1
|
Mandel S, Hanke T, Mathea S, Chatterjee D, Saraswati H, Berger BT, Schwalm MP, Yamamoto S, Tawada M, Takagi T, Ahmed M, Röhm S, Corrionero A, Alfonso P, Baena M, Elson L, Menge A, Krämer A, Pereira R, Müller S, Krause DS, Knapp S. Repurposing of the RIPK1-Selective Benzo[1,4]oxazepin-4-one Scaffold for the Development of a Type III LIMK1/2 Inhibitor. ACS Chem Biol 2025; 20:1087-1098. [PMID: 40227881 PMCID: PMC12090182 DOI: 10.1021/acschembio.5c00097] [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: 02/04/2025] [Revised: 03/25/2025] [Accepted: 03/31/2025] [Indexed: 04/16/2025]
Abstract
Benzoxazepinones have been extensively studied as exclusively selective RIP kinase 1 inhibitors. This scaffold binds to an allosteric pocket created by an αC-out/DFG-out conformation. This inactive conformation results in a large expansion of the kinase back pocket, a conformation that has also been reported for LIM kinases. Scaffold hopping is common in the design of orthosteric kinase inhibitors but has not been explored in the design of allosteric inhibitors, mainly due to the typically exclusive selectivity of type III inhibitors. Here, we hypothesized that the shared structural properties of LIMKs and RIPKs could lead to novel type III LIMK inhibitors using the benzoxazepinone scaffold. We report the discovery of a novel LIMK1/2 inhibitor that relies on this scaffold-based approach. The discovered compound 10 showed low nanomolar potency on LIMK1/2 and exceptional selectivity, as confirmed by a comprehensive selectivity panel with residual RIPK activity as the only off-target. The study provides one of the few examples for scaffold hopping for allosteric inhibitors, which are usually associated with exclusive target selectivity.
Collapse
Affiliation(s)
- Sebastian Mandel
- Institute
for Pharmaceutical Chemistry, Johann Wolfgang
Goethe University, Max von Laue Str. 9, 60438 Frankfurt am Main, Germany
- Structural
Genomics Consortium, Buchmann Institute for Molecular Life Sciences, Johann Wolfgang Goethe University, Max von Laue Str. 15, 60438 Frankfurt am Main, Germany
| | - Thomas Hanke
- Institute
for Pharmaceutical Chemistry, Johann Wolfgang
Goethe University, Max von Laue Str. 9, 60438 Frankfurt am Main, Germany
- Structural
Genomics Consortium, Buchmann Institute for Molecular Life Sciences, Johann Wolfgang Goethe University, Max von Laue Str. 15, 60438 Frankfurt am Main, Germany
| | - Sebastian Mathea
- Institute
for Pharmaceutical Chemistry, Johann Wolfgang
Goethe University, Max von Laue Str. 9, 60438 Frankfurt am Main, Germany
- Structural
Genomics Consortium, Buchmann Institute for Molecular Life Sciences, Johann Wolfgang Goethe University, Max von Laue Str. 15, 60438 Frankfurt am Main, Germany
| | - Deep Chatterjee
- Institute
for Pharmaceutical Chemistry, Johann Wolfgang
Goethe University, Max von Laue Str. 9, 60438 Frankfurt am Main, Germany
- Structural
Genomics Consortium, Buchmann Institute for Molecular Life Sciences, Johann Wolfgang Goethe University, Max von Laue Str. 15, 60438 Frankfurt am Main, Germany
| | - Hayuningbudi Saraswati
- Institute
of Transfusion Medicine−Transfusion Centre, Johannes Gutenberg University Medical Center, 55131 Mainz, Germany
| | - Benedict-Tilman Berger
- Institute
for Pharmaceutical Chemistry, Johann Wolfgang
Goethe University, Max von Laue Str. 9, 60438 Frankfurt am Main, Germany
- Structural
Genomics Consortium, Buchmann Institute for Molecular Life Sciences, Johann Wolfgang Goethe University, Max von Laue Str. 15, 60438 Frankfurt am Main, Germany
| | - Martin Peter Schwalm
- Institute
for Pharmaceutical Chemistry, Johann Wolfgang
Goethe University, Max von Laue Str. 9, 60438 Frankfurt am Main, Germany
- Structural
Genomics Consortium, Buchmann Institute for Molecular Life Sciences, Johann Wolfgang Goethe University, Max von Laue Str. 15, 60438 Frankfurt am Main, Germany
- German Cancer
Consortium (DKTK), German Cancer Research
Center (DKFZ), DKTK site Frankfurt-Mainz, 69120 Heidelberg, Germany
| | - Satoshi Yamamoto
- Neuroscience
Drug Discovery Unit, Research, Takeda Pharmaceutical
Company Limited, 26-1 Muraoka-Higashi 2-chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Michiko Tawada
- Neuroscience
Drug Discovery Unit, Research, Takeda Pharmaceutical
Company Limited, 26-1 Muraoka-Higashi 2-chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Terufumi Takagi
- Neuroscience
Drug Discovery Unit, Research, Takeda Pharmaceutical
Company Limited, 26-1 Muraoka-Higashi 2-chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Mahmood Ahmed
- Inaver Pharma
Consulting, 2 Havelock
Road, #07-12 Havelock 2, 059763 Singapore
| | - Sandra Röhm
- Institute
for Pharmaceutical Chemistry, Johann Wolfgang
Goethe University, Max von Laue Str. 9, 60438 Frankfurt am Main, Germany
- Structural
Genomics Consortium, Buchmann Institute for Molecular Life Sciences, Johann Wolfgang Goethe University, Max von Laue Str. 15, 60438 Frankfurt am Main, Germany
| | - Ana Corrionero
- Enzymlogic, Qube Technology Park, C/Santiago
Grisolía, 2, 28760 Madrid, Spain
| | - Patricia Alfonso
- Enzymlogic, Qube Technology Park, C/Santiago
Grisolía, 2, 28760 Madrid, Spain
| | - Maria Baena
- Enzymlogic, Qube Technology Park, C/Santiago
Grisolía, 2, 28760 Madrid, Spain
| | - Lewis Elson
- Institute
for Pharmaceutical Chemistry, Johann Wolfgang
Goethe University, Max von Laue Str. 9, 60438 Frankfurt am Main, Germany
- Structural
Genomics Consortium, Buchmann Institute for Molecular Life Sciences, Johann Wolfgang Goethe University, Max von Laue Str. 15, 60438 Frankfurt am Main, Germany
| | - Amelie Menge
- Institute
for Pharmaceutical Chemistry, Johann Wolfgang
Goethe University, Max von Laue Str. 9, 60438 Frankfurt am Main, Germany
- Structural
Genomics Consortium, Buchmann Institute for Molecular Life Sciences, Johann Wolfgang Goethe University, Max von Laue Str. 15, 60438 Frankfurt am Main, Germany
| | - Andreas Krämer
- Institute
for Pharmaceutical Chemistry, Johann Wolfgang
Goethe University, Max von Laue Str. 9, 60438 Frankfurt am Main, Germany
- Structural
Genomics Consortium, Buchmann Institute for Molecular Life Sciences, Johann Wolfgang Goethe University, Max von Laue Str. 15, 60438 Frankfurt am Main, Germany
- German Cancer
Consortium (DKTK), German Cancer Research
Center (DKFZ), DKTK site Frankfurt-Mainz, 69120 Heidelberg, Germany
| | - Raquel Pereira
- Institute
for Experimental Pediatric Hematology and Oncology, Goethe University Frankfurt, 60438 Frankfurt am Main, Germany
| | - Susanne Müller
- Institute
for Pharmaceutical Chemistry, Johann Wolfgang
Goethe University, Max von Laue Str. 9, 60438 Frankfurt am Main, Germany
- Structural
Genomics Consortium, Buchmann Institute for Molecular Life Sciences, Johann Wolfgang Goethe University, Max von Laue Str. 15, 60438 Frankfurt am Main, Germany
| | - Daniela S. Krause
- Institute
of Transfusion Medicine−Transfusion Centre, Johannes Gutenberg University Medical Center, 55131 Mainz, Germany
- German Cancer
Consortium (DKTK), German Cancer Research
Center (DKFZ), DKTK site Frankfurt-Mainz, 69120 Heidelberg, Germany
- Research
Center for Immunotherapy (FZI), University
Medical Center, University of Mainz, 55131 Mainz, Germany
| | - Stefan Knapp
- Institute
for Pharmaceutical Chemistry, Johann Wolfgang
Goethe University, Max von Laue Str. 9, 60438 Frankfurt am Main, Germany
- Structural
Genomics Consortium, Buchmann Institute for Molecular Life Sciences, Johann Wolfgang Goethe University, Max von Laue Str. 15, 60438 Frankfurt am Main, Germany
- German Cancer
Consortium (DKTK), German Cancer Research
Center (DKFZ), DKTK site Frankfurt-Mainz, 69120 Heidelberg, Germany
| |
Collapse
|
2
|
Baldwin AG, Foley DW, Collins R, Lee H, Jones DH, Wahab B, Waters L, Pedder J, Paine M, Feng GJ, Privitera L, Ashall-Kelly A, Thomas C, Gillespie JA, Schino L, Belelli D, Rocha C, Maussion G, Krahn AI, Durcan TM, Elkins JM, Lambert JJ, Atack JR, Ward SE. Discovery of MDI-114215: A Potent and Selective LIMK Inhibitor To Treat Fragile X Syndrome. J Med Chem 2025; 68:719-752. [PMID: 39711116 PMCID: PMC11726654 DOI: 10.1021/acs.jmedchem.4c02694] [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: 11/04/2024] [Revised: 11/29/2024] [Accepted: 12/04/2024] [Indexed: 12/24/2024]
Abstract
LIMKs are serine/threonine and tyrosine kinases responsible for controlling cytoskeletal dynamics as key regulators of actin stability, ensuring synaptic health through normal synaptic bouton structure and function. However, LIMK1 overactivation results in abnormal dendritic synaptic development that characterizes the pathogenesis of Fragile X Syndrome (FXS). As a result, the development of LIMK inhibitors represents an emerging disease-modifying therapeutic approach for FXS. We report the discovery of MDI-114215 (85), a novel, potent allosteric dual-LIMK1/2 inhibitor that demonstrates exquisite kinome selectivity. 85 reduces phospho-cofilin in mouse brain slices and rescues impaired hippocampal long-term potentiation in brain slices from FXS mice. We also show that LIMK inhibitors are effective in reducing phospho-cofilin levels in iPSC neurons derived from FXS patients, demonstrating 85 to be a potential therapeutic candidate for FXS that could have broad application to neurological disorders or cancers caused by LIMK1/2 overactivation and actin instability.
