1
|
Petshow S, Coblentz A, Hamilton AM, Sarkar D, Anisimova M, Flores JC, Zito K. Activity-dependent regulation of Cdc42 by Ephexin5 drives synapse growth and stabilization. SCIENCE ADVANCES 2025; 11:eadp5782. [PMID: 40138406 PMCID: PMC11939064 DOI: 10.1126/sciadv.adp5782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Accepted: 02/19/2025] [Indexed: 03/29/2025]
Abstract
Synaptic Rho guanosine triphosphatase (GTPase) guanine nucleotide exchange factors (RhoGEFs) play vital roles in regulating the activity-dependent neuronal plasticity that is critical for learning. Ephexin5, a RhoGEF implicated in the etiology of Alzheimer's disease and Angelman syndrome, was originally reported in neurons as a RhoA-specific GEF that negatively regulates spine synapse density. Here, we show that Ephexin5 activates both RhoA and Cdc42 in the brain. Furthermore, using live imaging of GTPase biosensors, we demonstrate that Ephexin5 regulates activity-dependent Cdc42, but not RhoA, signaling at single synapses. The selectivity of Ephexin5 for Cdc42 activation is regulated by tyrosine phosphorylation, which is regulated by neuronal activity. Last, in contrast to Ephexin5's role in negatively regulating synapse density, we show that, downstream of neuronal activity, Ephexin5 positively regulates synaptic growth and stabilization. Our results support a model in which plasticity-inducing neuronal activity regulates Ephexin5 tyrosine phosphorylation, driving Ephexin5-mediated activation of Cdc42 and the spine structural growth and stabilization vital for learning.
Collapse
Affiliation(s)
- Samuel Petshow
- Center for Neuroscience, University of California, Davis, Davis, CA 95618, USA
| | - Azariah Coblentz
- Center for Neuroscience, University of California, Davis, Davis, CA 95618, USA
| | - Andrew M. Hamilton
- Center for Neuroscience, University of California, Davis, Davis, CA 95618, USA
| | - Dipannita Sarkar
- Center for Neuroscience, University of California, Davis, Davis, CA 95618, USA
| | - Margarita Anisimova
- Center for Neuroscience, University of California, Davis, Davis, CA 95618, USA
| | - Juan C. Flores
- Center for Neuroscience, University of California, Davis, Davis, CA 95618, USA
| | - Karen Zito
- Center for Neuroscience, University of California, Davis, Davis, CA 95618, USA
| |
Collapse
|
2
|
Zelek-Molik A, Gądek-Michalska A, Wilczkowski M, Bielawski A, Maziarz K, Kreiner G, Nalepa I. Restraint stress effects on glutamate signaling protein levels in the rats' frontal cortex: Does β1 adrenoceptor activity matter? Front Pharmacol 2025; 15:1451895. [PMID: 39834820 PMCID: PMC11743458 DOI: 10.3389/fphar.2024.1451895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 12/04/2024] [Indexed: 01/22/2025] Open
Abstract
Introduction Stress-evoked dysfunctions of the frontal cortex (FC) are correlated with changes in the functioning of the glutamatergic system, and evidence demonstrates that noradrenergic transmission is an important regulator of this process. In the current study, we adopted a restraint stress (RS) model in male Wistar rats to investigate whether the blockade of β1 adrenergic receptors (β1AR) with betaxolol (BET) in stressed animals influences the body's stress response and the expression of selected signaling proteins in the medial prefrontal cortex (mPFC). Methods The study was divided into two parts. In the first part, rats were exposed to RS for 3, 7, or 14 days, and the expression of glutamate signaling proteins (p(S845)/t GluA1, p(Y1472)/t GluN2B, VGLUT1, and VGLUT2) in the FC was analyzed to determine the optimal RS duration for studying the mechanisms of hypofrontality. In the second part, rats were exposed to RS for 14 days, and BET (5 mg/kg, p. o.) was administered during the last 8 days immediately after RS. The body's stress reaction was assessed by analyzing body weight and blood levels of adrenocorticotropic hormone (ACTH) and corticosterone (CORT). Behavioral responses were evaluated using the novel object recognition (NOR) and elevated plus maze (EPM) tests. The impact of RS and BET on the expression of p(Y530)/t Fyn and p (S133)/t CREB in the mPFC was measured via Western blotting. Results and Discussion The first part of the study demonstrated a decreased level of glutamate receptors in rats exposed to 14 days of RS, following an initial increase observed after 7 days of RS. Results from the second part revealed that chronic RS reduced body weight, impaired recognition memory in the NOR test, augmented blood levels of ACTH, and increased the expression of p(Y530) Fyn in the mPFC. However, β1AR blockade did not alter the effects of RS on weight gain, cognitive function, or the expression of p(Y530) Fyn. β1AR blockade normalized only the blood concentration of ACTH. These results suggest that decreased Fyn kinase activity, indicated by phosphorylation at Y530, underlies the stress-evoked downregulation of GluN2B in the FC in a manner independent of β1AR activity.
Collapse
Affiliation(s)
- Agnieszka Zelek-Molik
- Department of Brain Biochemistry, Maj Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland
| | - Anna Gądek-Michalska
- Department of Physiology, Maj Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland
| | - Michał Wilczkowski
- Department of Brain Biochemistry, Maj Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland
| | - Adam Bielawski
- Department of Brain Biochemistry, Maj Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland
| | - Katarzyna Maziarz
- Department of Brain Biochemistry, Maj Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland
| | - Grzegorz Kreiner
- Department of Brain Biochemistry, Maj Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland
| | - Irena Nalepa
- Department of Brain Biochemistry, Maj Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland
| |
Collapse
|
3
|
Walters JM, Noblet HA, Chung HJ. An emerging role of STriatal-Enriched protein tyrosine Phosphatase in hyperexcitability-associated brain disorders. Neurobiol Dis 2024; 200:106641. [PMID: 39159894 DOI: 10.1016/j.nbd.2024.106641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Revised: 08/15/2024] [Accepted: 08/16/2024] [Indexed: 08/21/2024] Open
Abstract
STriatal-Enriched protein tyrosine Phosphatase (STEP) is a brain-specific tyrosine phosphatase that is associated with numerous neurological and neuropsychiatric disorders. STEP dephosphorylates and inactivates various kinases and phosphatases critical for neuronal function and health including Fyn, Pyk2, ERK1/2, p38, and PTPα. Importantly, STEP dephosphorylates NMDA and AMPA receptors, two major glutamate receptors that mediate fast excitatory synaptic transmission. This STEP-mediated dephosphorylation leads to their internalization and inhibits both Hebbian synaptic potentiation and homeostatic synaptic scaling. Hence, STEP has been widely accepted to weaken excitatory synaptic strength. However, emerging evidence implicates a novel role of STEP in neuronal hyperexcitability and seizure disorders. Genetic deletion and pharmacological blockade of STEP reduces seizure susceptibility in acute seizure mouse models and audiogenic seizures in a mouse model of Fragile X syndrome. Pharmacologic inhibition of STEP also decreases hippocampal activity and neuronal intrinsic excitability. Here, we will highlight the divergent roles of STEP in excitatory synaptic transmission and neuronal intrinsic excitability, present the potential underlying mechanisms, and discuss their impact on STEP-associated neurologic and neuropsychiatric disorders.
