertebrates.
Collapse
Affiliation(s)
- Mariapaola Sidoli
- Department
of Developmental Biology, School of Medicine, Stanford University, Stanford, California 94305, United States
| | - Ling-chun Chen
- Department
of Chemical and Systems Biology, School of Medicine, Stanford University, Stanford, California 94305, United States
| | - Alexander J. Lu
- Department
of Chemical and Systems Biology, School of Medicine, Stanford University, Stanford, California 94305, United States
| | - Thomas J. Wandless
- Department
of Chemical and Systems Biology, School of Medicine, Stanford University, Stanford, California 94305, United States
| | - William S. Talbot
- Department
of Developmental Biology, School of Medicine, Stanford University, Stanford, California 94305, United States
| |
Collapse
|
27
|
Yan Y, Gao S, Avasthi S, Zhao Y, Ye J, Tao Y, Wang W, Zhu X, Du F, O'Donnell JM, Xu Y. Protective effects of phosphodiesterase 2 inhibitor against Aβ 1-42 induced neuronal toxicity. Neuropharmacology 2022; 213:109128. [PMID: 35588859 DOI: 10.1016/j.neuropharm.2022.109128] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 04/19/2022] [Accepted: 05/09/2022] [Indexed: 01/12/2023]
Abstract
Our previous study suggested that inhibition of Phosphodiesterase 2 ameliorates memory loss upon exposure to oxidative stress. While whether memory enhancing effects of PDE2 inhibition on Alzheimer's disease mouse model are involved in antioxidant defense and neuronal remodeling, are largely unexplored. The present study addressed whether and how PDE2 inhibitor Bay 60-7550 rescued Aβ oligomers (Aβo)-induced neuronal damage and memory impairment. The results suggested that exposure of primary cortical neurons to Aβo induced neuronal cells damage and increased PDE2 expression, which were paralleled to an increase in the oxidative parameter malondialdehyde (MDA) level and cellular apoptosis. However, this Aβo-induced oxidative damage was blocked by pre-treatment with protein kinase A or G (PKA or PKG) inhibitor, suggesting the involvement of cAMP/cGMP signaling. Moreover, microinjection of Aβo into the prefrontal cortex of mice increased the MDA level; while Bay 60-7550 reversed this effect and increased antioxidant and anti-apoptotic factors, i.e. increased trolox-equivalent-antioxidant capacity and Bcl-2/Bax ratio. Bay 60-7550 also rescued Aβo-induced synaptic atrophy and memory deficits, as evidenced by the increased synaptic proteins' levels and spine density in the prefrontal cortex, and improved cognitive behaviors by decreased working memory errors in the eight-arm maze and increased discrimination index in the novel object recognition test. These findings suggest that inhibition of PDE2 contributes to antioxidant defense and neuronal remodeling by regulation of cAMP/cGMP signaling, which provide a theoretical basis for the future use of PDE2 inhibitors as the anti-AD drugs.
Collapse
Affiliation(s)
- Yuqing Yan
- Department of Anesthesiology, Rutgers New Jersey Medical School, The State University of New Jersey, Newark, NJ, 07101, USA; Department of Pharmaceutical Sciences, School of Pharmacy & Pharmaceutical Sciences, University at Buffalo, The State University of New York, Buffalo, NY, 14214, USA
| | - Shichao Gao
- Department of Pharmaceutical Sciences, School of Pharmacy & Pharmaceutical Sciences, University at Buffalo, The State University of New York, Buffalo, NY, 14214, USA
| | - Shivani Avasthi
- Department of Pharmaceutical Sciences, School of Pharmacy & Pharmaceutical Sciences, University at Buffalo, The State University of New York, Buffalo, NY, 14214, USA
| | - Yuhan Zhao
- Department of Pharmaceutical Sciences, School of Pharmacy & Pharmaceutical Sciences, University at Buffalo, The State University of New York, Buffalo, NY, 14214, USA
| | - Jianghong Ye
- Department of Anesthesiology, Rutgers New Jersey Medical School, The State University of New Jersey, Newark, NJ, 07101, USA
| | - Yuanxiang Tao
- Department of Anesthesiology, Rutgers New Jersey Medical School, The State University of New Jersey, Newark, NJ, 07101, USA
| | - Wei Wang
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, AZ, 85721, USA
| | - Xiongwei Zhu
- Department of Pathology, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Fu Du
- FD Neurotechnologies, Inc., Elicott City, MD, 21041, USA
| | - James M O'Donnell
- Department of Pharmaceutical Sciences, School of Pharmacy & Pharmaceutical Sciences, University at Buffalo, The State University of New York, Buffalo, NY, 14214, USA
| | - Ying Xu
- Department of Anesthesiology, Rutgers New Jersey Medical School, The State University of New Jersey, Newark, NJ, 07101, USA.
| |
Collapse
|
28
|
Kawata S, Mukai Y, Nishimura Y, Takahashi T, Saitoh N. Green fluorescent cAMP indicator of high speed and specificity suitable for neuronal live-cell imaging. Proc Natl Acad Sci U S A 2022; 119:e2122618119. [PMID: 35867738 PMCID: PMC9282276 DOI: 10.1073/pnas.2122618119] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 04/29/2022] [Indexed: 11/18/2022] Open
Abstract
Cyclic adenosine monophosphate (cAMP) is a canonical intracellular messenger playing diverse roles in cell functions. In neurons, cAMP promotes axonal growth during early development, and mediates sensory transduction and synaptic plasticity after maturation. The molecular cascades of cAMP are well documented, but its spatiotemporal profiles associated with neuronal functions remain hidden. Hence, we developed a genetically encoded cAMP indicator based on a bacterial cAMP-binding protein. This indicator "gCarvi" monitors [cAMP]i at 0.2 to 20 µM with a subsecond time resolution and a high specificity over cyclic guanosine monophosphate (cGMP). gCarvi can be converted to a ratiometric probe for [cAMP]i quantification and its expression can be specifically targeted to various subcellular compartments. Monomeric gCarvi also enables simultaneous multisignal monitoring in combination with other indicators. As a proof of concept, simultaneous cAMP/Ca2+ imaging in hippocampal neurons revealed a tight linkage of cAMP to Ca2+ signals. In cerebellar presynaptic boutons, forskolin induced nonuniform cAMP elevations among boutons, which positively correlated with subsequent increases in the size of the recycling pool of synaptic vesicles assayed using FM dye. Thus, the cAMP domain in presynaptic boutons is an important determinant of the synaptic strength.