Collapse
Affiliation(s)
- Alex G. Baldwin
- Medicines
Discovery Institute, School of Biosciences, Cardiff University, Main Building, Park Place, Cardiff CF10 3AT, U.K.
| | - David W. Foley
- Medicines
Discovery Institute, School of Biosciences, Cardiff University, Main Building, Park Place, Cardiff CF10 3AT, U.K.
| | - Ross Collins
- Medicines
Discovery Institute, School of Biosciences, Cardiff University, Main Building, Park Place, Cardiff CF10 3AT, U.K.
| | - Hyunah Lee
- Centre
for Medicines Discovery, University of Oxford, Roosevelt Drive, Oxford OX3 7DQ, U.K.
| | - D. Heulyn Jones
- Medicines
Discovery Institute, School of Biosciences, Cardiff University, Main Building, Park Place, Cardiff CF10 3AT, U.K.
| | - Ben Wahab
- Medicines
Discovery Institute, School of Biosciences, Cardiff University, Main Building, Park Place, Cardiff CF10 3AT, U.K.
| | - Loren Waters
- Medicines
Discovery Institute, School of Biosciences, Cardiff University, Main Building, Park Place, Cardiff CF10 3AT, U.K.
| | - Josephine Pedder
- Medicines
Discovery Institute, School of Biosciences, Cardiff University, Main Building, Park Place, Cardiff CF10 3AT, U.K.
| | - Marie Paine
- Medicines
Discovery Institute, School of Biosciences, Cardiff University, Main Building, Park Place, Cardiff CF10 3AT, U.K.
| | - Gui Jie Feng
- Medicines
Discovery Institute, School of Biosciences, Cardiff University, Main Building, Park Place, Cardiff CF10 3AT, U.K.
| | - Lucia Privitera
- Division
of Neuroscience, School of Medicine, Medical Sciences Institute, Dundee University, Dow Street, Dundee DD1
5HL, U.K.
| | - Alexander Ashall-Kelly
- Medicines
Discovery Institute, School of Biosciences, Cardiff University, Main Building, Park Place, Cardiff CF10 3AT, U.K.
| | - Carys Thomas
- Medicines
Discovery Institute, School of Biosciences, Cardiff University, Main Building, Park Place, Cardiff CF10 3AT, U.K.
| | - Jason A. Gillespie
- Medicines
Discovery Institute, School of Biosciences, Cardiff University, Main Building, Park Place, Cardiff CF10 3AT, U.K.
| | - Lauramariú Schino
- Medicines
Discovery Institute, School of Biosciences, Cardiff University, Main Building, Park Place, Cardiff CF10 3AT, U.K.
| | - Delia Belelli
- Division
of Neuroscience, School of Medicine, Medical Sciences Institute, Dundee University, Dow Street, Dundee DD1
5HL, U.K.
| | - Cecilia Rocha
- The
Neuro’s Early Drug Discovery Unit (EDDU), Department of Neurology
and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, 3801 University Street, Montreal, Quebec H3A 2B4, Canada
| | - Gilles Maussion
- The
Neuro’s Early Drug Discovery Unit (EDDU), Department of Neurology
and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, 3801 University Street, Montreal, Quebec H3A 2B4, Canada
| | - Andrea I. Krahn
- The
Neuro’s Early Drug Discovery Unit (EDDU), Department of Neurology
and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, 3801 University Street, Montreal, Quebec H3A 2B4, Canada
| | - Thomas M. Durcan
- The
Neuro’s Early Drug Discovery Unit (EDDU), Department of Neurology
and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, 3801 University Street, Montreal, Quebec H3A 2B4, Canada
| | - Jonathan M. Elkins
- Centre
for Medicines Discovery, University of Oxford, Roosevelt Drive, Oxford OX3 7DQ, U.K.
| | - Jeremy J. Lambert
- Division
of Neuroscience, School of Medicine, Medical Sciences Institute, Dundee University, Dow Street, Dundee DD1
5HL, U.K.
| | - John R. Atack
- Medicines
Discovery Institute, School of Biosciences, Cardiff University, Main Building, Park Place, Cardiff CF10 3AT, U.K.
| | - Simon E. Ward
- Medicines
Discovery Institute, School of Biosciences, Cardiff University, Main Building, Park Place, Cardiff CF10 3AT, U.K.
| |
Collapse
|
3
|
Vannelli A, Mariano V, Bagni C, Kanellopoulos AK. Activation of the 5-HT1A Receptor by Eltoprazine Restores Mitochondrial and Motor Deficits in a Drosophila Model of Fragile X Syndrome. Int J Mol Sci 2024; 25:8787. [PMID: 39201473 PMCID: PMC11354613 DOI: 10.3390/ijms25168787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 07/28/2024] [Accepted: 07/29/2024] [Indexed: 09/02/2024] Open
Abstract
Neurons rely on mitochondrial energy metabolism for essential functions like neurogenesis, neurotransmission, and synaptic plasticity. Mitochondrial dysfunctions are associated with neurodevelopmental disorders including Fragile X syndrome (FXS), the most common cause of inherited intellectual disability, which also presents with motor skill deficits. However, the precise role of mitochondria in the pathophysiology of FXS remains largely unknown. Notably, previous studies have linked the serotonergic system and mitochondrial activity to FXS. Our study investigates the potential therapeutic role of serotonin receptor 1A (5-HT1A) in FXS. Using the Drosophila model of FXS, we demonstrated that treatment with eltoprazine, a 5-HT1A agonist, can ameliorate synaptic transmission, correct mitochondrial deficits, and ultimately improve motor behavior. While these findings suggest that the 5-HT1A-mitochondrial axis may be a promising therapeutic target, further investigation is needed in the context of FXS.
Collapse
Affiliation(s)
- Anna Vannelli
- Department of Fundamental Neurosciences, University of Lausanne, 1005 Lausanne, Switzerland
| | - Vittoria Mariano
- Department of Fundamental Neurosciences, University of Lausanne, 1005 Lausanne, Switzerland
| | - Claudia Bagni
- Department of Fundamental Neurosciences, University of Lausanne, 1005 Lausanne, Switzerland
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133 Rome, Italy
| | | |
Collapse
|
4
|
Miles KD, Barker CM, Russell KP, Appel BH, Doll CA. Electrical Synapses Mediate Embryonic Hyperactivity in a Zebrafish Model of Fragile X Syndrome. J Neurosci 2024; 44:e2275232024. [PMID: 38969506 PMCID: PMC11293453 DOI: 10.1523/jneurosci.2275-23.2024] [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: 12/06/2023] [Revised: 06/19/2024] [Accepted: 06/27/2024] [Indexed: 07/07/2024] Open
Abstract
Although hyperactivity is associated with a wide variety of neurodevelopmental disorders, the early embryonic origins of locomotion have hindered investigation of pathogenesis of these debilitating behaviors. The earliest motor output in vertebrate animals is generated by clusters of early-born motor neurons (MNs) that occupy distinct regions of the spinal cord, innervating stereotyped muscle groups. Gap junction electrical synapses drive early spontaneous behavior in zebrafish, prior to the emergence of chemical neurotransmitter networks. We use a genetic model of hyperactivity to gain critical insight into the consequences of errors in motor circuit formation and function, finding that Fragile X syndrome model mutant zebrafish are hyperexcitable from the earliest phases of spontaneous behavior, show altered sensitivity to blockade of electrical gap junctions, and have increased expression of the gap junction protein Connexin 34/35. We further show that this hyperexcitable behavior can be rescued by pharmacological inhibition of electrical synapses. We also use functional imaging to examine MN and interneuron (IN) activity in early embryogenesis, finding genetic disruption of electrical gap junctions uncouples activity between mnx1 + MNs and INs. Taken together, our work highlights the importance of electrical synapses in motor development and suggests that the origins of hyperactivity in neurodevelopmental disorders may be established during the initial formation of locomotive circuits.
Collapse
Affiliation(s)
- Kaleb D Miles
- Section of Developmental Biology, Department of Pediatrics, Children's Hospital Colorado, University of Colorado, Anschutz Medical Campus, Aurora, Colorado 80045
| | - Chase M Barker
- Section of Developmental Biology, Department of Pediatrics, Children's Hospital Colorado, University of Colorado, Anschutz Medical Campus, Aurora, Colorado 80045
| | - Kristen P Russell
- Section of Developmental Biology, Department of Pediatrics, Children's Hospital Colorado, University of Colorado, Anschutz Medical Campus, Aurora, Colorado 80045
| | - Bruce H Appel
- Section of Developmental Biology, Department of Pediatrics, Children's Hospital Colorado, University of Colorado, Anschutz Medical Campus, Aurora, Colorado 80045
| | - Caleb A Doll
- Section of Developmental Biology, Department of Pediatrics, Children's Hospital Colorado, University of Colorado, Anschutz Medical Campus, Aurora, Colorado 80045
| |
Collapse
|
5
|
Stojkovic M, Petrovic M, Capovilla M, Milojevic S, Makevic V, Budimirovic DB, Corscadden L, He S, Protic D. Using a Combination of Novel Research Tools to Understand Social Interaction in the Drosophila melanogaster Model for Fragile X Syndrome. BIOLOGY 2024; 13:432. [PMID: 38927312 PMCID: PMC11200401 DOI: 10.3390/biology13060432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 05/31/2024] [Accepted: 06/06/2024] [Indexed: 06/28/2024]
Abstract
Fragile X syndrome (FXS), the most common monogenic cause of inherited intellectual disability and autism spectrum disorder, is caused by a full mutation (>200 CGG repeats) in the Fragile X Messenger Ribonucleoprotein 1 (FMR1) gene. Individuals with FXS experience various challenges related to social interaction (SI). Animal models, such as the Drosophila melanogaster model for FXS where the only ortholog of human FMR1 (dFMR1) is mutated, have played a crucial role in the understanding of FXS. The aim of this study was to investigate SI in the dFMR1B55 mutants (the groups of flies of both sexes simultaneously) using the novel Drosophila Shallow Chamber and a Python data processing pipeline based on social network analysis (SNA). In comparison with wild-type flies (w1118), SNA analysis in dFMR1B55 mutants revealed hypoactivity, fewer connections in their networks, longer interaction duration, a lower ability to transmit information efficiently, fewer alternative pathways for information transmission, a higher variability in the number of interactions they achieved, and flies tended to stay near the boundaries of the testing chamber. These observed alterations indicate the presence of characteristic strain-dependent social networks in dFMR1B55 flies, commonly referred to as the group phenotype. Finally, combining novel research tools is a valuable method for SI research in fruit flies.
Collapse
Affiliation(s)
- Maja Stojkovic
- Department of Pharmacology, Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia; (M.S.); (S.M.)
| | - Milan Petrovic
- Department of Informatics, University of Rijeka, 51000 Rijeka, Croatia;
- Center for Artificial Intelligence and Cybersecurity, University of Rijeka, 51000 Rijeka, Croatia
| | - Maria Capovilla
- UMR7275 CNRS-UCA, Institut de Pharmacologie Moléculaire et Cellulaire, 06560 Valbonne Sophia Antipolis, France;
| | - Sara Milojevic
- Department of Pharmacology, Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia; (M.S.); (S.M.)
| | - Vedrana Makevic
- Department of Pathophysiology, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia;
| | - Dejan B. Budimirovic
- Department of Psychiatry, Fragile X Clinic, Kennedy Krieger Institute, Baltimore, MD 21205, USA;
- Department of Psychiatry & Behavioral Sciences-Child Psychiatry, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA
| | | | - Shuhan He
- Lab of Computer Science, Department of Internal Medicine, Harvard Medical School, Boston, MA 02115, USA;
| | - Dragana Protic
- Department of Pharmacology, Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia; (M.S.); (S.M.)
- Fragile X Clinic, Special Hospital for Cerebral Palsy and Developmental Neurology, 11000 Belgrade, Serbia
| |
Collapse
|
6
|
Brija EA, Guan Z, Jetti SK, Littleton JT. Stochastic RNA editing of the Complexin C-terminus within single neurons regulates neurotransmitter release. Cell Rep 2023; 42:113152. [PMID: 37717212 PMCID: PMC10591831 DOI: 10.1016/j.celrep.2023.113152] [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: 07/19/2023] [Revised: 08/22/2023] [Accepted: 08/31/2023] [Indexed: 09/19/2023] Open
Abstract
Neurotransmitter release requires assembly of the SNARE complex fusion machinery, with multiple SNARE-binding proteins regulating when and where synaptic vesicle fusion occurs. The presynaptic protein Complexin (Cpx) controls spontaneous and evoked neurotransmitter release by modulating SNARE complex zippering. Although the central SNARE-binding helix is essential, post-translational modifications to Cpx's C-terminal membrane-binding amphipathic helix regulate its ability to control synaptic vesicle fusion. Here, we demonstrate that RNA editing of the Cpx C-terminus modifies its ability to clamp SNARE-mediated fusion and alters presynaptic output. RNA editing of Cpx across single neurons is stochastic, generating up to eight edit variants that fine tune neurotransmitter release by altering the subcellular localization and clamping properties of the protein. Similar stochastic editing rules for other synaptic genes were observed, indicating editing variability at single adenosines and across multiple mRNAs generates unique synaptic proteomes within the same population of neurons to fine tune presynaptic output.
Collapse
Affiliation(s)
- Elizabeth A Brija
- The Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Zhuo Guan
- The Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Suresh K Jetti
- The Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - J Troy Littleton
- The Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.
| |
Collapse
|
7
|
Westmark PR, Gholston AK, Swietlik TJ, Maganti RK, Westmark CJ. Ketogenic Diet Affects Sleep Architecture in C57BL/6J Wild Type and Fragile X Mice. Int J Mol Sci 2023; 24:14460. [PMID: 37833907 PMCID: PMC10572443 DOI: 10.3390/ijms241914460] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 09/15/2023] [Accepted: 09/18/2023] [Indexed: 10/15/2023] Open
Abstract
Nearly half of children with fragile X syndrome experience sleep problems including trouble falling asleep and frequent nighttime awakenings. The goals here were to assess sleep-wake cycles in mice in response to Fmr1 genotype and a dietary intervention that reduces hyperactivity. Electroencephalography (EEG) results were compared with published rest-activity patterns to determine if actigraphy is a viable surrogate for sleep EEG. Specifically, sleep-wake patterns in adult wild type and Fmr1KO littermate mice were recorded after EEG electrode implantation and the recordings manually scored for vigilance states. The data indicated that Fmr1KO mice exhibited sleep-wake patterns similar to wild type littermates when maintained on a control purified ingredient diet. Treatment with a high-fat, low-carbohydrate ketogenic diet increased the percentage of non-rapid eye movement (NREM) sleep in both wild type and Fmr1KO mice during the dark cycle, which corresponded to decreased activity levels. Treatment with a ketogenic diet flattened diurnal sleep periodicity in both wild type and Fmr1KO mice. Differences in several sleep microstructure outcomes (number and length of sleep and wake bouts) supported the altered sleep states in response to a ketogenic diet and were correlated with altered rest-activity cycles. While actigraphy may be a less expensive, reduced labor surrogate for sleep EEG during the dark cycle, daytime resting in mice did not correlate with EEG sleep states.