Collapse
Affiliation(s)
- Jennifer M Walters
- Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, IL, USA; Dept. of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Hayden A Noblet
- Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, IL, USA; Dept. of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Hee Jung Chung
- Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, IL, USA; Dept. of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL, USA; Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL, USA; Institute of Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL, USA.
| |
Collapse
|
4
|
Li H, Rajani V, Sengar AS, Salter MW. Src dependency of the regulation of LTP by alternative splicing of GRIN1 exon 5. Philos Trans R Soc Lond B Biol Sci 2024; 379:20230236. [PMID: 38853562 PMCID: PMC11343231 DOI: 10.1098/rstb.2023.0236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 01/31/2024] [Accepted: 02/11/2024] [Indexed: 06/11/2024] Open
Abstract
Alternative splicing of Grin1 exon 5 regulates induction of long-term potentiation (LTP) at Schaffer collateral-CA1 synapses: LTP in mice lacking the GluN1 exon 5-encoded N1 cassette (GluN1a mice) is significantly increased compared with that in mice compulsorily expressing this exon (GluN1b mice). The mechanism underlying this difference is unknown. Here, we report that blocking the non-receptor tyrosine kinase Src prevents induction of LTP in GluN1a mice but not in GluN1b. We find that activating Src enhances pharmacologically isolated synaptic N-methyl-d-aspartate receptor (NMDAR) currents in GluN1a mice but not in GluN1b. Moreover, we observe that Src activation increases the α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor component of Schaffer collateral-evoked excitatory post-synaptic potentials in GluN1a mice, but this increase is prevented by blocking NMDARs. We conclude that at these synapses, NMDARs in GluN1a mice are subject to upregulation by Src that mediates induction of LTP, whereas NMDARs in GluN1b mice are not regulated by Src, leading to Src-resistance of LTP. Thus, we have uncovered that a key regulatory mechanism for synaptic potentiation is gated by differential splicing of exon 5 of Grin1. This article is part of a discussion meeting issue 'Long-term potentiation: 50 years on'.
Collapse
Affiliation(s)
- Hongbin Li
- Program in Neurosciences & Mental Health, The Hospital for Sick Children, Toronto, ONM5G 1X8, Canada
| | - Vishaal Rajani
- Program in Neurosciences & Mental Health, The Hospital for Sick Children, Toronto, ONM5G 1X8, Canada
| | - Ameet S. Sengar
- Program in Neurosciences & Mental Health, The Hospital for Sick Children, Toronto, ONM5G 1X8, Canada
| | - Michael W. Salter
- Program in Neurosciences & Mental Health, The Hospital for Sick Children, Toronto, ONM5G 1X8, Canada
- Department of Physiology, University of Toronto, Toronto, ONM5S 1A8, Canada
| |
Collapse
|
5
|
Lee S, Kim J, Ryu HH, Jang H, Lee D, Lee S, Song JM, Lee YS, Ho Suh Y. SHP2 regulates GluA2 tyrosine phosphorylation required for AMPA receptor endocytosis and mGluR-LTD. Proc Natl Acad Sci U S A 2024; 121:e2316819121. [PMID: 38657042 PMCID: PMC11066993 DOI: 10.1073/pnas.2316819121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Accepted: 03/29/2024] [Indexed: 04/26/2024] Open
Abstract
Posttranslational modifications regulate the properties and abundance of synaptic α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors that mediate fast excitatory synaptic transmission and synaptic plasticity in the central nervous system. During long-term depression (LTD), protein tyrosine phosphatases (PTPs) dephosphorylate tyrosine residues in the C-terminal tail of AMPA receptor GluA2 subunit, which is essential for GluA2 endocytosis and group I metabotropic glutamate receptor (mGluR)-dependent LTD. However, as a selective downstream effector of mGluRs, the mGluR-dependent PTP responsible for GluA2 tyrosine dephosphorylation remains elusive at Schaffer collateral (SC)-CA1 synapses. In the present study, we find that mGluR5 stimulation activates Src homology 2 (SH2) domain-containing phosphatase 2 (SHP2) by increasing phospho-Y542 levels in SHP2. Under steady-state conditions, SHP2 plays a protective role in stabilizing phospho-Y869 of GluA2 by directly interacting with GluA2 phosphorylated at Y869, without affecting GluA2 phospho-Y876 levels. Upon mGluR5 stimulation, SHP2 dephosphorylates GluA2 at Y869 and Y876, resulting in GluA2 endocytosis and mGluR-LTD. Our results establish SHP2 as a downstream effector of mGluR5 and indicate a dual action of SHP2 in regulating GluA2 tyrosine phosphorylation and function. Given the implications of mGluR5 and SHP2 in synaptic pathophysiology, we propose SHP2 as a promising therapeutic target for neurodevelopmental and autism spectrum disorders.