Collapse
Affiliation(s)
- Seiko Kawata
- Department of Neurophysiology, Graduate School of Life and Medical Sciences, Doshisha University, Kyoto, 610-0394, Japan
| | - Yuki Mukai
- Department of Neurophysiology, Graduate School of Life and Medical Sciences, Doshisha University, Kyoto, 610-0394, Japan
| | - Yumi Nishimura
- Department of Neurophysiology, Graduate School of Life and Medical Sciences, Doshisha University, Kyoto, 610-0394, Japan
| | - Tomoyuki Takahashi
- Cellular and Molecular Synaptic Function Unit, Okinawa Institute of Science and Technology Graduate University, Okinawa, 904-0495, Japan
| | - Naoto Saitoh
- Department of Neurophysiology, Graduate School of Life and Medical Sciences, Doshisha University, Kyoto, 610-0394, Japan
| |
Collapse
|
29
|
The Role of NLRP3 Inflammasome Activation and Oxidative Stress in Varicocele-Mediated Male Hypofertility. Int J Mol Sci 2022; 23:ijms23095233. [PMID: 35563625 PMCID: PMC9102453 DOI: 10.3390/ijms23095233] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 05/03/2022] [Accepted: 05/05/2022] [Indexed: 12/11/2022] Open
Abstract
Varicocele (VC) is the most common abnormality identified in men evaluated for hypofertility. Increased levels of reactive oxygen species (ROS) and reduced antioxidants concentrations are key contributors in varicocele-mediated hypofertility. Moreover, inflammation and alterations in testicular immunity negatively impact male fertility. In particular, NLRP3 inflammasome activation was hypothesized to lead to seminal inflammation, in which the levels of specific cytokines, such as IL-1β and IL-18, are overexpressed. In this review, we described the role played by oxidative stress (OS), inflammation, and NLRP3 inflammasome activation in VC disease. The consequences of ROS overproduction in testis, including inflammation, lipid peroxidation, mitochondrial dysfunction, chromatin damage, and sperm DNA fragmentation, leading to abnormal testicular function and failed spermatogenesis, were highlighted. Finally, we described some therapeutic antioxidant strategies, with recognized beneficial effects in counteracting OS and inflammation in testes, as possible therapeutic drugs against varicocele-mediated hypofertility.
Collapse
|
30
|
Sears JC, Broadie K. Temporally and Spatially Localized PKA Activity within Learning and Memory Circuitry Regulated by Network Feedback. eNeuro 2022; 9:ENEURO.0450-21.2022. [PMID: 35301221 PMCID: PMC8982635 DOI: 10.1523/eneuro.0450-21.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2021] [Revised: 02/18/2022] [Accepted: 03/12/2022] [Indexed: 12/02/2022] Open
Abstract
Dynamic functional connectivity within brain circuits requires coordination of intercellular signaling and intracellular signal transduction. Critical roles for cAMP-dependent protein kinase A (PKA) signaling are well established in the Drosophila mushroom body (MB) learning and memory circuitry, but local PKA activity within this well-mapped neuronal network is uncharacterized. Here, we use an in vivo PKA activity sensor (PKA-SPARK) to test spatiotemporal regulatory requirements in the MB axon lobes. We find immature animals have little detectable PKA activity, whereas postcritical period adults show high field-selective activation primarily in just 3/16 defined output regions. In addition to the age-dependent PKA activity in distinct α'/β' lobe nodes, females show sex-dependent elevation compared with males in these same restricted regions. Loss of neural cell body Fragile X mental retardation protein (FMRP) and Rugose [human Neurobeachin (NBEA)] suppresses localized PKA activity, whereas overexpression (OE) of MB lobe PKA-synergist Meng-Po (human SBK1) promotes PKA activity. Elevated Meng-Po subverts the PKA age-dependence, with elevated activity in immature animals, and spatial-restriction, with striking γ lobe activity. Testing circuit signaling requirements with temperature-sensitive shibire (human Dynamin) blockade, we find broadly expanded PKA activity within the MB lobes. Using transgenic tetanus toxin to block MB synaptic output, we find greatly heightened PKA activity in virtually all MB lobe fields, although the age-dependence is maintained. We conclude spatiotemporally restricted PKA activity signaling within this well-mapped learning/memory circuit is age-dependent and sex-dependent, driven by FMRP-Rugose pathway activation, temporally promoted by Meng-Po kinase function, and restricted by output neurotransmission providing network feedback.
Collapse
Affiliation(s)
- James C Sears
- Department of Biological Sciences, Vanderbilt University and Medical Center, Nashville, TN 37235
- Vanderbilt Brain Institute, Vanderbilt University and Medical Center, Nashville, TN 37235
| | - Kendal Broadie
- Department of Biological Sciences, Vanderbilt University and Medical Center, Nashville, TN 37235
- Vanderbilt Brain Institute, Vanderbilt University and Medical Center, Nashville, TN 37235
- Department of Cell and Developmental Biology, Vanderbilt University and Medical Center, Nashville, TN 37235
- Department of Pharmacology, Vanderbilt University and Medical Center, Nashville, TN 37235
| |
Collapse
|
31
|
Philyaw TJ, Rothenfluh A, Titos I. The Use of Drosophila to Understand Psychostimulant Responses. Biomedicines 2022; 10:119. [PMID: 35052798 PMCID: PMC8773124 DOI: 10.3390/biomedicines10010119] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 12/31/2021] [Accepted: 12/31/2021] [Indexed: 01/27/2023] Open
Abstract
The addictive properties of psychostimulants such as cocaine, amphetamine, methamphetamine, and methylphenidate are based on their ability to increase dopaminergic neurotransmission in the reward system. While cocaine and methamphetamine are predominately used recreationally, amphetamine and methylphenidate also work as effective therapeutics to treat symptoms of disorders including attention deficit and hyperactivity disorder (ADHD) and autism spectrum disorder (ASD). Although both the addictive properties of psychostimulant drugs and their therapeutic efficacy are influenced by genetic variation, very few genes that regulate these processes in humans have been identified. This is largely due to population heterogeneity which entails a requirement for large samples. Drosophila melanogaster exhibits similar psychostimulant responses to humans, a high degree of gene conservation, and allow performance of behavioral assays in a large population. Additionally, amphetamine and methylphenidate reduce impairments in fly models of ADHD-like behavior. Therefore, Drosophila represents an ideal translational model organism to tackle the genetic components underlying the effects of psychostimulants. Here, we break down the many assays that reliably quantify the effects of cocaine, amphetamine, methamphetamine, and methylphenidate in Drosophila. We also discuss how Drosophila is an efficient and cost-effective model organism for identifying novel candidate genes and molecular mechanisms involved in the behavioral responses to psychostimulant drugs.
Collapse
Affiliation(s)
- Travis James Philyaw
- Molecular Biology Graduate Program, University of Utah, Salt Lake City, UT 84112, USA;
| | - Adrian Rothenfluh
- Department of Psychiatry, Huntsman Mental Health Institute, University of Utah, Salt Lake City, UT 84108, USA
- Molecular Medicine Program, University of Utah, Salt Lake City, UT 84112, USA
- Department of Neurobiology, University of Utah, Salt Lake City, UT 84132, USA
- Department of Human Genetics, University of Utah, Salt Lake City, UT 84112, USA
| | - Iris Titos
- Molecular Medicine Program, University of Utah, Salt Lake City, UT 84112, USA
| |
Collapse
|
32
|
Palumbos SD, Skelton R, McWhirter R, Mitchell A, Swann I, Heifner S, Von Stetina S, Miller DM. cAMP controls a trafficking mechanism that maintains the neuron specificity and subcellular placement of electrical synapses. Dev Cell 2021; 56:3235-3249.e4. [PMID: 34741804 DOI: 10.1016/j.devcel.2021.10.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 08/30/2021] [Accepted: 10/08/2021] [Indexed: 11/28/2022]
Abstract
Electrical synapses are established between specific neurons and within distinct subcellular compartments, but the mechanisms that direct gap junction assembly in the nervous system are largely unknown. Here, we show that a developmental program tunes cAMP signaling to direct the neuron-specific assembly and placement of electrical synapses in the C. elegans motor circuit. We use live-cell imaging to visualize electrical synapses in vivo and an optogenetic assay to confirm that they are functional. In ventral A class (VA) motor neurons, the UNC-4 transcription factor blocks expression of cAMP antagonists that promote gap junction miswiring. In unc-4 mutants, VA electrical synapses are established with an alternative synaptic partner and are repositioned from the VA axon to soma. cAMP counters these effects by driving gap junction trafficking into the VA axon for electrical synapse assembly. Thus, our experiments establish that cAMP regulates gap junction trafficking for the biogenesis of functional electrical synapses.