Collapse
Affiliation(s)
- Pamela R. Westmark
- Department of Neurology, University of Wisconsin, Madison, WI 53706, USA; (P.R.W.); (A.K.G.); (T.J.S.); (R.K.M.)
| | - Aaron K. Gholston
- Department of Neurology, University of Wisconsin, Madison, WI 53706, USA; (P.R.W.); (A.K.G.); (T.J.S.); (R.K.M.)
- Molecular Environmental Toxicology Center, University of Wisconsin, Madison, WI 53706, USA
| | - Timothy J. Swietlik
- Department of Neurology, University of Wisconsin, Madison, WI 53706, USA; (P.R.W.); (A.K.G.); (T.J.S.); (R.K.M.)
| | - Rama K. Maganti
- Department of Neurology, University of Wisconsin, Madison, WI 53706, USA; (P.R.W.); (A.K.G.); (T.J.S.); (R.K.M.)
| | - Cara J. Westmark
- Department of Neurology, University of Wisconsin, Madison, WI 53706, USA; (P.R.W.); (A.K.G.); (T.J.S.); (R.K.M.)
- Molecular Environmental Toxicology Center, University of Wisconsin, Madison, WI 53706, USA
| |
Collapse
|
8
|
Brija EA, Guan Z, Jetti SK, Littleton JT. Stochastic RNA editing of the Complexin C-terminus within single neurons regulates neurotransmitter release. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.30.542887. [PMID: 37398117 PMCID: PMC10312600 DOI: 10.1101/2023.05.30.542887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Neurotransmitter release requires assembly of the SNARE complex fusion machinery, with multiple SNARE-binding proteins regulating this process to control when and where synaptic vesicle fusion occurs. Complexin (Cpx) controls spontaneous and evoked neurotransmitter release by modulating SNARE complex zippering. Although the central SNARE-binding helix is essential, post-translational modifications to Cpx's C-terminal membrane-binding amphipathic helix modulate its activity. Here we demonstrate that RNA editing of the Cpx C-terminus regulates its ability to clamp SNARE-mediated fusion and alters presynaptic output. RNA editing of Cpx within single neurons is stochastic, generating up to eight edit variants that fine-tune neurotransmitter release by changing the subcellular localization and clamping properties of the protein. Similar editing rules for other synaptic genes were observed, indicating stochastic editing at single adenosines and across multiple mRNAs can generate unique synaptic proteomes within the same population of neurons to fine-tune presynaptic output.
Collapse
Affiliation(s)
- Elizabeth A Brija
- The Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Zhuo Guan
- The Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Suresh K Jetti
- The Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - J Troy Littleton
- The Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139
| |
Collapse
|
9
|
Geng J, Khaket TP, Pan J, Li W, Zhang Y, Ping Y, Cobos Sillero MI, Lu B. Deregulation of ER-mitochondria contact formation and mitochondrial calcium homeostasis mediated by VDAC in fragile X syndrome. Dev Cell 2023; 58:597-615.e10. [PMID: 37040696 PMCID: PMC10113018 DOI: 10.1016/j.devcel.2023.03.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Revised: 07/31/2022] [Accepted: 03/06/2023] [Indexed: 04/13/2023]
Abstract
Loss of fragile X messenger ribonucleoprotein (FMRP) causes fragile X syndrome (FXS), the most prevalent form of inherited intellectual disability. Here, we show that FMRP interacts with the voltage-dependent anion channel (VDAC) to regulate the formation and function of endoplasmic reticulum (ER)-mitochondria contact sites (ERMCSs), structures that are critical for mitochondrial calcium (mito-Ca2+) homeostasis. FMRP-deficient cells feature excessive ERMCS formation and ER-to-mitochondria Ca2+ transfer. Genetic and pharmacological inhibition of VDAC or other ERMCS components restored synaptic structure, function, and plasticity and rescued locomotion and cognitive deficits of the Drosophila dFmr1 mutant. Expressing FMRP C-terminal domain (FMRP-C), which confers FMRP-VDAC interaction, rescued the ERMCS formation and mito-Ca2+ homeostasis defects in FXS patient iPSC-derived neurons and locomotion and cognitive deficits in Fmr1 knockout mice. These results identify altered ERMCS formation and mito-Ca2+ homeostasis as contributors to FXS and offer potential therapeutic targets.
Collapse
Affiliation(s)
- Ji Geng
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Tejinder Pal Khaket
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Jie Pan
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Wen Li
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Yan Zhang
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai 200240, China; Shanghai Key Laboratory of Psychotic Disorders (No. 13dz2260500), Shanghai Mental Health Center, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Yong Ping
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai 200240, China; Shanghai Key Laboratory of Psychotic Disorders (No. 13dz2260500), Shanghai Mental Health Center, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China
| | | | - Bingwei Lu
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
10
|
Villalonga E, Mosrin C, Normand T, Girardin C, Serrano A, Žunar B, Doudeau M, Godin F, Bénédetti H, Vallée B. LIM Kinases, LIMK1 and LIMK2, Are Crucial Node Actors of the Cell Fate: Molecular to Pathological Features. Cells 2023; 12:cells12050805. [PMID: 36899941 PMCID: PMC10000741 DOI: 10.3390/cells12050805] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 02/28/2023] [Accepted: 03/02/2023] [Indexed: 03/08/2023] Open
Abstract
LIM kinase 1 (LIMK1) and LIM kinase 2 (LIMK2) are serine/threonine and tyrosine kinases and the only two members of the LIM kinase family. They play a crucial role in the regulation of cytoskeleton dynamics by controlling actin filaments and microtubule turnover, especially through the phosphorylation of cofilin, an actin depolymerising factor. Thus, they are involved in many biological processes, such as cell cycle, cell migration, and neuronal differentiation. Consequently, they are also part of numerous pathological mechanisms, especially in cancer, where their involvement has been reported for a few years and has led to the development of a wide range of inhibitors. LIMK1 and LIMK2 are known to be part of the Rho family GTPase signal transduction pathways, but many more partners have been discovered over the decades, and both LIMKs are suspected to be part of an extended and various range of regulation pathways. In this review, we propose to consider the different molecular mechanisms involving LIM kinases and their associated signalling pathways, and to offer a better understanding of their variety of actions within the physiology and physiopathology of the cell.
Collapse
Affiliation(s)
- Elodie Villalonga
- Centre de Biophysique Moléculaire; UPR4301, CNRS, University of Orleans and INSERM, CEDEX 2, 45071 Orleans, France
| | - Christine Mosrin
- Centre de Biophysique Moléculaire; UPR4301, CNRS, University of Orleans and INSERM, CEDEX 2, 45071 Orleans, France
| | - Thierry Normand
- Centre de Biophysique Moléculaire; UPR4301, CNRS, University of Orleans and INSERM, CEDEX 2, 45071 Orleans, France
| | - Caroline Girardin
- Centre de Biophysique Moléculaire; UPR4301, CNRS, University of Orleans and INSERM, CEDEX 2, 45071 Orleans, France
| | - Amandine Serrano
- Centre de Biophysique Moléculaire; UPR4301, CNRS, University of Orleans and INSERM, CEDEX 2, 45071 Orleans, France
| | - Bojan Žunar
- Laboratory for Biochemistry, Department of Chemistry and Biochemistry, Faculty of Food Technology and Biotechnology, University of Zagreb, 10000 Zagreb, Croatia
| | - Michel Doudeau
- Centre de Biophysique Moléculaire; UPR4301, CNRS, University of Orleans and INSERM, CEDEX 2, 45071 Orleans, France
| | - Fabienne Godin
- Centre de Biophysique Moléculaire; UPR4301, CNRS, University of Orleans and INSERM, CEDEX 2, 45071 Orleans, France
| | - Hélène Bénédetti
- Centre de Biophysique Moléculaire; UPR4301, CNRS, University of Orleans and INSERM, CEDEX 2, 45071 Orleans, France
| | - Béatrice Vallée
- Centre de Biophysique Moléculaire; UPR4301, CNRS, University of Orleans and INSERM, CEDEX 2, 45071 Orleans, France
- Correspondence: ; Tel.: +33-(0)2-38-25-76-11
| |
Collapse
|
11
|
Trajković J, Makevic V, Pesic M, Pavković-Lučić S, Milojevic S, Cvjetkovic S, Hagerman R, Budimirovic DB, Protic D. Drosophila melanogaster as a Model to Study Fragile X-Associated Disorders. Genes (Basel) 2022; 14:genes14010087. [PMID: 36672829 PMCID: PMC9859539 DOI: 10.3390/genes14010087] [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: 11/21/2022] [Revised: 12/22/2022] [Accepted: 12/23/2022] [Indexed: 12/30/2022] Open
Abstract
Fragile X syndrome (FXS) is a global neurodevelopmental disorder caused by the expansion of CGG trinucleotide repeats (≥200) in the Fragile X Messenger Ribonucleoprotein 1 (FMR1) gene. FXS is the hallmark of Fragile X-associated disorders (FXD) and the most common monogenic cause of inherited intellectual disability and autism spectrum disorder. There are several animal models used to study FXS. In the FXS model of Drosophila, the only ortholog of FMR1, dfmr1, is mutated so that its protein is missing. This model has several relevant phenotypes, including defects in the circadian output pathway, sleep problems, memory deficits in the conditioned courtship and olfactory conditioning paradigms, deficits in social interaction, and deficits in neuronal development. In addition to FXS, a model of another FXD, Fragile X-associated tremor/ataxia syndrome (FXTAS), has also been established in Drosophila. This review summarizes many years of research on FXD in Drosophila models.
Collapse
Affiliation(s)
- Jelena Trajković
- Faculty of Biology, University of Belgrade, 11000 Belgrade, Serbia
| | - Vedrana Makevic
- Department of Pathophysiology, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
| | - Milica Pesic
- Institute of Human Genetics, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
| | | | - Sara Milojevic
- Department of Pharmacology, Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
| | - Smiljana Cvjetkovic
- Department of Humanities, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
| | - Randi Hagerman
- Medical Investigation of Neurodevelopmental Disorders (MIND) Institute, University of California Davis, 2825 50th Street, Sacramento, CA 95817, USA
- Department of Pediatrics, University of California Davis School of Medicine, Sacramento, CA 95817, USA
| | - Dejan B. Budimirovic
- Department of Psychiatry, Fragile X Clinic, Kennedy Krieger Institute, Baltimore, MD 21205, USA
- Department of Psychiatry & Behavioral Sciences-Child Psychiatry, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Dragana Protic
- Department of Pharmacology, Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
- Correspondence:
| |
Collapse
|
12
|
Karagas NE, Gupta R, Rastegari E, Tan KL, Leung HH, Bellen HJ, Venkatachalam K, Wong CO. Loss of Activity-Induced Mitochondrial ATP Production Underlies the Synaptic Defects in a Drosophila Model of ALS. J Neurosci 2022; 42:8019-8037. [PMID: 36261266 PMCID: PMC9617612 DOI: 10.1523/jneurosci.2456-21.2022] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 08/23/2022] [Accepted: 08/27/2022] [Indexed: 11/21/2022] Open
Abstract
Mutations in the gene encoding vesicle-associated membrane protein B (VAPB) cause a familial form of amyotrophic lateral sclerosis (ALS). Expression of an ALS-related variant of vapb (vapbP58S ) in Drosophila motor neurons results in morphologic changes at the larval neuromuscular junction (NMJ) characterized by the appearance of fewer, but larger, presynaptic boutons. Although diminished microtubule stability is known to underlie these morphologic changes, a mechanism for the loss of presynaptic microtubules has been lacking. By studying flies of both sexes, we demonstrate the suppression of vapbP58S -induced changes in NMJ morphology by either a loss of endoplasmic reticulum (ER) Ca2+ release channels or the inhibition Ca2+/calmodulin (CaM)-activated kinase II (CaMKII). These data suggest that decreased stability of presynaptic microtubules at vapbP58S NMJs results from hyperactivation of CaMKII because of elevated cytosolic [Ca2+]. We attribute the Ca2+ dyshomeostasis to delayed extrusion of cytosolic Ca2+ Suggesting that this defect in Ca2+ extrusion arose from an insufficient response to the bioenergetic demand of neural activity, depolarization-induced mitochondrial ATP production was diminished in vapbP58S neurons. These findings point to bioenergetic dysfunction as a potential cause for the synaptic defects in vapbP58S -expressing motor neurons.SIGNIFICANCE STATEMENT Whether the synchrony between the rates of ATP production and demand is lost in degenerating neurons remains poorly understood. We report that expression of a gene equivalent to an amyotrophic lateral sclerosis (ALS)-causing variant of vesicle-associated membrane protein B (VAPB) in fly neurons decouples mitochondrial ATP production from neuronal activity. Consequently, levels of ATP in mutant neurons are unable to keep up with the bioenergetic burden of neuronal activity. Reduced rate of Ca2+ extrusion, which could result from insufficient energy to power Ca2+ ATPases, results in the accumulation of residual Ca2+ in mutant neurons and leads to alterations in synaptic vesicle (SV) release and synapse development. These findings suggest that synaptic defects in a model of ALS arise from the loss of activity-induced ATP production.