Collapse
Affiliation(s)
- Sanghyeon Lee
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul03080, South Korea
- Neuroscience Research Institute, Medical Research Center, Seoul National University College of Medicine, Seoul03080, South Korea
- Transplantation Research Institute, Medical Research Center, Seoul National University College of Medicine, Seoul03080, South Korea
| | - Jungho Kim
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul03080, South Korea
- Neuroscience Research Institute, Medical Research Center, Seoul National University College of Medicine, Seoul03080, South Korea
- Transplantation Research Institute, Medical Research Center, Seoul National University College of Medicine, Seoul03080, South Korea
| | - Hyun-Hee Ryu
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul03080, South Korea
- Neuroscience Research Institute, Medical Research Center, Seoul National University College of Medicine, Seoul03080, South Korea
- Department of Physiology, Seoul National University College of Medicine, Seoul03080, South Korea
| | - Hanbyul Jang
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul03080, South Korea
- Neuroscience Research Institute, Medical Research Center, Seoul National University College of Medicine, Seoul03080, South Korea
- Department of Physiology, Seoul National University College of Medicine, Seoul03080, South Korea
| | - DoEun Lee
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul03080, South Korea
- Neuroscience Research Institute, Medical Research Center, Seoul National University College of Medicine, Seoul03080, South Korea
- Transplantation Research Institute, Medical Research Center, Seoul National University College of Medicine, Seoul03080, South Korea
| | - Seungha Lee
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul03080, South Korea
- Neuroscience Research Institute, Medical Research Center, Seoul National University College of Medicine, Seoul03080, South Korea
- Transplantation Research Institute, Medical Research Center, Seoul National University College of Medicine, Seoul03080, South Korea
| | - Jae-man Song
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul03080, South Korea
- Neuroscience Research Institute, Medical Research Center, Seoul National University College of Medicine, Seoul03080, South Korea
- Transplantation Research Institute, Medical Research Center, Seoul National University College of Medicine, Seoul03080, South Korea
| | - Yong-Seok Lee
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul03080, South Korea
- Neuroscience Research Institute, Medical Research Center, Seoul National University College of Medicine, Seoul03080, South Korea
- Department of Physiology, Seoul National University College of Medicine, Seoul03080, South Korea
| | - Young Ho Suh
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul03080, South Korea
- Neuroscience Research Institute, Medical Research Center, Seoul National University College of Medicine, Seoul03080, South Korea
- Transplantation Research Institute, Medical Research Center, Seoul National University College of Medicine, Seoul03080, South Korea
| |
Collapse
|
6
|
Aberuagba A, Joel EB, Bello AJ, Igunnu A, Malomo SO, Olorunniji FJ. Thermophilic PHP Protein Tyrosine Phosphatases (Cap8C and Wzb) from Mesophilic Bacteria. Int J Mol Sci 2024; 25:1262. [PMID: 38279261 PMCID: PMC10816263 DOI: 10.3390/ijms25021262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Revised: 01/14/2024] [Accepted: 01/17/2024] [Indexed: 01/28/2024] Open
Abstract
Protein tyrosine phosphatases (PTPs) of the polymerase and histidinol phosphatase (PHP) superfamily with characteristic phosphatase activity dependent on divalent metal ions are found in many Gram-positive bacteria. Although members of this family are co-purified with metal ions, they still require the exogenous supply of metal ions for full activation. However, the specific roles these metal ions play during catalysis are yet to be well understood. Here, we report the metal ion requirement for phosphatase activities of S. aureus Cap8C and L. rhamnosus Wzb. AlphaFold-predicted structures of the two PTPs suggest that they are members of the PHP family. Like other PHP phosphatases, the two enzymes have a catalytic preference for Mn2+, Co2+ and Ni2+ ions. Cap8C and Wzb show an unusual thermophilic property with optimum activities over 75 °C. Consistent with this model, the activity-temperature profiles of the two enzymes are dependent on the divalent metal ion activating the enzyme.
Collapse
Affiliation(s)
- Adepeju Aberuagba
- School of Pharmacy & Biomolecular Sciences, Liverpool John Moores University, Byrom Street, Liverpool L3 3AF, UK; (A.A.); (E.B.J.); (A.J.B.)
| | - Enoch B. Joel
- School of Pharmacy & Biomolecular Sciences, Liverpool John Moores University, Byrom Street, Liverpool L3 3AF, UK; (A.A.); (E.B.J.); (A.J.B.)
- Department of Biochemistry, Faculty of Basic Medical Sciences, University of Jos, Jos 930003, Nigeria
| | - Adebayo J. Bello
- School of Pharmacy & Biomolecular Sciences, Liverpool John Moores University, Byrom Street, Liverpool L3 3AF, UK; (A.A.); (E.B.J.); (A.J.B.)
| | - Adedoyin Igunnu
- Department of Biochemistry, Faculty of Life Sciences, University of Ilorin, Ilorin 234031, Nigeria; (A.I.); (S.O.M.)
| | - Sylvia O. Malomo
- Department of Biochemistry, Faculty of Life Sciences, University of Ilorin, Ilorin 234031, Nigeria; (A.I.); (S.O.M.)
| | - Femi J. Olorunniji
- School of Pharmacy & Biomolecular Sciences, Liverpool John Moores University, Byrom Street, Liverpool L3 3AF, UK; (A.A.); (E.B.J.); (A.J.B.)
| |
Collapse
|
7
|
Bagwe PV, Deshpande RD, Juhasz G, Sathaye S, Joshi SV. Uncovering the Significance of STEP61 in Alzheimer's Disease: Structure, Substrates, and Interactome. Cell Mol Neurobiol 2023; 43:3099-3113. [PMID: 37219664 PMCID: PMC11410018 DOI: 10.1007/s10571-023-01364-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 05/15/2023] [Indexed: 05/24/2023]
Abstract
STEP (STriatal-Enriched Protein Tyrosine Phosphatase) is a brain-specific phosphatase that plays an important role in controlling signaling molecules involved in neuronal activity and synaptic development. The striatum is the main location of the STEP enzyme. An imbalance in STEP61 activity is a risk factor for Alzheimer's disease (AD). It can contribute to the development of numerous neuropsychiatric diseases, including Parkinson's disease (PD), schizophrenia, fragile X syndrome (FXS), Huntington's disease (HD), alcoholism, cerebral ischemia, and stress-related diseases. The molecular structure, chemistry, and molecular mechanisms associated with STEP61's two major substrates, Alpha-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid receptors (AMPAr) and N-methyl-D-aspartate receptors (NMDARs), are crucial in understanding the relationship between STEP61 and associated illnesses. STEP's interactions with its substrate proteins can alter the pathways of long-term potentiation and long-term depression. Therefore, understanding the role of STEP61 in neurological illnesses, particularly Alzheimer's disease-associated dementia, can provide valuable insights for possible therapeutic interventions. This review provides valuable insights into the molecular structure, chemistry, and molecular mechanisms associated with STEP61. This brain-specific phosphatase controls signaling molecules involved in neuronal activity and synaptic development. This review can aid researchers in gaining deep insights into the complex functions of STEP61.
Collapse
Affiliation(s)
- Pritam V Bagwe
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, Matunga, Mumbai, 400019, India
| | - Radni D Deshpande
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, Matunga, Mumbai, 400019, India
| | - Gabor Juhasz
- Clinical Research Unit (CRU Global Hungary Ltd.), Budapest, Hungary
| | - Sadhana Sathaye
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, Matunga, Mumbai, 400019, India.
| | - Shreerang V Joshi
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, Matunga, Mumbai, 400019, India.