Collapse
Affiliation(s)
- Sierra D Palumbos
- Neuroscience Program, Vanderbilt University, Nashville, TN 37212, USA
| | - Rachel Skelton
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN 37212, USA
| | - Rebecca McWhirter
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN 37212, USA
| | - Amanda Mitchell
- Vanderbilt Summer Science Academy, Vanderbilt University, Nashville, TN 37212, USA
| | - Isaiah Swann
- Vanderbilt Summer Science Academy, Vanderbilt University, Nashville, TN 37212, USA
| | | | - Stephen Von Stetina
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN 37212, USA
| | - David M Miller
- Neuroscience Program, Vanderbilt University, Nashville, TN 37212, USA; Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN 37212, USA.
| |
Collapse
|
33
|
Jankowska A, Satała G, Świerczek A, Pociecha K, Partyka A, Jastrzębska-Więsek M, Głuch-Lutwin M, Bojarski AJ, Wyska E, Chłoń-Rzepa G. A new class of 5-HT 1A receptor antagonists with procognitive and antidepressant properties. Future Med Chem 2021; 13:1497-1514. [PMID: 34253032 DOI: 10.4155/fmc-2020-0363] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Aims: 5-HT1A receptor antagonists constitute a potential group of drugs in the treatment of CNS diseases. The aim of this study was to search for new procognitive and antidepressant drugs among amide derivatives of aminoalkanoic acids with 5-HT1A receptor antagonistic properties. Materials & methods: Thirty-three amides were designed and evaluated in silico for their drug-likeness. The synthesized compounds were tested in vitro for their 5-HT1A receptor affinity and functional profile. Moreover, their selectivity over 5-HT7, 5-HT2A and D2 receptors and ability to inhibit phosphodiesterases were evaluated. Results: A selected 5-HT1A receptor antagonist 20 (Ki = 35 nM, Kb = 4.9 nM) showed procognitive and antidepressant activity in vivo. Conclusion: Novel 5-HT1A receptor antagonists were discovered and shown as potential psychotropic drugs.
Collapse
Affiliation(s)
- Agnieszka Jankowska
- Jagiellonian University Medical College, Faculty of Pharmacy, Department of Medicinal Chemistry, 9 Medyczna Street, 30-688, Kraków, Poland
| | - Grzegorz Satała
- Polish Academy of Sciences, Maj Institute of Pharmacology, Department of Medicinal Chemistry, 12 Smętna Street, 31-343, Kraków, Poland
| | - Artur Świerczek
- Jagiellonian University Medical College, Faculty of Pharmacy, Department of Pharmacokinetics & Physical Pharmacy, 9 Medyczna Street, 30-688, Kraków, Poland
| | - Krzysztof Pociecha
- Jagiellonian University Medical College, Faculty of Pharmacy, Department of Pharmacokinetics & Physical Pharmacy, 9 Medyczna Street, 30-688, Kraków, Poland
| | - Anna Partyka
- Jagiellonian University Medical College, Faculty of Pharmacy, Department of Clinical Pharmacy, 9 Medyczna Street, 30-688, Kraków, Poland
| | - Magdalena Jastrzębska-Więsek
- Jagiellonian University Medical College, Faculty of Pharmacy, Department of Clinical Pharmacy, 9 Medyczna Street, 30-688, Kraków, Poland
| | - Monika Głuch-Lutwin
- Jagiellonian University Medical College, Faculty of Pharmacy, Department of Pharmacobiology, 9 Medyczna Street, 30-688, Kraków, Poland
| | - Andrzej J Bojarski
- Polish Academy of Sciences, Maj Institute of Pharmacology, Department of Medicinal Chemistry, 12 Smętna Street, 31-343, Kraków, Poland
| | - Elżbieta Wyska
- Jagiellonian University Medical College, Faculty of Pharmacy, Department of Pharmacokinetics & Physical Pharmacy, 9 Medyczna Street, 30-688, Kraków, Poland
| | - Grażyna Chłoń-Rzepa
- Jagiellonian University Medical College, Faculty of Pharmacy, Department of Medicinal Chemistry, 9 Medyczna Street, 30-688, Kraków, Poland
| |
Collapse
|
34
|
Inhibiting PDE7A Enhances the Protective Effects of Neural Stem Cells on Neurodegeneration and Memory Deficits in Sevoflurane-Exposed Mice. eNeuro 2021; 8:ENEURO.0071-21.2021. [PMID: 34135002 PMCID: PMC8266220 DOI: 10.1523/eneuro.0071-21.2021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 05/21/2021] [Accepted: 05/25/2021] [Indexed: 12/03/2022] Open
Abstract
Sevoflurane is widely used in general anesthesia, especially for children. However, prolonged exposure to sevoflurane is reported to be associated with adverse effects on the development of brain in infant monkey. Neural stem cells (NSCs), with potent proliferation, differentiation, and renewing ability, provide an encouraging tool for basic research and clinical therapies for neurodegenerative diseases. We aim to explore the functional effects of injecting NSCs with phosphodiesterase 7A (PDE7A) knock-down in infant mice exposed to sevoflurane. The effects of PDE7A in NSCs proliferation and differentiation were determined by cell counting kit-8 (CCK-8) assay and differentiation-related gene expression assay, respectively. The effects of NSCs with modified PDE7A on mice’s long-term memory and learning ability were assessed by behavioral assays. Our data demonstrated that depleting PDE7A promoted, whereas forcing PDE7A suppressed the activation of cAMP/cAMP-response element binding protein (CREB) signaling as well as cell proliferation and neuronal differentiation of NSCs. Inhibition of PDE7A in NSCs exhibited profound improved effects on long-term memory and learning ability of mice exposed to sevoflurane. Our results for the first time show that knock-down of PDE7A improves the neurogenesis of NSCs in vitro and in vivo, and is beneficial for alleviating sevoflurane-induced brain damage in infant mice.