Collapse
Affiliation(s)
- Nicholas E Karagas
- Department of Integrative Biology and Pharmacology, McGovern Medical School at the University of Texas Health Sciences Center, Houston, Texas 77030
- Graduate Program in Biochemistry and Cell Biology, MD Anderson Cancer Center and University of Texas Health Sciences Center Graduate School of Biomedical Sciences, Houston, TX, 77030
| | - Richa Gupta
- Department of Integrative Biology and Pharmacology, McGovern Medical School at the University of Texas Health Sciences Center, Houston, Texas 77030
| | - Elham Rastegari
- Department of Integrative Biology and Pharmacology, McGovern Medical School at the University of Texas Health Sciences Center, Houston, Texas 77030
| | - Kai Li Tan
- Departments of Molecular and Human Genetics and Neuroscience, Baylor College of Medicine, Houston, TX 77030
- Graduate Program in Developmental Biology, Baylor College of Medicine, Houston, Texas 77030
- Duncan Neurological Research Institute, Texas Children Hospital, Houston, Texas 77030
| | - Ho Hang Leung
- Department of Biological Sciences, Rutgers University, Newark, New Jersey 07102
| | - Hugo J Bellen
- Departments of Molecular and Human Genetics and Neuroscience, Baylor College of Medicine, Houston, TX 77030
- Graduate Program in Developmental Biology, Baylor College of Medicine, Houston, Texas 77030
- Duncan Neurological Research Institute, Texas Children Hospital, Houston, Texas 77030
| | - Kartik Venkatachalam
- Department of Integrative Biology and Pharmacology, McGovern Medical School at the University of Texas Health Sciences Center, Houston, Texas 77030
- Graduate Program in Biochemistry and Cell Biology, MD Anderson Cancer Center and University of Texas Health Sciences Center Graduate School of Biomedical Sciences, Houston, TX, 77030
- Graduate Program in Neuroscience, MD Anderson Cancer Center and University of Texas Health Sciences Center Graduate School of Biomedical Sciences, Houston, TX, 77030
| | - Ching-On Wong
- Department of Biological Sciences, Rutgers University, Newark, New Jersey 07102
| |
Collapse
|
13
|
Kuznitsov-Yanovsky L, Shapira G, Gildin L, Shomron N, Ben-Yosef D. Transcriptomic Analysis of Human Fragile X Syndrome Neurons Reveals Neurite Outgrowth Modulation by the TGFβ/BMP Pathway. Int J Mol Sci 2022; 23:ijms23169278. [PMID: 36012539 PMCID: PMC9409179 DOI: 10.3390/ijms23169278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 08/11/2022] [Accepted: 08/14/2022] [Indexed: 11/25/2022] Open
Abstract
Fragile X Syndrome (FXS) is the main genetic reason for intellectual disability and is caused by the silencing of fragile X mental retardation protein (FMRP), an RNA-binding protein regulating the translation of many neuronal mRNAs. Neural differentiation of FX human embryonic stem cells (hESC) mimics the neurodevelopment of FXS fetuses and thus serves as a good model to explore the mechanisms underlining the development of FXS. Isogenic hESC clones with and without the FX mutation that share the same genetic background were in vitro differentiated into neurons, and their transcriptome was analyzed by RNA sequencing. FX neurons inactivating FMR1 expression presented delayed neuronal development and maturation, concomitant with dysregulation of the TGFβ/BMP signaling pathway, and genes related to the extracellular matrix. Migration assay showed decreased neurite outgrowth in FX neurons that was rescued by inhibition of the TGFβ/BMP signaling pathway. Our results provide new insights into the molecular pathway by which loss of FMRP affects neuronal network development. In FX neurons, the lack of FMRP dysregulates members of the BMP signaling pathway associated with ECM organization which, in a yet unknown mechanism, reduces the guidance of axonal growth cones, probably leading to the aberrant neuronal network function seen in FXS.
Collapse
Affiliation(s)
- Liron Kuznitsov-Yanovsky
- Wolfe PGD Stem Cell Lab, Racine IVF Unit, Lis Maternity Hospital Tel-Aviv Sourasky Medical Center, Tel Aviv 64239, Israel
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel-Aviv University, Tel Aviv 69978, Israel
| | - Guy Shapira
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel-Aviv University, Tel Aviv 69978, Israel
| | - Lital Gildin
- Wolfe PGD Stem Cell Lab, Racine IVF Unit, Lis Maternity Hospital Tel-Aviv Sourasky Medical Center, Tel Aviv 64239, Israel
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel-Aviv University, Tel Aviv 69978, Israel
| | - Noam Shomron
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel-Aviv University, Tel Aviv 69978, Israel
- Sagol School of Neuroscience, Tel-Aviv University, Tel Aviv 69978, Israel
| | - Dalit Ben-Yosef
- Wolfe PGD Stem Cell Lab, Racine IVF Unit, Lis Maternity Hospital Tel-Aviv Sourasky Medical Center, Tel Aviv 64239, Israel
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel-Aviv University, Tel Aviv 69978, Israel
- Sagol School of Neuroscience, Tel-Aviv University, Tel Aviv 69978, Israel
- Correspondence:
| |
Collapse
|
14
|
Chakravorty A, Sharma A, Sheeba V, Manjithaya R. Glutamatergic Synapse Dysfunction in Drosophila Neuromuscular Junctions Can Be Rescued by Proteostasis Modulation. Front Mol Neurosci 2022; 15:842772. [PMID: 35909443 PMCID: PMC9337869 DOI: 10.3389/fnmol.2022.842772] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Accepted: 04/25/2022] [Indexed: 11/29/2022] Open
Abstract
Glutamate is the major excitatory neurotransmitter in the nervous system, and the Drosophila glutamatergic neuromuscular junctions (NMJs) offer a tractable platform to understand excitatory synapse biology both in health and disease. Synaptopathies are neurodegenerative diseases that are associated with synaptic dysfunction and often display compromised proteostasis. One such rare, progressive neurodegenerative condition, Spinocerebellar Ataxia Type 3 (SCA3) or Machado-Joseph Disease (MJD), is characterized by cerebellar ataxia, Parkinsonism, and degeneration of motor neuron synapses. While the polyQ repeat mutant protein ataxin-3 is implicated in MJD, it is unclear how it leads to impaired synaptic function. In this study, we indicated that a Drosophila model of MJD recapitulates characteristics of neurodegenerative disorders marked by motor neuron dysfunction. Expression of 78 polyQ repeats of mutant ataxin-3 protein in Drosophila motor neurons resulted in behavioral defects, such as impaired locomotion in both larval and adult stages. Furthermore, defects in eclosion and lifespan were observed in adult flies. Detailed characterization of larval glutamatergic neuromuscular junctions (NMJs) revealed defects in morphological features along with compromised NMJ functioning. Autophagy, one of the key proteostasis pathways, is known to be impaired in the case of several synaptopathies. Our study reveals that overexpression of the autophagy-related protein Atg8a rescued behavioral defects. Thus, we present a model for glutamatergic synapse dysfunction that recapitulates synaptic and behavioral deficits and show that it is an amenable system for carrying out genetic and chemical biology screens to identify potential therapeutic targets for synaptopathies.
Collapse
Affiliation(s)
- Anushka Chakravorty
- Autophagy Laboratory, Molecular Biology and Genetics Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bangalore, India
| | - Ankit Sharma
- Chronobiology and Behavioural Neurogenetics Laboratory, Neuroscience Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bangalore, India
| | - Vasu Sheeba
- Chronobiology and Behavioural Neurogenetics Laboratory, Neuroscience Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bangalore, India
- *Correspondence: Vasu Sheeba
| | - Ravi Manjithaya
- Autophagy Laboratory, Molecular Biology and Genetics Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bangalore, India
- Neuroscience Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bangalore, India
- Ravi Manjithaya
| |
Collapse
|
15
|
Song C, Broadie K. Dysregulation of BMP, Wnt, and Insulin Signaling in Fragile X Syndrome. Front Cell Dev Biol 2022; 10:934662. [PMID: 35880195 PMCID: PMC9307498 DOI: 10.3389/fcell.2022.934662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Accepted: 06/06/2022] [Indexed: 01/21/2023] Open
Abstract
Drosophila models of neurological disease contribute tremendously to research progress due to the high conservation of human disease genes, the powerful and sophisticated genetic toolkit, and the rapid generation time. Fragile X syndrome (FXS) is the most prevalent heritable cause of intellectual disability and autism spectrum disorders, and the Drosophila FXS disease model has been critical for the genetic screening discovery of new intercellular secretion mechanisms. Here, we focus on the roles of three major signaling pathways: BMP, Wnt, and insulin-like peptides. We present Drosophila FXS model defects compared to mouse models in stem cells/embryos, the glutamatergic neuromuscular junction (NMJ) synapse model, and the developing adult brain. All three of these secreted signaling pathways are strikingly altered in FXS disease models, giving new mechanistic insights into impaired cellular outcomes and neurological phenotypes. Drosophila provides a powerful genetic screening platform to expand understanding of these secretory mechanisms and to test cellular roles in both peripheral and central nervous systems. The studies demonstrate the importance of exploring broad genetic interactions and unexpected regulatory mechanisms. We discuss a number of research avenues to pursue BMP, Wnt, and insulin signaling in future FXS investigations and the development of potential therapeutics.
Collapse
Affiliation(s)
- Chunzhu Song
- Department of Biological Sciences, College of Arts and Science, Vanderbilt University, Nashville, TN, United States
| | - Kendal Broadie
- Department of Biological Sciences, College of Arts and Science, Vanderbilt University, Nashville, TN, United States
- Department of Cell and Developmental Biology, School of Medicine, Vanderbilt University, Nashville, TN, United States
- Kennedy Center for Research on Human Development, Nashville, TN, United States
- Vanderbilt Brain Institute, School of Medicine, Vanderbilt University and Medical Center, Nashville, TN, United States
| |
Collapse
|
16
|
Welch C, Johnson E, Tupikova A, Anderson J, Tinsley B, Newman J, Widman E, Alfareh A, Davis A, Rodriguez L, Visger C, Miller-Schulze JP, Lee W, Mulligan K. Bisphenol a affects neurodevelopmental gene expression, cognitive function, and neuromuscular synaptic morphology in Drosophila melanogaster. Neurotoxicology 2022; 89:67-78. [PMID: 35041872 DOI: 10.1016/j.neuro.2022.01.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 01/05/2022] [Accepted: 01/12/2022] [Indexed: 01/02/2023]
Abstract
Bisphenol A (BPA) is an environmentally prevalent endocrine disrupting chemical that can impact human health and may be an environmental risk factor for neurodevelopmental disorders. BPA has been associated with behavioral impairment in children and a variety of neurodevelopmental phenotypes in model organisms. We used Drosophila melanogaster to explore the consequences of developmental BPA exposure on gene expression, cognitive function, and synapse development. Our transcriptome analysis indicated neurodevelopmentally relevant genes were predominantly downregulated by BPA. Among the misregulated genes were those with roles in learning, memory, and synapse development, as well as orthologs of human genes associated with neurodevelopmental and neuropsychiatric disorders. To examine how gene expression data corresponded to behavioral and cellular phenotypes, we first used a predator-response behavioral paradigm and found that BPA disrupts visual perception. Further analysis using conditioned courtship suppression showed that BPA impairs associative learning. Finally, we examined synapse morphology within the larval neuromuscular junction and found that BPA significantly increased the number of axonal branches. Given that our findings align with studies of BPA in mammalian model organisms, this data indicates that BPA impairs neurodevelopmental pathways that are functionally conserved from invertebrates to mammals. Further, because Drosophila do not possess classic estrogen receptors or estrogen, this research suggests that BPA can impact neurodevelopment by molecular mechanisms distinct from its role as an estrogen mimic.