| |
Collapse
|
8
|
Mallozzi C, Pepponi R, Gaddini L, Casella I, Chiodi V, Popoli P, Domenici MR. Functional Interaction between Adenosine A 2A and mGlu 5 Receptors Mediates STEP Phosphatase Activation and Promotes STEP/mGlu 5R Binding in Mouse Hippocampus and Neuroblastoma Cell Line. Biomolecules 2023; 13:1350. [PMID: 37759748 PMCID: PMC10527457 DOI: 10.3390/biom13091350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 08/22/2023] [Accepted: 08/24/2023] [Indexed: 09/29/2023] Open
Abstract
(1) Background: Recently, we found that adenosine A2A receptor (A2AR) stimulation results in an increase in STEP phosphatase activity. In order to delve into the mechanism through which A2AR stimulation induced STEP activation, we investigated the involvement of mGlu5R since it is well documented that A2AR and mGlu5R physically and functionally interact in several brain areas. (2) Methods: In a neuroblastoma cell line (SH-SY5Y) and in mouse hippocampal slices, we evaluated the enzymatic activity of STEP by using a para-nitrophenyl phosphate colorimetric assay. A co-immunoprecipitation assay and a Western blot analysis were used to evaluate STEP/mGlu5R binding. (3) Results: We found that the A2AR-dependent activation of STEP was mediated by the mGlu5R. Indeed, the A2AR agonist CGS 21680 significantly increased STEP activity, and this effect was prevented not only by the A2AR antagonist ZM 241385, as expected, but also by the mGlu5R antagonist MPEP. In addition, we found that mGlu5R agonist DHPG-induced STEP activation was reversed not only by the mGlu5R antagonist MPEP but also by ZM 241385. Finally, via co-immunoprecipitation experiments, we found that mGlu5R and STEP physically interact when both receptors are activated (4) Conclusions: These results demonstrated a close functional interaction between mGlu5 and A2A receptors in the modulation of STEP activity.
Collapse
Affiliation(s)
- Cinzia Mallozzi
- Department of Neuroscience, Istituto Superiore di Sanità, 00161 Rome, Italy
| | - Rita Pepponi
- National Centre for Drug Research and Evaluation, Istituto Superiore di Sanità, 00161 Rome, Italy; (R.P.); (L.G.); (I.C.); (V.C.); (P.P.)
| | - Lucia Gaddini
- National Centre for Drug Research and Evaluation, Istituto Superiore di Sanità, 00161 Rome, Italy; (R.P.); (L.G.); (I.C.); (V.C.); (P.P.)
| | - Ida Casella
- National Centre for Drug Research and Evaluation, Istituto Superiore di Sanità, 00161 Rome, Italy; (R.P.); (L.G.); (I.C.); (V.C.); (P.P.)
| | - Valentina Chiodi
- National Centre for Drug Research and Evaluation, Istituto Superiore di Sanità, 00161 Rome, Italy; (R.P.); (L.G.); (I.C.); (V.C.); (P.P.)
| | - Patrizia Popoli
- National Centre for Drug Research and Evaluation, Istituto Superiore di Sanità, 00161 Rome, Italy; (R.P.); (L.G.); (I.C.); (V.C.); (P.P.)
| | - Maria Rosaria Domenici
- National Centre for Drug Research and Evaluation, Istituto Superiore di Sanità, 00161 Rome, Italy; (R.P.); (L.G.); (I.C.); (V.C.); (P.P.)
| |
Collapse
|
9
|
Jin K, Yao Z, van Velthoven CTJ, Kaplan ES, Glattfelder K, Barlow ST, Boyer G, Carey D, Casper T, Chakka AB, Chakrabarty R, Clark M, Departee M, Desierto M, Gary A, Gloe J, Goldy J, Guilford N, Guzman J, Hirschstein D, Lee C, Liang E, Pham T, Reding M, Ronellenfitch K, Ruiz A, Sevigny J, Shapovalova N, Shulga L, Sulc J, Torkelson A, Tung H, Levi B, Sunkin SM, Dee N, Esposito L, Smith K, Tasic B, Zeng H. Cell-type specific molecular signatures of aging revealed in a brain-wide transcriptomic cell-type atlas. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.26.550355. [PMID: 38168182 PMCID: PMC10760145 DOI: 10.1101/2023.07.26.550355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
Biological aging can be defined as a gradual loss of homeostasis across various aspects of molecular and cellular function. Aging is a complex and dynamic process which influences distinct cell types in a myriad of ways. The cellular architecture of the mammalian brain is heterogeneous and diverse, making it challenging to identify precise areas and cell types of the brain that are more susceptible to aging than others. Here, we present a high-resolution single-cell RNA sequencing dataset containing ~1.2 million high-quality single-cell transcriptomic profiles of brain cells from young adult and aged mice across both sexes, including areas spanning the forebrain, midbrain, and hindbrain. We find age-associated gene expression signatures across nearly all 130+ neuronal and non-neuronal cell subclasses we identified. We detect the greatest gene expression changes in non-neuronal cell types, suggesting that different cell types in the brain vary in their susceptibility to aging. We identify specific, age-enriched clusters within specific glial, vascular, and immune cell types from both cortical and subcortical regions of the brain, and specific gene expression changes associated with cell senescence, inflammation, decrease in new myelination, and decreased vasculature integrity. We also identify genes with expression changes across multiple cell subclasses, pointing to certain mechanisms of aging that may occur across wide regions or broad cell types of the brain. Finally, we discover the greatest gene expression changes in cell types localized to the third ventricle of the hypothalamus, including tanycytes, ependymal cells, and Tbx3+ neurons found in the arcuate nucleus that are part of the neuronal circuits regulating food intake and energy homeostasis. These findings suggest that the area surrounding the third ventricle in the hypothalamus may be a hub for aging in the mouse brain. Overall, we reveal a dynamic landscape of cell-type-specific transcriptomic changes in the brain associated with normal aging that will serve as a foundation for the investigation of functional changes in the aging process and the interaction of aging and diseases.
Collapse
Affiliation(s)
- Kelly Jin
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Zizhen Yao
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | | | | | | | | | - Daniel Carey
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | | | | | | | - Max Departee
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | - Amanda Gary
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Jessica Gloe
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Jeff Goldy
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | | | | | - Changkyu Lee
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | | | | | | | | | - Josh Sevigny
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | | | - Josef Sulc
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | - Herman Tung
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Boaz Levi
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | - Nick Dee
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | | | | | - Hongkui Zeng
- Allen Institute for Brain Science, Seattle, WA, USA
| |
Collapse
|
10
|
Chudoba R, Dabrowska J. Distinct populations of corticotropin-releasing factor (CRF) neurons mediate divergent yet complementary defensive behaviors in response to a threat. Neuropharmacology 2023; 228:109461. [PMID: 36775096 PMCID: PMC10055972 DOI: 10.1016/j.neuropharm.2023.109461] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 01/31/2023] [Accepted: 02/09/2023] [Indexed: 02/12/2023]
Abstract
Defensive behaviors in response to a threat are shared across the animal kingdom. Active (fleeing, sheltering) or passive (freezing, avoiding) defensive responses are adaptive and facilitate survival. Selecting appropriate defensive strategy depends on intensity, proximity, temporal threat threshold, and past experiences. Hypothalamic corticotropin-releasing factor (CRF) is a major driver of an acute stress response, whereas extrahypothalamic CRF mediates stress-related affective behaviors. In this review, we shift the focus from a monolithic role of CRF as an anxiogenic peptide to comprehensively dissecting contributions of distinct populations of CRF neurons in mediating defensive behaviors. Direct interrogation of CRF neurons of the central amygdala (CeA) or the bed nucleus of the stria terminalis (BNST) show they drive unconditioned defensive responses, such as vigilance and avoidance of open spaces. Although both populations also contribute to learned fear responses in familiar, threatening contexts, CeA-CRF neurons are particularly attuned to the ever-changing environment. Depending on threat intensities, they facilitate discrimination of salient stimuli predicting manageable threats, and prevent their generalization. Finally, hypothalamic CRF neurons mediate initial threat assessment and active defense such as escape to shelter. Overall, these three major populations of CRF neurons demonstrate divergent, yet complementary contributions to the versatile defense system: heightened vigilance, discriminating salient threats, and active escape, representing three legs of the defense tripod. Despite the 'CRF exhaustion' in the field of affective neuroscience, understanding contributions of specific CRF neurons during adaptive defensive behaviors is needed in order to understand the implications of their dysregulation in fear- and anxiety-related psychiatric disorders. This article is part of the Special Issue on "Fear, Anxiety and PTSD".