Collapse
|
35
|
Pentoxifylline Enhances Antioxidative Capability and Promotes Mitochondrial Biogenesis in D-Galactose-Induced Aging Mice by Increasing Nrf2 and PGC-1 α through the cAMP-CREB Pathway. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:6695613. [PMID: 34257818 PMCID: PMC8245236 DOI: 10.1155/2021/6695613] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 04/21/2021] [Accepted: 05/13/2021] [Indexed: 02/07/2023]
Abstract
Aging is a complex phenomenon associated with oxidative stress and mitochondrial dysfunction. The objective of this study was to investigate the potential ameliorative effects of the phosphodiesterase inhibitor pentoxifylline (PTX) on the aging process and its underlying mechanisms. We treated D-galactose- (D-gal-) induced aging mice with PTX and measured the changes in behavior, degree of oxidative damage, and mitochondrial ultrastructure and content as well as the expression of nuclear factor erythroid 2-related factor 2- (Nrf2-) mediated antioxidant genes and peroxisome proliferator-activated receptor-gamma coactivator 1-alpha- (PGC-1α-) dependent mitochondrial biogenesis genes. The results demonstrated that PTX improved cognitive deficits, reduced oxidative damage, ameliorated abnormal mitochondrial ultrastructure, increased mitochondrial content and Nrf2 activation, and upregulated antioxidant and mitochondrial biogenesis gene expression in the hippocampus of wild-type aging mice. However, the above antiaging effects of PTX were obviously decreased in the brains of Nrf2-deficient D-gal-induced aging mice. Moreover, in hydrogen peroxide-treated SH-SY5Y cells, we found that cyclic adenosine monophosphate (cAMP) response element-binding protein (CREB) and Nrf2/PGC-1α act in a linear way by CREB siRNA transfection. Thus, PTX administration improved the aging-related decline in brain function by enhancing antioxidative capability and promoting mitochondrial biogenesis, which might depend on increasing Nrf2 and PGC-1α by activating the cAMP-CREB pathway.
Collapse
|
36
|
Abstract
Circadian clocks are biochemical time-keeping machines that synchronize animal behavior and physiology with planetary rhythms. In Drosophila, the core components of the clock comprise a transcription/translation feedback loop and are expressed in seven neuronal clusters in the brain. Although it is increasingly evident that the clocks in each of the neuronal clusters are regulated differently, how these clocks communicate with each other across the circadian neuronal network is less clear. Here, we review the latest evidence that describes the physical connectivity of the circadian neuronal network . Using small ventral lateral neurons as a starting point, we summarize how one clock may communicate with another, highlighting the signaling pathways that are both upstream and downstream of these clocks. We propose that additional efforts are required to understand how temporal information generated in each circadian neuron is integrated across a neuronal circuit to regulate rhythmic behavior.
Collapse
Affiliation(s)
- Myra Ahmad
- Department of Pediatrics, Division of Medical Genetics, Dalhousie University, Halifax, NS, Canada
- Department of Pharmacology, Dalhousie University, Halifax, NS, Canada
| | - Wanhe Li
- Laboratory of Genetics, The Rockefeller University, New York, NY, USA
| | - Deniz Top
- Department of Pediatrics, Division of Medical Genetics, Dalhousie University, Halifax, NS, Canada
- Department of Pharmacology, Dalhousie University, Halifax, NS, Canada
| |
Collapse
|
37
|
Bardeci NG, Tofolón E, Trajtenberg F, Caramelo J, Larrieux N, Rossi S, Buschiazzo A, Moreno S. The crystal structure of yeast regulatory subunit reveals key evolutionary insights into Protein Kinase A oligomerization. J Struct Biol 2021; 213:107732. [PMID: 33819633 DOI: 10.1016/j.jsb.2021.107732] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 03/15/2021] [Accepted: 03/30/2021] [Indexed: 02/08/2023]
Abstract
Protein Kinase A (PKA) is a widespread enzyme that plays a key role in many signaling pathways from lower eukaryotes to metazoans. In mammals, the regulatory (R) subunits sequester and target the catalytic (C) subunits to proper subcellular locations. This targeting is accomplished by the dimerization and docking (D/D) domain of the R subunits. The activation of the holoenzyme depends on the binding of the second messenger cAMP. The only available structures of the D/D domain proceed from mammalian sources. Unlike dimeric mammalian counterparts, the R subunit from Saccharomyces cerevisiae (Bcy1) forms tetramers in solution. Here we describe the first high-resolution structure of a non-mammalian D/D domain. The tetramer in the crystals of the Bcy1 D/D domain is a dimer of dimers that retain the classical D/D domain fold. By using phylogenetic and structural analyses combined with site-directed mutagenesis, we found that fungal R subunits present an insertion of a single amino acid at the D/D domain that shifts the position of a downstream, conserved arginine. This residue participates in intra-dimer interactions in mammalian D/D domains, while due to this insertion it is involved in inter-dimer contacts in Bcy1, which are crucial for the stability of the tetramer. This surprising finding challenges well-established concepts regarding the oligomeric state within the PKAR protein family and provides important insights into the yet unexplored structural diversity of the D/D domains and the molecular determinants of R subunit oligomerization.
Collapse
Affiliation(s)
- Nicolás González Bardeci
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires C1428EHA, Argentina; Instituto de Química Biológica, Facultad de Ciencias Exactas y Naturales (IQUIBICEN-CONICET), Buenos Aires C1428EHA, Argentina.
| | - Enzo Tofolón
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires C1428EHA, Argentina; Instituto de Química Biológica, Facultad de Ciencias Exactas y Naturales (IQUIBICEN-CONICET), Buenos Aires C1428EHA, Argentina
| | - Felipe Trajtenberg
- Laboratory of Molecular and Structural Microbiology, Institut Pasteur de Montevideo, Montevideo 11400, Uruguay
| | - Julio Caramelo
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires C1428EHA, Argentina; Fundación Instituto Leloir, Instituto de investigaciones Bioquímicas de Buenos Aires (IIBBA- CONICET), Buenos Aires C1405BWE, Argentina
| | - Nicole Larrieux
- Laboratory of Molecular and Structural Microbiology, Institut Pasteur de Montevideo, Montevideo 11400, Uruguay
| | - Silvia Rossi
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires C1428EHA, Argentina; Instituto de Química Biológica, Facultad de Ciencias Exactas y Naturales (IQUIBICEN-CONICET), Buenos Aires C1428EHA, Argentina
| | - Alejandro Buschiazzo
- Laboratory of Molecular and Structural Microbiology, Institut Pasteur de Montevideo, Montevideo 11400, Uruguay
| | - Silvia Moreno
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires C1428EHA, Argentina; Instituto de Química Biológica, Facultad de Ciencias Exactas y Naturales (IQUIBICEN-CONICET), Buenos Aires C1428EHA, Argentina.