Collapse
Affiliation(s)
- Chloe Welch
- Department of Biological Sciences, California State University, Sacramento, 6000 J Street, Sacramento, CA, 95819-6077, USA
| | - Eden Johnson
- Department of Computer Science, San José State University, 1 Washington Sq, San Jose, CA, 95192, USA
| | - Angelina Tupikova
- Department of Biological Sciences, California State University, Sacramento, 6000 J Street, Sacramento, CA, 95819-6077, USA
| | - Judith Anderson
- Department of Biological Sciences, California State University, Sacramento, 6000 J Street, Sacramento, CA, 95819-6077, USA
| | - Brendan Tinsley
- Department of Biological Sciences, California State University, Sacramento, 6000 J Street, Sacramento, CA, 95819-6077, USA
| | - Johnathan Newman
- Department of Biological Sciences, California State University, Sacramento, 6000 J Street, Sacramento, CA, 95819-6077, USA
| | - Erin Widman
- Department of Biological Sciences, California State University, Sacramento, 6000 J Street, Sacramento, CA, 95819-6077, USA
| | - Adam Alfareh
- Department of Biological Sciences, California State University, Sacramento, 6000 J Street, Sacramento, CA, 95819-6077, USA
| | - Alexandra Davis
- Department of Biological Sciences, California State University, Sacramento, 6000 J Street, Sacramento, CA, 95819-6077, USA
| | - Lucero Rodriguez
- Department of Chemistry, California State University, Sacramento, 6000 J Street, Sacramento, CA, 95819-6077, USA
| | - Clayton Visger
- Department of Biological Sciences, California State University, Sacramento, 6000 J Street, Sacramento, CA, 95819-6077, USA
| | - Justin P Miller-Schulze
- Department of Chemistry, California State University, Sacramento, 6000 J Street, Sacramento, CA, 95819-6077, USA
| | - Wendy Lee
- Department of Computer Science, San José State University, 1 Washington Sq, San Jose, CA, 95192, USA
| | - Kimberly Mulligan
- Department of Biological Sciences, California State University, Sacramento, 6000 J Street, Sacramento, CA, 95819-6077, USA.
| |
Collapse
|
17
|
LIM-Kinases in Synaptic Plasticity, Memory, and Brain Diseases. Cells 2021; 10:cells10082079. [PMID: 34440848 PMCID: PMC8391678 DOI: 10.3390/cells10082079] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 07/30/2021] [Accepted: 08/04/2021] [Indexed: 12/12/2022] Open
Abstract
Learning and memory require structural and functional modifications of synaptic connections, and synaptic deficits are believed to underlie many brain disorders. The LIM-domain-containing protein kinases (LIMK1 and LIMK2) are key regulators of the actin cytoskeleton by affecting the actin-binding protein, cofilin. In addition, LIMK1 is implicated in the regulation of gene expression by interacting with the cAMP-response element-binding protein. Accumulating evidence indicates that LIMKs are critically involved in brain function and dysfunction. In this paper, we will review studies on the roles and underlying mechanisms of LIMKs in the regulation of long-term potentiation (LTP) and depression (LTD), the most extensively studied forms of long-lasting synaptic plasticity widely regarded as cellular mechanisms underlying learning and memory. We will also discuss the involvement of LIMKs in the regulation of the dendritic spine, the structural basis of synaptic plasticity, and memory formation. Finally, we will discuss recent progress on investigations of LIMKs in neurological and mental disorders, including Alzheimer’s, Parkinson’s, Williams–Beuren syndrome, schizophrenia, and autism spectrum disorders.
Collapse
|
18
|
Titus MB, Wright EG, Bono JM, Poliakon AK, Goldstein BR, Super MK, Young LA, Manaj M, Litchford M, Reist NE, Killian DJ, Olesnicky EC. The conserved alternative splicing factor caper regulates neuromuscular phenotypes during development and aging. Dev Biol 2021; 473:15-32. [PMID: 33508255 PMCID: PMC7987824 DOI: 10.1016/j.ydbio.2021.01.011] [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: 06/23/2020] [Revised: 01/18/2021] [Accepted: 01/19/2021] [Indexed: 12/17/2022]
Abstract
RNA-binding proteins play an important role in the regulation of post-transcriptional gene expression throughout the nervous system. This is underscored by the prevalence of mutations in genes encoding RNA splicing factors and other RNA-binding proteins in a number of neurodegenerative and neurodevelopmental disorders. The highly conserved alternative splicing factor Caper is widely expressed throughout the developing embryo and functions in the development of various sensory neural subtypes in the Drosophila peripheral nervous system. Here we find that caper dysfunction leads to aberrant neuromuscular junction morphogenesis, as well as aberrant locomotor behavior during larval and adult stages. Despite its widespread expression, our results indicate that caper function is required to a greater extent within the nervous system, as opposed to muscle, for neuromuscular junction development and for the regulation of adult locomotor behavior. Moreover, we find that Caper interacts with the RNA-binding protein Fmrp to regulate adult locomotor behavior. Finally, we show that caper dysfunction leads to various phenotypes that have both a sex and age bias, both of which are commonly seen in neurodegenerative disorders in humans.
Collapse
Affiliation(s)
- M Brandon Titus
- Department of Biology, University of Colorado Colorado Springs, Colorado Springs, CO, 80918, USA
| | - Ethan G Wright
- Department of Biology, University of Colorado Colorado Springs, Colorado Springs, CO, 80918, USA
| | - Jeremy M Bono
- Department of Biology, University of Colorado Colorado Springs, Colorado Springs, CO, 80918, USA
| | - Andrea K Poliakon
- Department of Biology, University of Colorado Colorado Springs, Colorado Springs, CO, 80918, USA
| | - Brandon R Goldstein
- Department of Biology, University of Colorado Colorado Springs, Colorado Springs, CO, 80918, USA
| | - Meg K Super
- Department of Biology, University of Colorado Colorado Springs, Colorado Springs, CO, 80918, USA
| | - Lauren A Young
- Department of Biology, University of Colorado Colorado Springs, Colorado Springs, CO, 80918, USA
| | - Melpomeni Manaj
- Department of Biology, University of Colorado Colorado Springs, Colorado Springs, CO, 80918, USA
| | - Morgan Litchford
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO, 80523, USA
| | - Noreen E Reist
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO, 80523, USA
| | - Darrell J Killian
- Department of Molecular Biology, Colorado College, Colorado Springs, CO, 80903, USA
| | - Eugenia C Olesnicky
- Department of Biology, University of Colorado Colorado Springs, Colorado Springs, CO, 80918, USA.
| |
Collapse
|
19
|
Belalcazar HM, Hendricks EL, Zamurrad S, Liebl FLW, Secombe J. The histone demethylase KDM5 is required for synaptic structure and function at the Drosophila neuromuscular junction. Cell Rep 2021; 34:108753. [PMID: 33596422 PMCID: PMC7945993 DOI: 10.1016/j.celrep.2021.108753] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 12/14/2020] [Accepted: 01/25/2021] [Indexed: 02/08/2023] Open
Abstract
Mutations in the genes encoding the lysine demethylase 5 (KDM5) family of histone demethylases are observed in individuals with intellectual disability (ID). Despite clear evidence linking KDM5 function to neurodevelopmental pathways, how this family of proteins impacts transcriptional programs to mediate synaptic structure and activity remains unclear. Using the Drosophila larval neuromuscular junction (NMJ), we show that KDM5 is required presynaptically for neuroanatomical development and synaptic function. The Jumonji C (JmjC) domain-encoded histone demethylase activity of KDM5, which is expected to be diminished by many ID-associated alleles, is required for appropriate synaptic morphology and neurotransmission. The activity of the C5HC2 zinc finger is also required, as an ID-associated mutation in this motif reduces NMJ bouton number, increases bouton size, and alters microtubule dynamics. KDM5 therefore uses demethylase-dependent and independent mechanisms to regulate NMJ structure and activity, highlighting the complex nature by which this chromatin modifier carries out its neuronal gene-regulatory programs.
Collapse
Affiliation(s)
- Helen M Belalcazar
- Department of Genetics, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461, USA
| | - Emily L Hendricks
- Department of Biological Sciences, Southern Illinois University Edwardsville, 44 Circle Drive, Edwardsville, IL 62026, USA
| | - Sumaira Zamurrad
- Department of Genetics, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461, USA
| | - Faith L W Liebl
- Department of Biological Sciences, Southern Illinois University Edwardsville, 44 Circle Drive, Edwardsville, IL 62026, USA
| | - Julie Secombe
- Department of Genetics, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461, USA; Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, 1410 Pelham Parkway South, Bronx, NY 10461, USA.
| |
Collapse
|
20
|
Worpenberg L, Paolantoni C, Longhi S, Mulorz MM, Lence T, Wessels HH, Dassi E, Aiello G, Sutandy FXR, Scheibe M, Edupuganti RR, Busch A, Möckel MM, Vermeulen M, Butter F, König J, Notarangelo M, Ohler U, Dieterich C, Quattrone A, Soldano A, Roignant JY. Ythdf is a N6-methyladenosine reader that modulates Fmr1 target mRNA selection and restricts axonal growth in Drosophila. EMBO J 2021; 40:e104975. [PMID: 33428246 PMCID: PMC7883056 DOI: 10.15252/embj.2020104975] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 11/18/2020] [Accepted: 11/30/2020] [Indexed: 12/19/2022] Open
Abstract
N6‐methyladenosine (m6A) regulates a variety of physiological processes through modulation of RNA metabolism. This modification is particularly enriched in the nervous system of several species, and its dysregulation has been associated with neurodevelopmental defects and neural dysfunctions. In Drosophila, loss of m6A alters fly behavior, albeit the underlying molecular mechanism and the role of m6A during nervous system development have remained elusive. Here we find that impairment of the m6A pathway leads to axonal overgrowth and misguidance at larval neuromuscular junctions as well as in the adult mushroom bodies. We identify Ythdf as the main m6A reader in the nervous system, being required to limit axonal growth. Mechanistically, we show that the m6A reader Ythdf directly interacts with Fmr1, the fly homolog of Fragile X mental retardation RNA binding protein (FMRP), to inhibit the translation of key transcripts involved in axonal growth regulation. Altogether, this study demonstrates that the m6A pathway controls development of the nervous system and modulates Fmr1 target transcript selection.