Collapse
Affiliation(s)
- Rachel Chudoba
- Center for the Neurobiology of Stress Resilience and Psychiatric Disorders, Rosalind Franklin University of Medicine and Science, North Chicago, IL, United States; Discipline of Cellular and Molecular Pharmacology, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL, United States; School of Graduate and Postdoctoral Studies, Rosalind Franklin University of Medicine and Science, North Chicago, IL, United States
| | - Joanna Dabrowska
- Center for the Neurobiology of Stress Resilience and Psychiatric Disorders, Rosalind Franklin University of Medicine and Science, North Chicago, IL, United States; Discipline of Cellular and Molecular Pharmacology, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL, United States; School of Graduate and Postdoctoral Studies, Rosalind Franklin University of Medicine and Science, North Chicago, IL, United States.
| |
Collapse
|
11
|
Pan L, Li T, Wang R, Deng W, Pu H, Deng M. Roles of Phosphorylation of N-Methyl-D-Aspartate Receptor in Chronic Pain. Cell Mol Neurobiol 2023; 43:155-175. [PMID: 35032275 PMCID: PMC11415214 DOI: 10.1007/s10571-022-01188-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 01/03/2022] [Indexed: 01/07/2023]
Abstract
Phosphorylation of N-methyl-D-aspartate receptor (NMDAR) is widely regarded as a vital modification of synaptic function. Various protein kinases are responsible for direct phosphorylation of NMDAR, such as cyclic adenosine monophosphate-dependent protein kinase A, protein kinase C, Ca2+/calmodulin-dependent protein kinase II, Src family protein tyrosine kinases, cyclin-dependent kinase 5, and casein kinase II. The detailed function of these kinases on distinct subunits of NMDAR has been reported previously and contributes to phosphorylation at sites predominately within the C-terminal of NMDAR. Phosphorylation underlies both structural and functional changes observed in chronic pain, and studies have demonstrated that inhibitors of kinases are significantly effective in alleviating pain behavior in different chronic pain models. In addition, the exploration of drugs that aim to disrupt the interaction between kinases and NMDAR is promising in clinical research. Based on research regarding the modulation of NMDAR in chronic pain models, this review provides an overview of the phosphorylation of NMDAR-related mechanisms underlying chronic pain to elucidate molecular and pharmacologic references for chronic pain management.
Collapse
Affiliation(s)
- Liangyu Pan
- Department of Biochemistry and Molecular Biology and Hunan Province Key Laboratory of Basic and Applied Hematology, School of Life Sciences, Central South University, Changsha, 410013, Hunan, China
- Hunan Key Laboratory of Animal Models for Human Diseases & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, 410013, Hunan, China
| | - Tiansheng Li
- Department of Biochemistry and Molecular Biology and Hunan Province Key Laboratory of Basic and Applied Hematology, School of Life Sciences, Central South University, Changsha, 410013, Hunan, China
- Hunan Key Laboratory of Animal Models for Human Diseases & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, 410013, Hunan, China
| | - Rui Wang
- Department of Biochemistry and Molecular Biology and Hunan Province Key Laboratory of Basic and Applied Hematology, School of Life Sciences, Central South University, Changsha, 410013, Hunan, China
- Hunan Key Laboratory of Animal Models for Human Diseases & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, 410013, Hunan, China
| | - Weiheng Deng
- Department of Biochemistry and Molecular Biology and Hunan Province Key Laboratory of Basic and Applied Hematology, School of Life Sciences, Central South University, Changsha, 410013, Hunan, China
- Hunan Key Laboratory of Animal Models for Human Diseases & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, 410013, Hunan, China
| | - Huangsheng Pu
- College of Advanced Interdisciplinary Studies, National University of Defense Technology, Changsha, 410073, Hunan, China.
| | - Meichun Deng
- Department of Biochemistry and Molecular Biology and Hunan Province Key Laboratory of Basic and Applied Hematology, School of Life Sciences, Central South University, Changsha, 410013, Hunan, China.
- Hunan Key Laboratory of Animal Models for Human Diseases & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, 410013, Hunan, China.
| |
Collapse
|
12
|
Baranowski MR, Wu J, Han YN, Lambert LJ, Cosford NDP, Tautz L. Protein Tyrosine Phosphatase Biochemical Inhibition Assays. Bio Protoc 2022; 12:e4510. [PMID: 36248604 PMCID: PMC9516250 DOI: 10.21769/bioprotoc.4510] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 07/13/2022] [Accepted: 07/20/2022] [Indexed: 12/29/2022] Open
Abstract
Disturbance of the dynamic balance between protein tyrosine phosphorylation and dephosphorylation, modulated by protein tyrosine kinases (PTKs) and protein tyrosine phosphatases (PTPs), is known to be crucial for the development of many human diseases. The discovery of agents that restore this balance has been the subject of many drug research efforts, most of which have focused on tyrosine kinase inhibitors (TKIs), resulting in the development of more than 50 FDA-approved TKIs during the past two decades. More recently, accumulating evidence has suggested that members of the PTP superfamily are also promising drug targets, and efforts to discover tyrosine phosphatase inhibitors (TPIs) have increased dramatically. Here, we provide protocols for determining the potency of TPIs in vitro. We focus on the use of fluorescence-based substrates, which exhibit a dramatic increase in fluorescence emission when dephosphorylated by the PTP, and thus allow setting up highly sensitive and miniaturized phosphatase activity assays using 384-well or 1536-well microplates and a continuous (kinetic) assay format. The protocols cover PTP specific activity assays, Michaelis-Menten kinetics, dose-response inhibition assays, and dose-response data analysis for determining IC 50 values. Potential pitfalls are also discussed. While advanced instrumentation is utilized for compound spotting and liquid dispensing, all the assays can be adapted to existing equipment in most laboratories. Assays are described for selected PTP drug targets, including SHP2 ( PTPN11 ), PTP1B ( PTPN1 ), STEP ( PTPN5 ), and VHR ( DUSP3 ). However, all protocols are applicable to members of the PTP enzyme family in general. Graphical abstract.