| |
Collapse
|
38
|
D’Angelo V, Giorgi M, Paldino E, Cardarelli S, Fusco FR, Saverioni I, Sorge R, Martella G, Biagioni S, Mercuri NB, Pisani A, Sancesario G. A2A Receptor Dysregulation in Dystonia DYT1 Knock-Out Mice. Int J Mol Sci 2021; 22:2691. [PMID: 33799994 PMCID: PMC7962104 DOI: 10.3390/ijms22052691] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Accepted: 03/01/2021] [Indexed: 01/28/2023] Open
Abstract
We aimed to investigate A2A receptors in the basal ganglia of a DYT1 mouse model of dystonia. A2A was studied in control Tor1a+/+ and Tor1a+/- knock-out mice. A2A expression was assessed by anti-A2A antibody immunofluorescence and Western blotting. The co-localization of A2A was studied in striatal cholinergic interneurons identified by anti-choline-acetyltransferase (ChAT) antibody. A2A mRNA and cyclic adenosine monophosphate (cAMP) contents were also assessed. In Tor1a+/+, Western blotting detected an A2A 45 kDa band, which was stronger in the striatum and the globus pallidus than in the entopeduncular nucleus. Moreover, in Tor1a+/+, immunofluorescence showed A2A roundish aggregates, 0.3-0.4 μm in diameter, denser in the neuropil of the striatum and the globus pallidus than in the entopeduncular nucleus. In Tor1a+/-, A2A Western blotting expression and immunofluorescence aggregates appeared either increased in the striatum and the globus pallidus, or reduced in the entopeduncular nucleus. Moreover, in Tor1a+/-, A2A aggregates appeared increased in number on ChAT positive interneurons compared to Tor1a+/+. Finally, in Tor1a+/-, an increased content of cAMP signal was detected in the striatum, while significant levels of A2A mRNA were neo-expressed in the globus pallidus. In Tor1a+/-, opposite changes of A2A receptors' expression in the striatal-pallidal complex and the entopeduncular nucleus suggest that the pathophysiology of dystonia is critically dependent on a composite functional imbalance of the indirect over the direct pathway in basal ganglia.
Collapse
Affiliation(s)
- Vincenza D’Angelo
- Department of Systems Medicine, Tor Vergata University of Rome, 00133 Rome, Italy; (V.D.); (R.S.); (G.M.); (N.B.M.)
| | - Mauro Giorgi
- Department of Biology and Biotechnology “Charles Darwin”, Sapienza University of Rome, 00185 Rome, Italy; (M.G.); (S.C.); (I.S.); (S.B.)
| | - Emanuela Paldino
- IRCCS Santa Lucia Foundation, 00179 Rome, Italy; (E.P.); (F.R.F.)
| | - Silvia Cardarelli
- Department of Biology and Biotechnology “Charles Darwin”, Sapienza University of Rome, 00185 Rome, Italy; (M.G.); (S.C.); (I.S.); (S.B.)
| | | | - Ilaria Saverioni
- Department of Biology and Biotechnology “Charles Darwin”, Sapienza University of Rome, 00185 Rome, Italy; (M.G.); (S.C.); (I.S.); (S.B.)
| | - Roberto Sorge
- Department of Systems Medicine, Tor Vergata University of Rome, 00133 Rome, Italy; (V.D.); (R.S.); (G.M.); (N.B.M.)
| | - Giuseppina Martella
- Department of Systems Medicine, Tor Vergata University of Rome, 00133 Rome, Italy; (V.D.); (R.S.); (G.M.); (N.B.M.)
- IRCCS Santa Lucia Foundation, 00179 Rome, Italy; (E.P.); (F.R.F.)
| | - Stefano Biagioni
- Department of Biology and Biotechnology “Charles Darwin”, Sapienza University of Rome, 00185 Rome, Italy; (M.G.); (S.C.); (I.S.); (S.B.)
| | - Nicola B. Mercuri
- Department of Systems Medicine, Tor Vergata University of Rome, 00133 Rome, Italy; (V.D.); (R.S.); (G.M.); (N.B.M.)
| | - Antonio Pisani
- IRCCS Mondino Foundation, 27100 Pavia, Italy;
- Department of Brain and Behavioral Sciences, University of Pavia, 27100 Pavia, Italy
| | - Giuseppe Sancesario
- Department of Systems Medicine, Tor Vergata University of Rome, 00133 Rome, Italy; (V.D.); (R.S.); (G.M.); (N.B.M.)
| |
Collapse
|
39
|
Zhang J, Zhang C, Chen X, Wang B, Ma W, Yang Y, Zheng R, Huang Z. PKA-RIIβ autophosphorylation modulates PKA activity and seizure phenotypes in mice. Commun Biol 2021; 4:263. [PMID: 33649504 PMCID: PMC7921646 DOI: 10.1038/s42003-021-01748-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 12/22/2020] [Indexed: 11/20/2022] Open
Abstract
Temporal lobe epilepsy (TLE) is one of the most common and intractable neurological disorders in adults. Dysfunctional PKA signaling is causally linked to the TLE. However, the mechanism underlying PKA involves in epileptogenesis is still poorly understood. In the present study, we found the autophosphorylation level at serine 114 site (serine 112 site in mice) of PKA-RIIβ subunit was robustly decreased in the epileptic foci obtained from both surgical specimens of TLE patients and seizure model mice. The p-RIIβ level was negatively correlated with the activities of PKA. Notably, by using a P-site mutant that cannot be autophosphorylated and thus results in the released catalytic subunit to exert persistent phosphorylation, an increase in PKA activities through transduction with AAV-RIIβ-S112A in hippocampal DG granule cells decreased mIPSC frequency but not mEPSC, enhanced neuronal intrinsic excitability and seizure susceptibility. In contrast, a reduction of PKA activities by RIIβ knockout led to an increased mIPSC frequency, a reduction in neuronal excitability, and mice less prone to experimental seizure onset. Collectively, our data demonstrated that the autophosphorylation of RIIβ subunit plays a critical role in controlling neuronal and network excitabilities by regulating the activities of PKA, providing a potential therapeutic target for TLE.
Collapse
Affiliation(s)
- Jingliang Zhang
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences, Peking University Health Science Center, Beijing, China
| | - Chenyu Zhang
- Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Xiaoling Chen
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences, Peking University Health Science Center, Beijing, China
| | - Bingwei Wang
- Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Weining Ma
- Department of Neurology, Shengjing Hospital Affiliated to China Medical University, Shenyang, China
| | - Yang Yang
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University College of Pharmacy, West Lafayette, IN, USA
| | - Ruimao Zheng
- Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China.
- Neuroscience Research Institute, Peking University, Beijing, China.
- Key Laboratory for Neuroscience, Ministry of Education, Beijing, China.
- Key Laboratory for Neuroscience of National Health Commission, Beijing, China.
| | - Zhuo Huang
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences, Peking University Health Science Center, Beijing, China.
- Key Laboratory for Neuroscience, Ministry of Education, Beijing, China.
- Key Laboratory for Neuroscience of National Health Commission, Beijing, China.
| |
Collapse
|
40
|
Serotonin receptor 5-HT7 in Drosophila mushroom body neurons mediates larval appetitive olfactory learning. Sci Rep 2020; 10:21267. [PMID: 33277559 PMCID: PMC7718245 DOI: 10.1038/s41598-020-77910-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 11/09/2020] [Indexed: 11/29/2022] Open
Abstract
Serotonin (5-HT) and dopamine are critical neuromodulators known to regulate a range of behaviors in invertebrates and mammals, such as learning and memory. Effects of both serotonin and dopamine are mediated largely through their downstream G-protein coupled receptors through cAMP-PKA signaling. While the role of dopamine in olfactory learning in Drosophila is well described, the function of serotonin and its downstream receptors on Drosophila olfactory learning remain largely unexplored. In this study we show that the output of serotonergic neurons, possibly through points of synaptic contacts on the mushroom body (MB), is essential for training during olfactory associative learning in Drosophila larvae. Additionally, we demonstrate that the regulation of olfactory associative learning by serotonin is mediated by its downstream receptor (d5-HT7) in a cAMP-dependent manner. We show that d5-HT7 expression specifically in the MB, an anatomical structure essential for olfactory learning in Drosophila, is critical for olfactory associative learning. Importantly our work shows that spatio-temporal restriction of d5-HT7 expression to the MB is sufficient to rescue olfactory learning deficits in a d5-HT7 null larvae. In summary, our results establish a critical, and previously unknown, role of d5-HT7 in olfactory learning.