Collapse
Affiliation(s)
- Lina Worpenberg
- Center for Integrative Genomics, Génopode Building, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Chiara Paolantoni
- Center for Integrative Genomics, Génopode Building, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Sara Longhi
- Laboratory of Translational Genomics, Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Trento, Italy
| | | | - Tina Lence
- Institute of Molecular Biology (IMB), Mainz, Germany
| | - Hans-Hermann Wessels
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin Institute for Medical Systems Biology (BIMSB), Berlin, Germany.,Department of Biology, Humboldt University Berlin, Berlin, Germany
| | - Erik Dassi
- Laboratory of RNA Regulatory Networks, Department CIBIO, University of Trento, Trento, Italy
| | - Giuseppe Aiello
- Armenise-Harvard Laboratory of Brain Disorders and Cancer, Department CIBIO, University of Trento, Trento, Italy
| | | | | | - Raghu R Edupuganti
- Department of Molecular Biology, Faculty of Science, Radboud Institute for Molecular Life Sciences, Oncode Institute, Radboud University Nijmegen, Nijmegen, The Netherlands
| | - Anke Busch
- Bioinformatics Core Facility, IMB, Mainz, Germany
| | | | - Michiel Vermeulen
- Department of Molecular Biology, Faculty of Science, Radboud Institute for Molecular Life Sciences, Oncode Institute, Radboud University Nijmegen, Nijmegen, The Netherlands
| | - Falk Butter
- Institute of Molecular Biology (IMB), Mainz, Germany
| | - Julian König
- Institute of Molecular Biology (IMB), Mainz, Germany
| | - Michela Notarangelo
- Laboratory of Translational Genomics, Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Trento, Italy
| | - Uwe Ohler
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin Institute for Medical Systems Biology (BIMSB), Berlin, Germany.,Department of Biology, Humboldt University Berlin, Berlin, Germany
| | - Christoph Dieterich
- Klaus Tschira Institute for Integrative Computational Cardiology and Department of Internal Medicine III, University Hospital Heidelberg, Heidelberg, Germany.,German Center for Cardiovascular Research (DZHK), Partner site Heidelberg-Mannheim, Heidelberg, Germany
| | - Alessandro Quattrone
- Center for Integrative Genomics, Génopode Building, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland.,Laboratory of Translational Genomics, Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Trento, Italy
| | - Alessia Soldano
- Laboratory of Translational Genomics, Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Trento, Italy
| | - Jean-Yves Roignant
- Center for Integrative Genomics, Génopode Building, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland.,Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg-University Mainz, Mainz, Germany
| |
Collapse
|
21
|
Takarada-Iemata M, Yoshihara T, Okitani N, Iwata K, Hattori T, Ishii H, Roboon J, Nguyen DT, Fan Q, Tamatani T, Nishiuchi T, Asano M, Hori O. Abnormal social behavior and altered gene expression in mice lacking NDRG2. Neurosci Lett 2020; 743:135563. [PMID: 33359046 DOI: 10.1016/j.neulet.2020.135563] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 11/20/2020] [Accepted: 12/08/2020] [Indexed: 10/22/2022]
Abstract
N-myc downstream-regulated gene 2 (NDRG2), a member of the NDRG family, has multiple functions in cell proliferation, differentiation, and stress responses, and is predominantly expressed by astrocytes in the central nervous system. Previous studies including ours demonstrated that NDRG2 is involved in various central nervous system pathologies. However, the significance of NDRG2 in neurodevelopment is not fully understood. Here, we investigated the expression profile of NDRG2 during postnatal brain development, the role of NDRG2 in social behavior, and transcriptome changes in the brain of NDRG2-deficient mice. NDRG2 expression in the brain increased over time from postnatal day 1 to adulthood. Deletion of NDRG2 resulted in abnormal social behavior, as indicated by reduced exploratory activity toward a novel mouse in a three-chamber social interaction test. Microarray analysis identified genes differentially expressed in the NDRG2-deficient brain, and upregulated gene expression of Bmp4 and Per2 was confirmed by quantitative PCR analysis. Expression of both these genes and the encoded proteins increased over time during postnatal brain development, similar to NDRG2. Gene expression of Bmp4 and Per2 was upregulated in cultured astrocytes isolated from NDRG2-deficient mice. These results suggest that NDRG2 contributes to brain development required for proper social behavior by modulating gene expression in astrocytes.
Collapse
Affiliation(s)
- Mika Takarada-Iemata
- Department of Neuroanatomy, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Ishikawa, 920-8640, Japan.
| | - Toru Yoshihara
- Institute of Laboratory Animals, Graduate School of Medicine, Kyoto University, Sakyo-ku, Kyoto, 606-8501, Japan
| | - Nahoko Okitani
- Department of Neuroanatomy, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Ishikawa, 920-8640, Japan
| | - Keiko Iwata
- Research Center for Child Mental Development, University of Fukui, Yoshida-gun, Fukui, 910-1193, Japan
| | - Tsuyoshi Hattori
- Department of Neuroanatomy, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Ishikawa, 920-8640, Japan
| | - Hiroshi Ishii
- Department of Neuroanatomy, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Ishikawa, 920-8640, Japan
| | - Jureepon Roboon
- Department of Neuroanatomy, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Ishikawa, 920-8640, Japan
| | - Dinh Thi Nguyen
- Department of Neuroanatomy, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Ishikawa, 920-8640, Japan
| | - Qiyan Fan
- Department of Neuroanatomy, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Ishikawa, 920-8640, Japan
| | - Takashi Tamatani
- Department of Neuroanatomy, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Ishikawa, 920-8640, Japan
| | - Takumi Nishiuchi
- Institute for Gene Research, Advanced Science Research Center, Kanazawa University, Kanazawa, Ishikawa, 920-8640, Japan
| | - Masahide Asano
- Institute of Laboratory Animals, Graduate School of Medicine, Kyoto University, Sakyo-ku, Kyoto, 606-8501, Japan
| | - Osamu Hori
- Department of Neuroanatomy, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Ishikawa, 920-8640, Japan
| |
Collapse
|
22
|
Nguyen U, Tinsley B, Sen Y, Stein J, Palacios Y, Ceballos A, Welch C, Nzenkue K, Penn A, Murphy L, Leodones K, Casiquin J, Ivory I, Ghenta K, Danziger K, Widman E, Newman J, Triplehorn M, Hindi Z, Mulligan K. Exposure to bisphenol A differentially impacts neurodevelopment and behavior in Drosophila melanogaster from distinct genetic backgrounds. Neurotoxicology 2020; 82:146-157. [PMID: 33309840 DOI: 10.1016/j.neuro.2020.12.007] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 11/30/2020] [Accepted: 12/06/2020] [Indexed: 12/13/2022]
Abstract
Bisphenol A (BPA) is a ubiquitous environmental chemical that has been linked to behavioral differences in children and shown to impact critical neurodevelopmental processes in animal models. Though data is emerging, we still have an incomplete picture of how BPA disrupts neurodevelopment; in particular, how its impacts may vary across different genetic backgrounds. Given the genetic tractability of Drosophila melanogaster, they present a valuable model to address this question. Fruit flies are increasingly being used for assessment of neurotoxicants because of their relatively simple brain structure and variety of measurable behaviors. Here we investigated the neurodevelopmental impacts of BPA across two genetic strains of Drosophila-w1118 (control) and the Fragile X Syndrome (FXS) model-by examining both behavioral and neuronal phenotypes. We show that BPA induces hyperactivity in larvae, increases repetitive grooming behavior in adults, reduces courtship behavior, impairs axon guidance in the mushroom body, and disrupts neural stem cell development in the w1118 genetic strain. Remarkably, for every behavioral and neuronal phenotype examined, the impact of BPA in FXS flies was either insignificant or contrasted with the phenotypes observed in the w1118 strain. This data indicates that the neurodevelopmental impacts of BPA can vary widely depending on genetic background and suggests BPA may elicit a gene-environment interaction with Drosophila fragile X mental retardation 1 (dFmr1)-the ortholog of human FMR1, which causes Fragile X Syndrome and is associated with autism spectrum disorder.
Collapse
Affiliation(s)
- U Nguyen
- Department of Biological Sciences, California State University, Sacramento, 6000 J Street, Sacramento, CA, 95819, United States
| | - B Tinsley
- Department of Biological Sciences, California State University, Sacramento, 6000 J Street, Sacramento, CA, 95819, United States
| | - Y Sen
- Department of Biological Sciences, California State University, Sacramento, 6000 J Street, Sacramento, CA, 95819, United States
| | - J Stein
- Department of Biological Sciences, California State University, Sacramento, 6000 J Street, Sacramento, CA, 95819, United States
| | - Y Palacios
- Department of Biological Sciences, California State University, Sacramento, 6000 J Street, Sacramento, CA, 95819, United States
| | - A Ceballos
- Department of Biological Sciences, California State University, Sacramento, 6000 J Street, Sacramento, CA, 95819, United States
| | - C Welch
- Department of Biological Sciences, California State University, Sacramento, 6000 J Street, Sacramento, CA, 95819, United States
| | - K Nzenkue
- Department of Biological Sciences, California State University, Sacramento, 6000 J Street, Sacramento, CA, 95819, United States
| | - A Penn
- Department of Biological Sciences, California State University, Sacramento, 6000 J Street, Sacramento, CA, 95819, United States
| | - L Murphy
- Department of Biological Sciences, California State University, Sacramento, 6000 J Street, Sacramento, CA, 95819, United States
| | - K Leodones
- Department of Biological Sciences, California State University, Sacramento, 6000 J Street, Sacramento, CA, 95819, United States
| | - J Casiquin
- Department of Biological Sciences, California State University, Sacramento, 6000 J Street, Sacramento, CA, 95819, United States
| | - I Ivory
- Department of Biological Sciences, California State University, Sacramento, 6000 J Street, Sacramento, CA, 95819, United States
| | - K Ghenta
- Department of Biological Sciences, California State University, Sacramento, 6000 J Street, Sacramento, CA, 95819, United States
| | - K Danziger
- Department of Biological Sciences, California State University, Sacramento, 6000 J Street, Sacramento, CA, 95819, United States
| | - E Widman
- Department of Biological Sciences, California State University, Sacramento, 6000 J Street, Sacramento, CA, 95819, United States
| | - J Newman
- Department of Biological Sciences, California State University, Sacramento, 6000 J Street, Sacramento, CA, 95819, United States
| | - M Triplehorn
- Department of Biological Sciences, California State University, Sacramento, 6000 J Street, Sacramento, CA, 95819, United States
| | - Z Hindi
- Department of Biological Sciences, California State University, Sacramento, 6000 J Street, Sacramento, CA, 95819, United States
| | - K Mulligan
- Department of Biological Sciences, California State University, Sacramento, 6000 J Street, Sacramento, CA, 95819, United States.
| |
Collapse
|
23
|
Kennedy T, Rinker D, Broadie K. Genetic background mutations drive neural circuit hyperconnectivity in a fragile X syndrome model. BMC Biol 2020; 18:94. [PMID: 32731855 PMCID: PMC7392683 DOI: 10.1186/s12915-020-00817-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Accepted: 06/19/2020] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND Neural circuits are initially assembled during development when neurons synapse with potential partners and later refined as appropriate connections stabilize into mature synapses while inappropriate contacts are eliminated. Disruptions to this synaptogenic process impair connectivity optimization and can cause neurodevelopmental disorders. Intellectual disability (ID) and autism spectrum disorder (ASD) are often characterized by synaptic overgrowth, with the maintenance of immature or inappropriate synapses. Such synaptogenic defects can occur through mutation of a single gene, such as fragile X mental retardation protein (FMRP) loss causing the neurodevelopmental disorder fragile X syndrome (FXS). FXS represents the leading heritable cause of ID and ASD, but many other genes that play roles in ID and ASD have yet to be identified. RESULTS In a Drosophila FXS disease model, one dfmr150M null mutant stock exhibits previously unreported axonal overgrowths at developmental and mature stages in the giant fiber (GF) escape circuit. These excess axon projections contain both chemical and electrical synapse markers, indicating mixed synaptic connections. Extensive analyses show these supernumerary synapses connect known GF circuit neurons, rather than new, inappropriate partners, indicating hyperconnectivity within the circuit. Despite the striking similarities to well-characterized FXS synaptic defects, this new GF circuit hyperconnectivity phenotype is driven by genetic background mutations in this dfmr150M stock. Similar GF circuit synaptic overgrowth is not observed in independent dfmr1 null alleles. Bulked segregant analysis (BSA) was combined with whole genome sequencing (WGS) to identify the quantitative trait loci (QTL) linked to neural circuit hyperconnectivity. The results reveal 8 QTL associated with inappropriate synapse formation and maintenance in the dfmr150M mutant background. CONCLUSIONS Synaptogenesis is a complex, precisely orchestrated neurodevelopmental process with a large cohort of gene products coordinating the connectivity, synaptic strength, and excitatory/inhibitory balance between neuronal partners. This work identifies a number of genetic regions that contain mutations disrupting proper synaptogenesis within a particularly well-mapped neural circuit. These QTL regions contain potential new genes involved in synapse formation and refinement. Given the similarity of the synaptic overgrowth phenotype to known ID and ASD inherited conditions, identifying these genes should increase our understanding of these devastating neurodevelopmental disease states.
Collapse
Affiliation(s)
- Tyler Kennedy
- Department of Biological Sciences, Vanderbilt University and Medical Center, Nashville, TN, 37235, USA
| | - David Rinker
- Department of Biological Sciences, Vanderbilt University and Medical Center, Nashville, TN, 37235, USA
| | - Kendal Broadie
- Department of Biological Sciences, Vanderbilt University and Medical Center, Nashville, TN, 37235, USA.
- Department of Cell and Developmental Biology, Vanderbilt University and Medical Center, Nashville, TN, 37235, USA.