Collapse
Affiliation(s)
- Marek R. Baranowski
- NCI-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, 10901 N Torrey Pines Rd, La Jolla, CA 92037, USA
- Division of Biophysics, Institute of Experimental Physics, Faculty of Physics, University of Warsaw, Ludwika Pasteura 5, 02-093 Warsaw, Poland
| | - Jiaqian Wu
- NCI-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, 10901 N Torrey Pines Rd, La Jolla, CA 92037, USA
| | - Ye Na Han
- NCI-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, 10901 N Torrey Pines Rd, La Jolla, CA 92037, USA
| | - Lester J. Lambert
- NCI-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, 10901 N Torrey Pines Rd, La Jolla, CA 92037, USA
| | - Nicholas D. P. Cosford
- NCI-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, 10901 N Torrey Pines Rd, La Jolla, CA 92037, USA
| | - Lutz Tautz
- NCI-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, 10901 N Torrey Pines Rd, La Jolla, CA 92037, USA
| |
Collapse
|
13
|
Walters JM, Kim EC, Zhang J, Jeong HG, Bajaj A, Baculis B, Tracy G, Ibrahim B, Christian-Hinman CA, Llano DA, Huesmann GR, Chung HJ. Pharmacological inhibition of STriatal-Enriched protein tyrosine Phosphatase by TC-2153 reduces hippocampal excitability and seizure propensity. Epilepsia 2022; 63:1211-1224. [PMID: 35188269 PMCID: PMC9586517 DOI: 10.1111/epi.17192] [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: 08/02/2021] [Revised: 12/30/2021] [Accepted: 02/01/2022] [Indexed: 11/29/2022]
Abstract
OBJECTIVE STriatal-Enriched protein tyrosine Phosphatase (STEP) is a brain-specific tyrosine phosphatase. Membrane-bound STEP61 is the only isoform expressed in hippocampus and cortex. Genetic deletion of STEP enhances excitatory synaptic currents and long-term potentiation in the hippocampus. However, whether STEP61 affects seizure susceptibility is unclear. Here we investigated the effects of STEP inhibitor TC-2153 on seizure propensity in a murine model displaying kainic acid (KA)-induced status epilepticus and its effect on hippocampal excitability. METHODS Adult male and female C57BL/6J mice received intraperitoneal injection of either vehicle (2.8% dimethylsulfoxide [DMSO] in saline) or TC-2153 (10 mg/kg) and then either saline or KA (30 mg/kg) 3 h later before being monitored for behavioral seizures. A subset of female mice was ovariectomized (OVX). Acute hippocampal slices from Thy1-GCaMP6s mice were treated with either DMSO or TC-2153 (10 μM) for 1 h, and then incubated in artificial cerebrospinal fluid (ACSF) and potassium chloride (15 mM) for 2 min prior to live calcium imaging. Pyramidal neurons in dissociated rat hippocampal culture (DIV 8-10) were pre-treated with DMSO or TC-2153 (10 µM) for 1 h before whole-cell patch-clamp recording. RESULTS TC-2153 treatment significantly reduced KA-induced seizure severity, with greater trend seen in female mice. OVX abolished this TC-2153-induced decrease in seizure severity in female mice. TC-2153 application significantly decreased overall excitability of acute hippocampal slices from both sexes. Surprisingly, TC-2153 treatment hyperpolarized resting membrane potential and decreased firing rate, sag voltage, and hyperpolarization-induced current (Ih ) of cultured hippocampal pyramidal neurons. SIGNIFICANCE This study is the first to demonstrate that pharmacological inhibition of STEP with TC-2153 decreases seizure severity and hippocampal activity in both sexes, and dampens hippocampal neuronal excitability and Ih . We propose that the antiseizure effects of TC-2153 are mediated by its unexpected action on suppressing neuronal intrinsic excitability.
Collapse
Affiliation(s)
- Jennifer M. Walters
- Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, IL, USA
- Dept. of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Eung Chang Kim
- Dept. of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Jiaren Zhang
- Dept. of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Han Gil Jeong
- Dept. of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Archit Bajaj
- Dept. of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Brian Baculis
- Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, IL, USA
- Dept. of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Gregory Tracy
- Dept. of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Baher Ibrahim
- Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, IL, USA
- Dept. of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL, USA
- Beckman Institute, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Catherine A. Christian-Hinman
- Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, IL, USA
- Dept. of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL, USA
- Beckman Institute, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Daniel A. Llano
- Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, IL, USA
- Dept. of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL, USA
- Beckman Institute, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Graham R. Huesmann
- Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, IL, USA
- Dept. of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL, USA
- Beckman Institute, University of Illinois at Urbana-Champaign, Urbana, IL, USA
- Department of Neurology, Carle Foundation Hospital, Urbana, IL, USA
- Carle Illinois College of Medicine, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Hee Jung Chung
- Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, IL, USA
- Dept. of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL, USA
- Beckman Institute, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| |
Collapse
|
14
|
Zhao Y, Yang WQ, Yu L, Yang J, Zhu HR, Zhang L. Dl-3-n-butylphthalide alleviates cognitive impairment in amyloid precursor protein/presenilin 1 transgenic mice by regulating the striatal-enriched protein tyrosine phosphatase/ERK/cAMP-response element-binding protein signaling pathway. Exp Ther Med 2022; 23:319. [PMID: 35350668 PMCID: PMC8943801 DOI: 10.3892/etm.2022.11248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Accepted: 01/04/2022] [Indexed: 12/27/2022] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder characterized by cognitive impairment and the deposition of amyloid plaques in the brain. In a transgenic mouse model of AD, cognitive impairment and synaptic dysfunction were revealed to be associated with soluble amyloid oligomers and to occur prior to plaque formation. The results of our previous studies revealed that striatal-enriched protein tyrosine phosphatase (STEP)61 negatively regulated the β-amyloid protein-mediated ERK/cAMP-response element-binding protein (CREB) signaling pathway. Dl-3-n-butylphthalide (NBP) is a synthetic compound approved by the Food and Drug Administration of China for the treatment of ischemic stroke in 2002. Studies have shown that the neuroprotective effects of NBP involve multiple mechanisms. The present study further explored the mechanism of NBP therapy in amyloid precursor protein (APP)/presenilin 1 (PS1) transgenic mice, and the involvement of the STEP/ERK/CREB signaling pathway. The results suggested that NBP treatment effectively ameliorated the spatial learning and memory impairment of the APP/PS1 transgenic mice, which was assessed using a Morris water maze. In addition, NBP reduced amyloid-induced activation of STEP61 levels, while increasing phosphorylated (p)-ERK1/2 and p-CREB levels in the cerebral cortex and hippocampus of APP/PS1 transgenic mice by western blotting and immunostaining. In conclusion, the present study provided evidence to suggest that the new drug NBP improved amyloid-induced learning and memory deficits, likely through the regulation of the STEP/ERK/CREB pathway. The results revealed that NBP, as a multi-target drug, may exert a neuroprotective effect. Therefore, NBP may serve as an effective treatment for AD.