Collapse
|
41
|
Pharmacological inhibition of phosphodiesterase 7 enhances consolidation processes of spatial memory. Neurobiol Learn Mem 2020; 177:107357. [PMID: 33278592 DOI: 10.1016/j.nlm.2020.107357] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 11/23/2020] [Accepted: 11/30/2020] [Indexed: 12/26/2022]
Abstract
Augmentation of cAMP signaling through inhibition of phosphodiesterases (PDE) is known to enhance plasticity and memory. Inhibition of PDE4 enhances consolidation into memory, but less is known about the role of other cAMP specific PDEs. Here, we tested the effects of oral treatment with a selective inhibitor of PDE7 of nanomolar potency on spatial and contextual memory. In an object location task, doses of 0.3-3 mg/kg administered 3 h after training dose-dependently attenuated time-dependent forgetting in rats. Significant enhancement of memory occurred at a dose of 3 mg/kg with corresponding brain levels consistent with PDE7 inhibition. The same dose given prior to training augmented contextual fear conditioning. In mice, daily dosing before training enhanced spatial memory in two different incremental learning paradigms in the Barnes Maze. Drug treated mice made significantly less errors locating the escape in a probe-test 24 h after the end of training, and they exhibited hippocampal-dependent spatial search strategies more frequently than controls, which tended to show serial sampling of escape locations. Acquisition and short-term memory, in contrast, were unaffected. Our data provide evidence for a role of PDE7 in the consolidation of hippocampal-dependent memory. We suggest that targeting PDE7 for memory enhancement may provide an alternative to PDE4 inhibitors, which tend to have undesirable gastrointestinal side-effects.
Collapse
|
42
|
Wang Y, Kang Y, Qi C, Zhang T, Zhao H, Ji X, Yan W, Huang Y, Cui R, Zhang G, Shi G. Pentoxifylline enhances antioxidative capability and promotes mitochondrial biogenesis for improving age-related behavioral deficits. Aging (Albany NY) 2020; 12:25487-25504. [PMID: 33231568 PMCID: PMC7803534 DOI: 10.18632/aging.104155] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 09/09/2020] [Indexed: 12/15/2022]
Abstract
Pentoxifylline (PTX) is a non-specific phosphodiesterase inhibitor with pleiotropic effects that is routinely used to treat peripheral vascular disease. In this study, we tested whether PTX could also counteract the detrimental effects of aging in the brain. To accomplish that, we treated aged rats with PTX and measured resulting behavioral alterations as well as changes in dopaminergic neurochemical levels, oxidative balance markers, mitochondrial function, nuclear factor erythroid 2-related factor 2 (Nrf2), peroxisome proliferator activated receptor-gamma coactivator 1-alpha (PGC-1α) and downstream gene expression, and cyclic adenosine monophosphate (cAMP) content in the brain. The results demonstrated that PTX improved motor and cognitive deficits and restored levels of dopamine and its metabolites in the brains of aged rats. PTX also reduced malondialdehyde levels and increased the GSH/GSSG ratio, mitochondrial ATP, nuclear Nrf2, and cAMP levels, and upregulated PGC-1α, nuclear respiratory factor 1, and mitochondrial transcription factor A expression in the substantia nigra and hippocampus of aged rats. Thus, increased nuclear Nrf2 levels and upregulation of PGC-1α, which enhance antioxidative capability and promote mitochondrial biogenesis, may be responsible for PTX-induced amelioration of behavioral deficits in aged rats.
Collapse
Affiliation(s)
- Yu Wang
- Department of Neurobiology, Hebei Medical University, Shijiazhuang 050017, China
| | - Yunxiao Kang
- Department of Neurobiology, Hebei Medical University, Shijiazhuang 050017, China
| | - Chunxiao Qi
- Department of Anatomy, Hebei Medical University, Shijiazhuang 050017, China
| | - Tianyun Zhang
- Department of Neurobiology, Hebei Medical University, Shijiazhuang 050017, China
| | - Hui Zhao
- Department of Neurobiology, Hebei Medical University, Shijiazhuang 050017, China
| | - Xiaoming Ji
- Department of Neurobiology, Hebei Medical University, Shijiazhuang 050017, China
| | - Wensheng Yan
- Department of Sports Medicine, Hebei Sport University, Shijiazhuang 050017, China
| | - Yuanxiang Huang
- Department of Neurobiology, Hebei Medical University, Shijiazhuang 050017, China
| | - Rui Cui
- Department of Anatomy, Hebei Medical University, Shijiazhuang 050017, China
| | - Guoliang Zhang
- Department of Neurobiology, Hebei Medical University, Shijiazhuang 050017, China.,Department of Anatomy, Hebei Medical University, Shijiazhuang 050017, China
| | - Geming Shi
- Department of Neurobiology, Hebei Medical University, Shijiazhuang 050017, China.,Neuroscience Research Center, Hebei Medical University, Shijiazhuang 050017, China.,Hebei Key Laboratory of Neurodegenerative Disease Mechanism, Hebei Medical University, Shijiazhuang 050017, China
| |
Collapse
|
43
|
Sears JC, Broadie K. FMRP-PKA Activity Negative Feedback Regulates RNA Binding-Dependent Fibrillation in Brain Learning and Memory Circuitry. Cell Rep 2020; 33:108266. [PMID: 33053340 PMCID: PMC7590955 DOI: 10.1016/j.celrep.2020.108266] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 09/07/2020] [Accepted: 09/22/2020] [Indexed: 11/29/2022] Open
Abstract
Fragile X mental retardation protein (FMRP) promotes cyclic AMP (cAMP) signaling. Using an in vivo protein kinase A activity sensor (PKA-SPARK), we find that Drosophila FMRP (dFMRP) and human FMRP (hFMRP) enhance PKA activity in a central brain learning and memory center. Increasing neuronal PKA activity suppresses FMRP in Kenyon cells, demonstrating an FMRP-PKA negative feedback loop. A patient-derived R140Q FMRP point mutation mislocalizes PKA-SPARK activity, whereas deletion of the RNA-binding argi-nine-glycine-glycine (RGG) box (hFMRP-ΔRGG) produces fibrillar PKA-SPARK assemblies colocalizing with ribonucleoprotein (RNP) and aggregation (thioflavin T) markers, demonstrating fibrillar partitioning of cytosolic protein aggregates. hFMRP-ΔRGG reduces dFMRP levels, indicating RGG-independent regulation. Short-term hFMRP-ΔRGG induction produces activated PKA-SPARK puncta, whereas long induction drives fibrillar assembly. Elevated temperature disassociates hFMRP-ΔRGG aggregates and blocks activated PKA-SPARK localization. These results suggest that FMRP regulates compartmentalized signaling via complex assembly, directing PKA activity localization, with FMRP RGG box RNA binding restricting separation via low-complexity interactions. FMRP is required for brain cAMP induction and cAMP-dependent PKA activation, but the FMRP mechanism is uncharacterized. Sears and Broadie test FXS patient-derived and FMRP domain-deficient mutants to reveal conserved FMRP functions regulating PKA activation, subcellular localization, and reversible partitioning into elongated fibrillar assemblies in brain learning/ memory circuit neurons.