- Vanderbilt Brain Institute, Vanderbilt University and Medical Center, Nashville, TN, 37235, USA.
| |
Collapse
|
24
|
Lessons from LIMK1 enzymology and their impact on inhibitor design. Biochem J 2020; 476:3197-3209. [PMID: 31652302 PMCID: PMC6835155 DOI: 10.1042/bcj20190517] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Revised: 10/07/2019] [Accepted: 10/11/2019] [Indexed: 12/14/2022]
Abstract
LIM domain kinase 1 (LIMK1) is a key regulator of actin dynamics. It is thereby a potential therapeutic target for the prevention of fragile X syndrome and amyotrophic lateral sclerosis. Herein, we use X-ray crystallography and activity assays to describe how LIMK1 accomplishes substrate specificity, to suggest a unique ‘rock-and-poke’ mechanism of catalysis and to explore the regulation of the kinase by activation loop phosphorylation. Based on these findings, a differential scanning fluorimetry assay and a RapidFire mass spectrometry activity assay were established, leading to the discovery and confirmation of a set of small-molecule LIMK1 inhibitors. Interestingly, several of the inhibitors were inactive towards the closely related isoform LIMK2. Finally, crystal structures of the LIMK1 kinase domain in complex with inhibitors (PF-477736 and staurosporine, respectively) are presented, providing insights into LIMK1 plasticity upon inhibitor binding.
Collapse
|
25
|
Ueoka I, Pham HTN, Matsumoto K, Yamaguchi M. Autism Spectrum Disorder-Related Syndromes: Modeling with Drosophila and Rodents. Int J Mol Sci 2019; 20:E4071. [PMID: 31438473 PMCID: PMC6747505 DOI: 10.3390/ijms20174071] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2019] [Revised: 08/17/2019] [Accepted: 08/18/2019] [Indexed: 12/11/2022] Open
Abstract
Whole exome analyses have identified a number of genes associated with autism spectrum disorder (ASD) and ASD-related syndromes. These genes encode key regulators of synaptogenesis, synaptic plasticity, cytoskeleton dynamics, protein synthesis and degradation, chromatin remodeling, transcription, and lipid homeostasis. Furthermore, in silico studies suggest complex regulatory networks among these genes. Drosophila is a useful genetic model system for studies of ASD and ASD-related syndromes to clarify the in vivo roles of ASD-associated genes and the complex gene regulatory networks operating in the pathogenesis of ASD and ASD-related syndromes. In this review, we discuss what we have learned from studies with vertebrate models, mostly mouse models. We then highlight studies with Drosophila models. We also discuss future developments in the related field.
Collapse
Affiliation(s)
- Ibuki Ueoka
- Department of Applied Biology, Kyoto Institute of Technology, Matsugasaki, Sakyo-ku, Kyoto 603-8585, Japan
| | - Hang Thi Nguyet Pham
- Department of Pharmacology and Biochemistry, National Institute of Medicinal Materials, Hanoi 110100, Vietnam
| | - Kinzo Matsumoto
- Division of Medicinal Pharmacology, Institute of Natural Medicine, University of Toyama, Toyama 930-0194, Japan
| | - Masamitsu Yamaguchi
- Department of Applied Biology, Kyoto Institute of Technology, Matsugasaki, Sakyo-ku, Kyoto 603-8585, Japan.
| |
Collapse
|
26
|
Sears JC, Choi WJ, Broadie K. Fragile X Mental Retardation Protein positively regulates PKA anchor Rugose and PKA activity to control actin assembly in learning/memory circuitry. Neurobiol Dis 2019; 127:53-64. [PMID: 30771457 DOI: 10.1016/j.nbd.2019.02.004] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2018] [Accepted: 02/04/2019] [Indexed: 01/09/2023] Open
Abstract
Recent work shows Fragile X Mental Retardation Protein (FMRP) drives the translation of very large proteins (>2000 aa) mediating neurodevelopment. Loss of function results in Fragile X syndrome (FXS), the leading heritable cause of intellectual disability (ID) and autism spectrum disorder (ASD). Using the Drosophila FXS disease model, we discover FMRP positively regulates the translation of the very large A-Kinase Anchor Protein (AKAP) Rugose (>3000 aa), homolog of ASD-associated human Neurobeachin (NBEA). In the central brain Mushroom Body (MB) circuit, where Protein Kinase A (PKA) signaling is necessary for learning/memory, FMRP loss reduces Rugose levels and targeted FMRP overexpression elevates Rugose levels. Using a new in vivo transgenic PKA activity reporter (PKA-SPARK), we find FMRP loss reduces PKA activity in MB Kenyon cells whereas FMRP overexpression elevates PKA activity. Consistently, loss of Rugose reduces PKA activity, but Rugose overexpression has no independent effect. A well-established PKA output is regulation of F-actin cytoskeleton dynamics. In the FXS disease model, F-actin is aberrantly accumulated in MB lobes and single MB Kenyon cells. Consistently, Rugose loss results in similar F-actin accumulation. Moreover, targeted FMRP, Rugose and PKA overexpression all result in increased F-actin accumulation in the MB circuit. These findings uncover a FMRP-Rugose-PKA mechanism regulating actin cytoskeleton. This study reveals a novel FMRP mechanism controlling neuronal PKA activity, and demonstrates a shared mechanistic connection between FXS and NBEA associated ASD disease states, with a common link to PKA and F-actin misregulation in brain neural circuits. SIGNIFICANCE STATEMENT: Autism spectrum disorder (ASD) arises from a wide array of genetic lesions, and it is therefore critical to identify common underlying molecular mechanisms. Here, we link two ASD states; Neurobeachin (NBEA) associated ASD and Fragile X syndrome (FXS), the most common inherited ASD. Using established Drosophila disease models, we find Fragile X Mental Retardation Protein (FMRP) positively regulates translation of NBEA homolog Rugose, consistent with a recent advance showing FMRP promotes translation of very large proteins associated with ASD. FXS exhibits reduced cAMP induction, a potent activator of PKA, and Rugose/NBEA is a PKA anchor. Consistently, we find brain PKA activity strikingly reduced in both ASD models. We discover this pathway regulation controls actin cytoskeleton dynamics in brain neural circuits.
Collapse
Affiliation(s)
- James C Sears
- Vanderbilt Brain Institute, Departments of Biological Sciences, Vanderbilt University and Medical Center, Nashville, TN 37235, USA
| | - Woong Jae Choi
- Departments of Biological Sciences, Vanderbilt University and Medical Center, Nashville, TN 37235, USA
| | - Kendal Broadie
- Vanderbilt Brain Institute, Departments of Biological Sciences, Cell and Developmental Biology, and Pharmacology, Vanderbilt University and Medical Center, Nashville, TN 37235, USA.
| |
Collapse
|
27
|
Olesnicky EC, Wright EG. Drosophila as a Model for Assessing the Function of RNA-Binding Proteins during Neurogenesis and Neurological Disease. J Dev Biol 2018; 6:E21. [PMID: 30126171 PMCID: PMC6162566 DOI: 10.3390/jdb6030021] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Revised: 08/15/2018] [Accepted: 08/15/2018] [Indexed: 12/16/2022] Open
Abstract
An outstanding question in developmental neurobiology is how RNA processing events contribute to the regulation of neurogenesis. RNA processing events are increasingly recognized as playing fundamental roles in regulating multiple developmental events during neurogenesis, from the asymmetric divisions of neural stem cells, to the generation of complex and diverse neurite morphologies. Indeed, both asymmetric cell division and neurite morphogenesis are often achieved by mechanisms that generate asymmetric protein distributions, including post-transcriptional gene regulatory mechanisms such as the transport of translationally silent messenger RNAs (mRNAs) and local translation of mRNAs within neurites. Additionally, defects in RNA splicing have emerged as a common theme in many neurodegenerative disorders, highlighting the importance of RNA processing in maintaining neuronal circuitry. RNA-binding proteins (RBPs) play an integral role in splicing and post-transcriptional gene regulation, and mutations in RBPs have been linked with multiple neurological disorders including autism, dementia, amyotrophic lateral sclerosis (ALS), spinal muscular atrophy (SMA), Fragile X syndrome (FXS), and X-linked intellectual disability disorder. Despite their widespread nature and roles in neurological disease, the molecular mechanisms and networks of regulated target RNAs have been defined for only a small number of specific RBPs. This review aims to highlight recent studies in Drosophila that have advanced our knowledge of how RBP dysfunction contributes to neurological disease.
Collapse
Affiliation(s)
- Eugenia C Olesnicky
- Department of Biology, University of Colorado Colorado Springs, 1420 Austin Bluffs Parkway, Colorado Springs, CO 80918, USA.
| | - Ethan G Wright
- Department of Biology, University of Colorado Colorado Springs, 1420 Austin Bluffs Parkway, Colorado Springs, CO 80918, USA.
| |
Collapse
|
28
|
Sunamura N, Iwashita S, Enomoto K, Kadoshima T, Isono F. Loss of the fragile X mental retardation protein causes aberrant differentiation in human neural progenitor cells. Sci Rep 2018; 8:11585. [PMID: 30072797 PMCID: PMC6072755 DOI: 10.1038/s41598-018-30025-4] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Accepted: 07/20/2018] [Indexed: 12/20/2022] Open
Abstract
Fragile X syndrome (FXS) is caused by transcriptional silencing of the FMR1 gene during embryonic development with the consequent loss of the encoded fragile X mental retardation protein (FMRP). The pathological mechanisms of FXS have been extensively studied using the Fmr1-knockout mouse, and the findings suggest important roles for FMRP in synaptic plasticity and proper functioning of neural networks. However, the function of FMRP during early development in the human nervous system remains to be confirmed. Here we describe human neural progenitor cells (NPCs) as a model for studying FMRP functions and FXS pathology. Transcriptome analysis of the NPCs derived from FMR1-knockout human induced pluripotent stem cells (iPSCs) showed altered expression of neural differentiation markers, particularly a marked induction of the astrocyte marker glial fibrillary acidic protein (GFAP). When induced to differentiate, FMRP-deficient neurons continued to express GFAP, and showed less spontaneous calcium bursts than the parental iPSC-derived neurons. Interestingly, the aberrant expression of GFAP and the impaired firing was corrected by treatment with the protein kinase inhibitor LX7101. These findings underscore the modulatory roles of FMRP in human neurogenesis, and further demonstrate that the defective phenotype of FXS could be reversed at least partly by small molecule kinase inhibitors.
Collapse
Affiliation(s)
- Naohiro Sunamura
- Asubio Pharma Co., Ltd., 6-4-3 Minatojima-minamimachi, Chuo-ku, Kobe, 650-0047, Japan
| | - Shinzo Iwashita
- Asubio Pharma Co., Ltd., 6-4-3 Minatojima-minamimachi, Chuo-ku, Kobe, 650-0047, Japan
| | - Kei Enomoto
- Asubio Pharma Co., Ltd., 6-4-3 Minatojima-minamimachi, Chuo-ku, Kobe, 650-0047, Japan
| | - Taisuke Kadoshima
- Asubio Pharma Co., Ltd., 6-4-3 Minatojima-minamimachi, Chuo-ku, Kobe, 650-0047, Japan.
| | - Fujio Isono
- Asubio Pharma Co., Ltd., 6-4-3 Minatojima-minamimachi, Chuo-ku, Kobe, 650-0047, Japan.
| |
Collapse
|
29
|
Drozd M, Bardoni B, Capovilla M. Modeling Fragile X Syndrome in Drosophila. Front Mol Neurosci 2018; 11:124. [PMID: 29713264 PMCID: PMC5911982 DOI: 10.3389/fnmol.2018.00124] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Accepted: 03/29/2018] [Indexed: 01/18/2023] Open
Abstract
Intellectual disability (ID) and autism are hallmarks of Fragile X Syndrome (FXS), a hereditary neurodevelopmental disorder. The gene responsible for FXS is Fragile X Mental Retardation gene 1 (FMR1) encoding the Fragile X Mental Retardation Protein (FMRP), an RNA-binding protein involved in RNA metabolism and modulating the expression level of many targets. Most cases of FXS are caused by silencing of FMR1 due to CGG expansions in the 5'-UTR of the gene. Humans also carry the FXR1 and FXR2 paralogs of FMR1 while flies have only one FMR1 gene, here called dFMR1, sharing the same level of sequence homology with all three human genes, but functionally most similar to FMR1. This enables a much easier approach for FMR1 genetic studies. Drosophila has been widely used to investigate FMR1 functions at genetic, cellular, and molecular levels since dFMR1 mutants have many phenotypes in common with the wide spectrum of FMR1 functions that underlay the disease. In this review, we present very recent Drosophila studies investigating FMRP functions at genetic, cellular, molecular, and electrophysiological levels in addition to research on pharmacological treatments in the fly model. These studies have the potential to aid the discovery of pharmacological therapies for FXS.