Collapse
Affiliation(s)
- Yan Zhao
- Life Science Institution, Jinzhou Medical University, Jinzhou, Liaoning 121001, P.R. China
| | - Wen-Qiang Yang
- Life Science Institution, Jinzhou Medical University, Jinzhou, Liaoning 121001, P.R. China
| | - Lu Yu
- Provincial Key Laboratory of Cardiovascular and Cerebrovascular Drug Basic Research, Jinzhou Medical University, Jinzhou, Liaoning 121001, P.R. China
| | - Jing Yang
- Provincial Key Laboratory of Cardiovascular and Cerebrovascular Drug Basic Research, Jinzhou Medical University, Jinzhou, Liaoning 121001, P.R. China
| | - Hai-Rong Zhu
- Department of Neurology, Affiliated Taizhou Hospital of Nanjing University of Chinese Medicine, Taizhou, Jiangsu 225300, P.R. China
| | - Lin Zhang
- Department of Neurology, Affiliated Wenling Hospital of Wenzhou Medical University, Wenling, Zhejiang 317500, P.R. China,Correspondence to: Dr Lin Zhang, Department of Neurology, Affiliated Wenling Hospital of Wenzhou Medical University, 333 Chuan'an South Road, Chengxi Street, Wenling, Zhejiang 317500, P.R. China
| |
Collapse
|
15
|
Moskaliuk VS, Kozhemyakina RV, Bazovkina DV, Terenina E, Khomenko TM, Volcho KP, Salakhutdinov NF, Kulikov AV, Naumenko VS, Kulikova E. On an association between fear-induced aggression and striatal-enriched protein tyrosine phosphatase (STEP) in the brain of Norway rats. Pharmacotherapy 2022; 147:112667. [DOI: 10.1016/j.biopha.2022.112667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 01/14/2022] [Accepted: 01/24/2022] [Indexed: 11/28/2022]
|
16
|
Elhassan RM, Hou X, Fang H. Recent advances in the development of allosteric protein tyrosine phosphatase inhibitors for drug discovery. Med Res Rev 2021; 42:1064-1110. [PMID: 34791703 DOI: 10.1002/med.21871] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 09/26/2021] [Accepted: 10/24/2021] [Indexed: 01/07/2023]
Abstract
Protein tyrosine phosphatases (PTPs) superfamily catalyzes tyrosine de-phosphorylation which affects a myriad of cellular processes. Imbalance in signal pathways mediated by PTPs has been associated with development of many human diseases including cancer, metabolic, and immunological diseases. Several compelling evidence suggest that many members of PTP family are novel therapeutic targets. However, the clinical development of conventional PTP-based active-site inhibitors originally was hampered by the poor selectivity and pharmacokinetic properties. In this regard, PTPs has been widely dismissed as "undruggable." Nonetheless, allosteric modulation has become increasingly an influential and alternative approach that can be exploited for drug development against PTPs. Unlike active-site inhibitors, allosteric inhibitors exhibit a remarkable target-selectivity, drug-likeness, potency, and in vivo activity. Intriguingly, there has been a high interest in novel allosteric PTPs inhibitors within the last years. In this review, we focus on the recent advances of allosteric inhibitors that have been explored in drug discovery and have shown an excellent result in the development of PTPs-based therapeutics. A special emphasis is placed on the structure-activity relationship and molecular mechanistic studies illustrating applications in chemical biology and medicinal chemistry.
Collapse
Affiliation(s)
- Reham M Elhassan
- Department of Medicinal Chemistry and Key Laboratory of Chemical Biology of Natural Products (MOE), School of Pharmacy, Shandong University, Jinan, Shandong, China
| | - Xuben Hou
- Department of Medicinal Chemistry and Key Laboratory of Chemical Biology of Natural Products (MOE), School of Pharmacy, Shandong University, Jinan, Shandong, China
| | - Hao Fang
- Department of Medicinal Chemistry and Key Laboratory of Chemical Biology of Natural Products (MOE), School of Pharmacy, Shandong University, Jinan, Shandong, China
| |
Collapse
|
17
|
de Pins B, Mendes T, Giralt A, Girault JA. The Non-receptor Tyrosine Kinase Pyk2 in Brain Function and Neurological and Psychiatric Diseases. Front Synaptic Neurosci 2021; 13:749001. [PMID: 34690733 PMCID: PMC8527176 DOI: 10.3389/fnsyn.2021.749001] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 09/14/2021] [Indexed: 12/28/2022] Open
Abstract
Pyk2 is a non-receptor tyrosine kinase highly enriched in forebrain neurons. Pyk2 is closely related to focal adhesion kinase (FAK), which plays an important role in sensing cell contacts with extracellular matrix and other extracellular signals controlling adhesion and survival. Pyk2 shares some of FAK’s characteristics including recruitment of Src-family kinases after autophosphorylation, scaffolding by interacting with multiple partners, and activation of downstream signaling pathways. Pyk2, however, has the unique property to respond to increases in intracellular free Ca2+, which triggers its autophosphorylation following stimulation of various receptors including glutamate NMDA receptors. Pyk2 is dephosphorylated by the striatal-enriched phosphatase (STEP) that is highly expressed in the same neuronal populations. Pyk2 localization in neurons is dynamic, and altered following stimulation, with post-synaptic and nuclear enrichment. As a signaling protein Pyk2 is involved in multiple pathways resulting in sometimes opposing functions depending on experimental models. Thus Pyk2 has a dual role on neurites and dendritic spines. With Src family kinases Pyk2 participates in postsynaptic regulations including of NMDA receptors and is necessary for specific types of synaptic plasticity and spatial memory tasks. The diverse functions of Pyk2 are also illustrated by its role in pathology. Pyk2 is activated following epileptic seizures or ischemia-reperfusion and may contribute to the consequences of these insults whereas Pyk2 deficit may contribute to the hippocampal phenotype of Huntington’s disease. Pyk2 gene, PTK2B, is associated with the risk for late-onset Alzheimer’s disease. Studies of underlying mechanisms indicate a complex contribution with involvement in amyloid toxicity and tauopathy, combined with possible functional deficits in neurons and contribution in microglia. A role of Pyk2 has also been proposed in stress-induced depression and cocaine addiction. Pyk2 is also important for the mobility of astrocytes and glioblastoma cells. The implication of Pyk2 in various pathological conditions supports its potential interest for therapeutic interventions. This is possible through molecules inhibiting its activity or increasing it through inhibition of STEP or other means, depending on a precise evaluation of the balance between positive and negative consequences of Pyk2 actions.