Collapse
Affiliation(s)
- James C Sears
- Vanderbilt Brain Institute, Vanderbilt University Medical Center, Nashville, TN 37235, USA; Department of Biological Sciences, Vanderbilt University, Nashville, TN 37235, USA.
| | - Kendal Broadie
- Vanderbilt Brain Institute, Vanderbilt University Medical Center, Nashville, TN 37235, USA; Department of Biological Sciences, Vanderbilt University, Nashville, TN 37235, USA; Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37235, USA; Department of Cell and Developmental Biology, Vanderbilt University Medical Center, Nashville, TN 37235, USA.
| |
Collapse
|
44
|
Blosser JA, Podolsky E, Lee D. L-DOPA-Induced Dyskinesia in a Genetic Drosophila Model of Parkinson's Disease. Exp Neurobiol 2020; 29:273-284. [PMID: 32921640 PMCID: PMC7492844 DOI: 10.5607/en20028] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Revised: 08/27/2020] [Accepted: 08/31/2020] [Indexed: 01/05/2023] Open
Abstract
Motor symptoms in Parkinson’s disease (PD) are directly related to the reduction of a neurotransmitter dopamine. Therefore, its precursor L-DOPA became the gold standard for PD treatment. However, chronic use of L-DOPA causes uncontrollable, involuntary movements, called L-DOPA-induced dyskinesia (LID) in the majority of PD patients. LID is complicated and very difficult to manage. Current rodent and non-human primate models have been developed to study LID mainly using neurotoxins. Therefore, it is necessary to develop a LID animal model with defects in genetic factors causing PD in order to study the relation between LID and PD genes such as α-synuclein. In this study, we first showed that a low concentration of L-DOPA (100 µM) rescues locomotion defects (i.e., speed, angular velocity, pause time) in Drosophila larvae expressing human mutant α-synuclein (A53T). This A53T larval model of PD was used to further examine dyskinetic behaviors. High concentrations of L-DOPA (5 or 10 mM) causes hyperactivity such as body bending behavior (BBB) in A53T larva, which resembles axial dyskinesia in rodents. Using ImageJ plugins and other third party software, dyskinetic BBB has been accurately and efficiently quantified. Further, we showed that a dopamine agonist pramipexole (PRX) partially rescues BBB caused by high L-DOPA. Our Drosophila genetic LID model will provide an important experimental platform to examine molecular and cellular mechanisms underlying LID, to study the role of PD causing genes in the development of LID, and to identify potential targets to slow/reverse LID pathology.
Collapse
Affiliation(s)
- Joshua A Blosser
- Neuroscience Program, Department of Biological Sciences, Ohio University, Athens, OH 45701, USA
| | - Eric Podolsky
- Neuroscience Program, Department of Biological Sciences, Ohio University, Athens, OH 45701, USA
| | - Daewoo Lee
- Neuroscience Program, Department of Biological Sciences, Ohio University, Athens, OH 45701, USA
| |
Collapse
|
45
|
Zhu C, Hui L, Zheng K, Liu L, Liu J, Lv W. Silencing of RGS2 enhances hippocampal neuron regeneration and rescues depression-like behavioral impairments through activation of cAMP pathway. Brain Res 2020; 1746:147018. [PMID: 32679115 DOI: 10.1016/j.brainres.2020.147018] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 07/01/2020] [Accepted: 07/10/2020] [Indexed: 12/31/2022]
Abstract
Depression is one of the most common mental disorders with an increasing incidence. However, factors involved in depression are so complex, thus it is difficult to find effective strategies to reverse the impairments. This study aims to verify the role of regulator of G protein signaling 2 (RGS2) in the mouse mode of unpredictable mild stress-induced depression-like behaviors. Knockdown of RGS2 was achieved by transfection of siRNA-RGS2 in mouse hippocampal (HT-22) cells in vitro and injection of recombinant adenovirus expressing siRNA-RGS2 in mice in vivo. An aberrant high expression of RGS2 was found in mice with depression-like behaviors through immunohistochemical analysis. Silencing of RGS2 or Forskolin (activator of cAMP pathway) developed sweet water consumption, reduced inflammation and oxidative stress injury, and attenuated cognitive impairment and neuronal damage in mice with depression-like behaviors. Furthermore, regeneration was enhanced and apoptosis was repressed in mouse hippocampal neurons in the presence of RGS2 knockdown and Forskolin. Mechanistic studies indicated that silencing of RGS2 promoted the activation of cAMP pathway, thus rescuing depression-like behaviors of mice. Collectively, our study uncovered the role of RGS2-dependent cAMP pathway in regulation of cognitive impairment and hippocampal neuron regeneration in depression-like behaviors of mice, which may be a potential therapeutic target for impairments and symptoms associated with depression.
Collapse
Affiliation(s)
- Cheng Zhu
- Department Clinical Psychology, The Affiliated Kangning Hospital to Wenzhou Medical University, Wenzhou 325000, PR China.
| | - Li Hui
- Institute of Mental Health, Suzhou Guangji Hospital, The Affiliated Guangji Hospital of Soochow University, Soochow University, Suzhou, 215137, PR China
| | - Ke Zheng
- Department of Psychiatry, The Affiliated Kangning Hospital to Wenzhou Medical University, Wenzhou 325000, PR China
| | - Linjing Liu
- Department Clinical Psychology, The Affiliated Kangning Hospital to Wenzhou Medical University, Wenzhou 325000, PR China
| | - Jiahong Liu
- Department Clinical Psychology, The Affiliated Kangning Hospital to Wenzhou Medical University, Wenzhou 325000, PR China
| | - Wei Lv
- Department of Psychiatry, The Affiliated Kangning Hospital to Wenzhou Medical University, Wenzhou 325000, PR China
| |
Collapse
|
46
|
Phosphodiesterase 10A Inhibition Leads to Brain Region-Specific Recovery Based on Stroke Type. Transl Stroke Res 2020; 12:303-315. [PMID: 32378029 PMCID: PMC7644574 DOI: 10.1007/s12975-020-00819-8] [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: 02/26/2020] [Revised: 04/19/2020] [Accepted: 04/22/2020] [Indexed: 01/08/2023]
Abstract
Stroke is the leading cause of adult disability. Recovery of function after stroke involves signaling events that are mediated by cAMP and cGMP pathways, such as axonal sprouting, neurogenesis, and synaptic plasticity. cAMP and cGMP are degraded by phosphodiesterases (PDEs), which are differentially expressed in brain regions. PDE10A is highly expressed in the basal ganglia/striatum. We tested a novel PDE10A inhibitor (TAK-063) for its effects on functional recovery. Stroke was produced in mice in the cortex or the striatum. Behavioral recovery was measured to 9 weeks. Tissue outcome measures included analysis of growth factor levels, angiogenesis, neurogenesis, gliogenesis, and inflammation. TAK-063 improved motor recovery after striatal stroke in a dose-related manner, but not in cortical stroke. Recovery of motor function correlated with increases in striatal brain-derived neurotrophic factor. TAK-063 treatment also increased motor system axonal connections. Stroke affects distinct brain regions, with each comprising different cellular and molecular elements. Inhibition of PDE10A improved recovery of function after striatal but not cortical stroke, consistent with its brain localization. This experiment is the first demonstration of brain region-specific enhanced functional recovery after stroke, and indicates that differential molecular signaling between brain regions can be exploited to improve recovery based on stroke subtype.