Collapse
Affiliation(s)
- Małgorzata Drozd
- Université Côte d'Azur, CNRS, IPMC, Valbonne, France.,CNRS LIA (Neogenex), Valbonne, France
| | - Barbara Bardoni
- CNRS LIA (Neogenex), Valbonne, France.,Université Côte d'Azur, INSERM, CNRS, IPMC, Valbonne, France
| | - Maria Capovilla
- Université Côte d'Azur, CNRS, IPMC, Valbonne, France.,CNRS LIA (Neogenex), Valbonne, France
| |
Collapse
|
30
|
Vasta JD, Corona CR, Wilkinson J, Zimprich CA, Hartnett JR, Ingold MR, Zimmerman K, Machleidt T, Kirkland TA, Huwiler KG, Ohana RF, Slater M, Otto P, Cong M, Wells CI, Berger BT, Hanke T, Glas C, Ding K, Drewry DH, Huber KVM, Willson TM, Knapp S, Müller S, Meisenheimer PL, Fan F, Wood KV, Robers MB. Quantitative, Wide-Spectrum Kinase Profiling in Live Cells for Assessing the Effect of Cellular ATP on Target Engagement. Cell Chem Biol 2018; 25:206-214.e11. [PMID: 29174542 PMCID: PMC5814754 DOI: 10.1016/j.chembiol.2017.10.010] [Citation(s) in RCA: 189] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Revised: 08/24/2017] [Accepted: 10/24/2017] [Indexed: 02/03/2023]
Abstract
For kinase inhibitors, intracellular target selectivity is fundamental to pharmacological mechanism. Although a number of acellular techniques have been developed to measure kinase binding or enzymatic inhibition, such approaches can fail to accurately predict engagement in cells. Here we report the application of an energy transfer technique that enabled the first broad-spectrum, equilibrium-based approach to quantitatively profile target occupancy and compound affinity in live cells. Using this method, we performed a selectivity profiling for clinically relevant kinase inhibitors against 178 full-length kinases, and a mechanistic interrogation of the potency offsets observed between cellular and biochemical analysis. For the multikinase inhibitor crizotinib, our approach accurately predicted cellular potency and revealed improved target selectivity compared with biochemical measurements. Due to cellular ATP, a number of putative crizotinib targets are unexpectedly disengaged in live cells at a clinically relevant drug dose.
Collapse
Affiliation(s)
- James D Vasta
- Promega Corporation, 2800 Woods Hollow Road, Fitchburg, WI 53711, USA
| | - Cesear R Corona
- Promega Biosciences Incorporated, 277 Granada Drive, San Luis Obispo, CA 93401, USA
| | | | - Chad A Zimprich
- Promega Corporation, 2800 Woods Hollow Road, Fitchburg, WI 53711, USA
| | - James R Hartnett
- Promega Corporation, 2800 Woods Hollow Road, Fitchburg, WI 53711, USA
| | - Morgan R Ingold
- Promega Corporation, 2800 Woods Hollow Road, Fitchburg, WI 53711, USA
| | | | - Thomas Machleidt
- Promega Corporation, 2800 Woods Hollow Road, Fitchburg, WI 53711, USA
| | - Thomas A Kirkland
- Promega Biosciences Incorporated, 277 Granada Drive, San Luis Obispo, CA 93401, USA
| | - Kristin G Huwiler
- Promega Corporation, 2800 Woods Hollow Road, Fitchburg, WI 53711, USA
| | | | - Michael Slater
- Promega Corporation, 2800 Woods Hollow Road, Fitchburg, WI 53711, USA
| | - Paul Otto
- Promega Corporation, 2800 Woods Hollow Road, Fitchburg, WI 53711, USA
| | - Mei Cong
- Promega Corporation, 2800 Woods Hollow Road, Fitchburg, WI 53711, USA
| | - Carrow I Wells
- Structural Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Benedict-Tilman Berger
- Structural Genomics Consortium, Institute for Pharmaceutical Chemistry, Johann Wolfgang Goethe-University, Max-von-Laue-Straße 9, 60438 Frankfurt am Main, Germany; Structural Genomics Consortium, Buchmann Institute for Molecular Life Sciences, Johann Wolfgang Goethe-University, Max-von-Laue-Straße 15, 60438 Frankfurt am Main, Germany
| | - Thomas Hanke
- Structural Genomics Consortium, Institute for Pharmaceutical Chemistry, Johann Wolfgang Goethe-University, Max-von-Laue-Straße 9, 60438 Frankfurt am Main, Germany
| | - Carina Glas
- Structural Genomics Consortium, Nuffield Department of Medicine, University of Oxford, Oxford, UK; Target Discovery Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Ke Ding
- State Key Laboratory of Respiratory Diseases, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 190 Kaiyuan Avenue, Guangzhou 510530, China; School of Pharmacy, Jinan University, 601 Huangpu Avenue West, Guangzhou 510632, China
| | - David H Drewry
- Structural Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Kilian V M Huber
- Structural Genomics Consortium, Nuffield Department of Medicine, University of Oxford, Oxford, UK; Target Discovery Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Timothy M Willson
- Structural Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Stefan Knapp
- Structural Genomics Consortium, Institute for Pharmaceutical Chemistry, Johann Wolfgang Goethe-University, Max-von-Laue-Straße 9, 60438 Frankfurt am Main, Germany; Structural Genomics Consortium, Buchmann Institute for Molecular Life Sciences, Johann Wolfgang Goethe-University, Max-von-Laue-Straße 15, 60438 Frankfurt am Main, Germany
| | - Susanne Müller
- Structural Genomics Consortium, Buchmann Institute for Molecular Life Sciences, Johann Wolfgang Goethe-University, Max-von-Laue-Straße 15, 60438 Frankfurt am Main, Germany
| | | | - Frank Fan
- Promega Corporation, 2800 Woods Hollow Road, Fitchburg, WI 53711, USA
| | - Keith V Wood
- Promega Corporation, 2800 Woods Hollow Road, Fitchburg, WI 53711, USA
| | - Matthew B Robers
- Promega Corporation, 2800 Woods Hollow Road, Fitchburg, WI 53711, USA.
| |
Collapse
|
31
|
Kashima R, Hata A. The role of TGF-β superfamily signaling in neurological disorders. Acta Biochim Biophys Sin (Shanghai) 2018; 50:106-120. [PMID: 29190314 PMCID: PMC5846707 DOI: 10.1093/abbs/gmx124] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Revised: 11/02/2017] [Indexed: 12/12/2022] Open
Abstract
The TGF-β superfamily signaling is involved in a variety of biological processes during embryogenesis and in adult tissue homeostasis. Faulty regulation of the signaling pathway that transduces the TGF-β superfamily signals accordingly leads to a number of ailments, such as cancer and cardiovascular, metabolic, urinary, intestinal, skeletal, and immune diseases. In recent years, a number of studies have elucidated the essential roles of TGF-βs and BMPs during neuronal development in the maintenance of appropriate innervation and neuronal activity. The new advancement implicates significant roles of the aberrant TGF-β superfamily signaling in the pathogenesis of neurological disorders. In this review, we compile a number of reports implicating the deregulation of TGF-β/BMP signaling pathways in the pathogenesis of cognitive and neurodegenerative disorders in animal models and patients. We apologize in advance that the review falls short of providing details of the role of TGF-β/BMP signaling or mechanisms underlying the pathogenesis of neurological disorders. The goal of this article is to reveal a gap in our knowledge regarding the association between TGF-β/BMP signaling pathways and neuronal tissue homeostasis and development and facilitate the research with a potential to develop new therapies for neurological ailments by modulating the pathways.
Collapse
Affiliation(s)
- Risa Kashima
- Cardiovascular Research Institute, University of California, San Francisco, CA 94143, USA
| | - Akiko Hata
- Cardiovascular Research Institute, University of California, San Francisco, CA 94143, USA
- Department of Biochemistry and Biophysics, University of California, San Francisco, CA 94143, USA
| |
Collapse
|
32
|
Pyronneau A, He Q, Hwang JY, Porch M, Contractor A, Zukin RS. Aberrant Rac1-cofilin signaling mediates defects in dendritic spines, synaptic function, and sensory perception in fragile X syndrome. Sci Signal 2017; 10:10/504/eaan0852. [PMID: 29114038 DOI: 10.1126/scisignal.aan0852] [Citation(s) in RCA: 101] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Fragile X syndrome (FXS) is the most common inherited cause of intellectual disabilities and a leading cause of autism. FXS is caused by a trinucleotide expansion in the gene FMR1 on the X chromosome. The neuroanatomical hallmark of FXS is an overabundance of immature dendritic spines, a factor thought to underlie synaptic dysfunction and impaired cognition. We showed that aberrantly increased activity of the Rho GTPase Rac1 inhibited the actin-depolymerizing factor cofilin, a major determinant of dendritic spine structure, and caused disease-associated spine abnormalities in the somatosensory cortex of FXS model mice. Increased cofilin phosphorylation and actin polymerization coincided with abnormal dendritic spines and impaired synaptic maturation. Viral delivery of a constitutively active cofilin mutant (cofilinS3A) into the somatosensory cortex of Fmr1-deficient mice rescued the immature dendritic spine phenotype and increased spine density. Inhibition of the Rac1 effector PAK1 with a small-molecule inhibitor rescued cofilin signaling in FXS mice, indicating a causal relationship between PAK1 and cofilin signaling. PAK1 inhibition rescued synaptic signaling (specifically the synaptic ratio of NMDA/AMPA in layer V pyramidal neurons) and improved sensory processing in FXS mice. These findings suggest a causal relationship between increased Rac1-cofilin signaling, synaptic defects, and impaired sensory processing in FXS and uncover a previously unappreciated role for impaired Rac1-cofilin signaling in the aberrant spine morphology and spine density associated with FXS.
Collapse
Affiliation(s)
- Alexander Pyronneau
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, New York, NY 10461, USA
| | - Qionger He
- Department of Physiology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Jee-Yeon Hwang
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, New York, NY 10461, USA
| | - Morgan Porch
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, New York, NY 10461, USA
| | - Anis Contractor
- Department of Physiology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA.,Department of Neurobiology, Weinberg College of Arts and Sciences, Northwestern University, Evanston, IL 60208, USA
| | - R Suzanne Zukin
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, New York, NY 10461, USA.
| |
Collapse
|
33
|
Abstract
In this issue of
Science Signaling
, three papers uncover molecular mechanisms of neurological dysfunction in animal models of fragile X syndrome.
Collapse
|
34
|
Davis JK, Broadie K. Multifarious Functions of the Fragile X Mental Retardation Protein. Trends Genet 2017; 33:703-714. [PMID: 28826631 PMCID: PMC5610095 DOI: 10.1016/j.tig.2017.07.008] [Citation(s) in RCA: 87] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Revised: 07/20/2017] [Accepted: 07/24/2017] [Indexed: 01/01/2023]
Abstract
Fragile X syndrome (FXS), a heritable intellectual and autism spectrum disorder (ASD), results from the loss of Fragile X mental retardation protein (FMRP). This neurodevelopmental disease state exhibits neural circuit hyperconnectivity and hyperexcitability. Canonically, FMRP functions as an mRNA-binding translation suppressor, but recent findings have enormously expanded its proposed roles. Although connections between burgeoning FMRP functions remain unknown, recent advances have extended understanding of its involvement in RNA, channel, and protein binding that modulate calcium signaling, activity-dependent critical period development, and the excitation-inhibition (E/I) neural circuitry balance. In this review, we contextualize 3 years of FXS model research. Future directions extrapolated from recent advances focus on discovering links between FMRP roles to determine whether FMRP has a multitude of unrelated functions or whether combinatorial mechanisms can explain its multifaceted existence.
Collapse
Affiliation(s)
- Jenna K Davis
- Department of Biological Sciences, Kennedy Center for Research on Human Development, Vanderbilt University, Nashville, TN 37235, USA
| | - Kendal Broadie
- Department of Biological Sciences, Kennedy Center for Research on Human Development, Vanderbilt University, Nashville, TN 37235, USA.
| |
Collapse
|
35
|
Ferrarelli LK. New connections: Treating fragile X syndrome. Sci Signal 2017; 10:10/477/eaan5441. [DOI: 10.1126/scisignal.aan5441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
An in vivo drug screen can identify inhibitors of the signaling pathway that is activated in fragile X syndrome.
Collapse
|