Collapse
|
47
|
Liu X, Zhou Y, Yang D, Li S, Liu X, Wang Z. Type 3 adenylyl cyclase in the MOE is involved in learning and memory in mice. Behav Brain Res 2020; 383:112533. [DOI: 10.1016/j.bbr.2020.112533] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Revised: 01/27/2020] [Accepted: 02/01/2020] [Indexed: 02/06/2023]
|
48
|
Malpe MS, McSwain LF, Kudyba K, Ng CL, Nicholson J, Brady M, Qian Y, Choksi V, Hudson AG, Parrott BB, Schulz C. G-protein signaling is required for increasing germline stem cell division frequency in response to mating in Drosophila males. Sci Rep 2020; 10:3888. [PMID: 32127590 PMCID: PMC7054589 DOI: 10.1038/s41598-020-60807-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Accepted: 02/17/2020] [Indexed: 02/07/2023] Open
Abstract
Adult stem cells divide to renew the stem cell pool and replenish specialized cells that are lost due to death or usage. However, little is known about the mechanisms regulating how stem cells adjust to a demand for specialized cells. A failure of the stem cells to respond to this demand can have serious consequences, such as tissue loss, or prolonged recovery post injury. Here, we challenge the male germline stem cells (GSCs) of Drosophila melanogaster for the production of specialized cells, sperm cells, using mating experiments. We show that repeated mating reduced the sperm pool and increased the percentage of GSCs in M- and S-phase of the cell cycle. The increase in dividing GSCs depended on the activity of the highly conserved G-proteins. Germline expression of RNA-Interference (RNA-i) constructs against G-proteins, or a dominant negative G-protein eliminated the increase in GSC division frequency in mated males. Consistent with a role for the G-proteins in regulating GSC division frequency, RNA-i against seven out of 35 G-protein coupled receptors (GPCRs) within the germline cells also eliminated the capability of males to increase the numbers of dividing GSCs in response to mating.
Collapse
Affiliation(s)
- Manashree S Malpe
- Department of Cellular Biology, University of Georgia, Athens, GA, 30602, USA
| | - Leon F McSwain
- Winship Cancer Institute, Emory University, Atlanta, GA, 30322, USA
| | - Karl Kudyba
- Department of Cellular Biology, University of Georgia, Athens, GA, 30602, USA
| | - Chun L Ng
- University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Jennie Nicholson
- Department of Cellular Biology, University of Georgia, Athens, GA, 30602, USA
| | - Maximilian Brady
- Department of Cellular Biology, University of Georgia, Athens, GA, 30602, USA
| | - Yue Qian
- University of North Georgia, Department of Biology, Oakwood, GA, 30566, USA
| | - Vinay Choksi
- School of Medicine, Duke University, Durham, NC, 27708, USA
| | - Alicia G Hudson
- Department of Cellular Biology, University of Georgia, Athens, GA, 30602, USA
| | | | - Cordula Schulz
- Department of Cellular Biology, University of Georgia, Athens, GA, 30602, USA.
| |
Collapse
|
49
|
Ohadi D, Rangamani P. Geometric Control of Frequency Modulation of cAMP Oscillations due to Calcium in Dendritic Spines. Biophys J 2019; 117:1981-1994. [PMID: 31668747 PMCID: PMC7018999 DOI: 10.1016/j.bpj.2019.10.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 07/30/2019] [Accepted: 10/02/2019] [Indexed: 12/12/2022] Open
Abstract
The spatiotemporal regulation of cyclic adenosine monophosphate (cAMP) and its dynamic interactions with other second messengers such as calcium are critical features of signaling specificity required for neuronal development and connectivity. cAMP is known to contribute to long-term potentiation and memory formation by controlling the formation and regulation of dendritic spines. Despite the recent advances in biosensing techniques for monitoring spatiotemporal cAMP dynamics, the underlying molecular mechanisms that attribute to the subcellular modulation of cAMP remain unknown. In this work, we model the spatiotemporal dynamics of calcium-induced cAMP signaling pathway in dendritic spines. Using a three-dimensional reaction-diffusion model, we investigate the effect of different spatial characteristics of cAMP dynamics that may be responsible for subcellular regulation of cAMP concentrations. Our model predicts that the volume/surface ratio of the spine, regulated through the spine head size, spine neck size, and the presence of physical barriers (spine apparatus), is an important regulator of cAMP dynamics. Furthermore, localization of the enzymes responsible for the synthesis and degradation of cAMP in different compartments also modulates the oscillatory patterns of cAMP through exponential relationships. Our findings shed light on the significance of complex geometric and localization relationships for cAMP dynamics in dendritic spines.
Collapse
Affiliation(s)
- Donya Ohadi
- Department of Mechanical and Aerospace Engineering, University of California, San Diego, La Jolla, California
| | - Padmini Rangamani
- Department of Mechanical and Aerospace Engineering, University of California, San Diego, La Jolla, California.
| |
Collapse
|
50
|
Sub-Chronic Consumption of Dark Chocolate Enhances Cognitive Function and Releases Nerve Growth Factors: A Parallel-Group Randomized Trial. Nutrients 2019; 11:nu11112800. [PMID: 31744119 PMCID: PMC6893800 DOI: 10.3390/nu11112800] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 11/12/2019] [Accepted: 11/14/2019] [Indexed: 01/18/2023] Open
Abstract
Previous research has shown that habitual chocolate intake is related to cognitive performance and that frequent chocolate consumption is significantly associated with improved memory. However, little is known about the effects of the subchronic consumption of dark chocolate (DC) on cognitive function and neurotrophins. Eighteen healthy young subjects (both sexes; 20-31 years old) were randomly divided into two groups: a DC intake group (n = 10) and a cacao-free white chocolate (WC) intake group (n = 8). The subjects then consumed chocolate daily for 30 days. Blood samples were taken to measure plasma levels of theobromine (a methylxanthine most often present in DC), nerve growth factor (NGF), and brain-derived neurotrophic factor, and to analyze hemodynamic parameters. Cognitive function was assessed using a modified Stroop color word test and digital cancellation test. Prefrontal cerebral blood flow was measured during the tests. DC consumption increased the NGF and theobromine levels in plasma, enhancing cognitive function performance in both tests. Interestingly, the DC-mediated enhancement of cognitive function was observed three weeks after the end of chocolate intake. WC consumption did not affect NGF and theobromine levels or cognitive performance. These results suggest that DC consumption has beneficial effects on human health by enhancing cognitive function.
Collapse
|