Collapse
Affiliation(s)
- Benoit de Pins
- Institut du Fer à Moulin, Paris, France.,Inserm UMR-S 1270, Paris, France.,Faculté des Sciences et Ingénierie, Sorbonne Université, Paris, France
| | - Tiago Mendes
- Institut du Fer à Moulin, Paris, France.,Inserm UMR-S 1270, Paris, France.,Faculté des Sciences et Ingénierie, Sorbonne Université, Paris, France
| | - Albert Giralt
- Departament de Biomedicina, Facultat de Medicina i Ciències de la Salut, Institut de Neurociències, Universitat de Barcelona, Barcelona, Spain.,Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.,Production and Validation Center of Advanced Therapies (Creatio), Faculty of Medicine and Health Science, University of Barcelona, Barcelona, Spain
| | - Jean-Antoine Girault
- Institut du Fer à Moulin, Paris, France.,Inserm UMR-S 1270, Paris, France.,Faculté des Sciences et Ingénierie, Sorbonne Université, Paris, France
| |
Collapse
|
18
|
Carvajal FJ, Cerpa W. Regulation of Phosphorylated State of NMDA Receptor by STEP 61 Phosphatase after Mild-Traumatic Brain Injury: Role of Oxidative Stress. Antioxidants (Basel) 2021; 10:antiox10101575. [PMID: 34679709 PMCID: PMC8533270 DOI: 10.3390/antiox10101575] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 09/14/2021] [Accepted: 09/28/2021] [Indexed: 01/21/2023] Open
Abstract
Traumatic Brain Injury (TBI) mediates neuronal death through several events involving many molecular pathways, including the glutamate-mediated excitotoxicity for excessive stimulation of N-methyl-D-aspartate receptors (NMDARs), producing activation of death signaling pathways. However, the contribution of NMDARs (distribution and signaling-associated to the distribution) remains incompletely understood. We propose a critical role of STEP61 (Striatal-Enriched protein tyrosine phosphatase) in TBI; this phosphatase regulates the dephosphorylated state of the GluN2B subunit through two pathways: by direct dephosphorylation of tyrosine-1472 and indirectly via dephosphorylation and inactivation of Fyn kinase. We previously demonstrated oxidative stress’s contribution to NMDAR signaling and distribution using SOD2+/− mice such a model. We performed TBI protocol using a controlled frontal impact device using C57BL/6 mice and SOD2+/− animals. After TBI, we found alterations in cognitive performance, NMDAR-dependent synaptic function (decreased synaptic form of NMDARs and decreased synaptic current NMDAR-dependent), and increased STEP61 activity. These changes are reduced partially with the STEP61-inhibitor TC-2153 treatment in mice subjected to TBI protocol. This study contributes with evidence about the role of STEP61 in the neuropathological progression after TBI and also the alteration in their activity, such as an early biomarker of synaptic damage in traumatic lesions.
Collapse
Affiliation(s)
- Francisco J. Carvajal
- Laboratorio de Función y Patología Neuronal, Centro de Envejecimiento y Regeneración (CARE), Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago 8331150, Chile;
- Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Universidad de Magallanes, Punta Arenas 6200000, Chile
| | - Waldo Cerpa
- Laboratorio de Función y Patología Neuronal, Centro de Envejecimiento y Regeneración (CARE), Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago 8331150, Chile;
- Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Universidad de Magallanes, Punta Arenas 6200000, Chile
- Correspondence: ; Tel.: +56-2-2354-2656; Fax: +56-2-2354-2660
| |
Collapse
|
19
|
Foster SL, Lustberg DJ, Harbin NH, Bramlett SN, Hepler JR, Weinshenker D. RGS14 modulates locomotor behavior and ERK signaling induced by environmental novelty and cocaine within discrete limbic structures. Psychopharmacology (Berl) 2021; 238:2755-2773. [PMID: 34184126 PMCID: PMC8455459 DOI: 10.1007/s00213-021-05892-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Accepted: 06/01/2021] [Indexed: 12/14/2022]
Abstract
RATIONALE In rodents, exposure to novel environments or psychostimulants promotes locomotion. Indeed, locomotor reactivity to novelty strongly predicts behavioral responses to psychostimulants in animal models of addiction. RGS14 is a plasticity-restricting protein with unique functional domains that enable it to suppress ERK-dependent signaling as well as regulate G protein activity. Although recent studies show that RGS14 is expressed in multiple limbic regions implicated in psychostimulant- and novelty-induced hyperlocomotion, its function has been examined mostly in the context of hippocampal physiology and memory. OBJECTIVE We investigated whether RGS14 modulates novelty- and cocaine-induced locomotion (NIL and CIL, respectively) and neuronal activity. METHODS We assessed Rgs14 knockout (RGS14 KO) mice and wild-type (WT) littermate controls using NIL and CIL behavioral tests, followed by quantification of c-fos and phosphorylated ERK (pERK) induction in limbic regions that normally express RGS14. RESULTS RGS14 KO mice were less active than WT controls in the NIL test, driven by avoidance of the center of the novel environment. By contrast, RGS14 KO mice demonstrated augmented peripheral locomotion in the CIL test conducted in either a familiar or novel environment. RGS14 KO mice exhibited increased thigmotaxis, as well as greater c-fos and pERK induction in the central amygdala and dorsal hippocampus, when cocaine and novelty were paired. CONCLUSIONS RGS14 KO mice exhibited anti-correlated locomotor responses to novelty and cocaine, but displayed increased thigmotaxis in response to either stimuli which was augmented by their combination. Our findings also suggest RGS14 may reduce neuronal activity in limbic subregions by inhibiting ERK-dependent signaling.
Collapse
Affiliation(s)
- Stephanie L Foster
- , Department of Human Genetics, Emory University School of Medicine, 615 Michael St., Whitehead 301, Atlanta, GA, 30322, USA
| | - Daniel J Lustberg
- , Department of Human Genetics, Emory University School of Medicine, 615 Michael St., Whitehead 301, Atlanta, GA, 30322, USA
| | - Nicholas H Harbin
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, 1510 Clifton Rd, Atlanta, GA, 30322, USA
| | - Sara N Bramlett
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, 1510 Clifton Rd, Atlanta, GA, 30322, USA
| | - John R Hepler
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, 1510 Clifton Rd, Atlanta, GA, 30322, USA.
| | - David Weinshenker
- , Department of Human Genetics, Emory University School of Medicine, 615 Michael St., Whitehead 301, Atlanta, GA, 30322, USA.
| |
Collapse
|
20
|
Kulikova EA, Fursenko DV, Bazhenova EY, Kulikov AV. Decrease in the Activity of Striatal-Enriched Protein-Tyrosine-Phosphatase (STEP) in the Brain of Danio rerio Treated with p-Chlorophenylalanine and Pargyline. Mol Biol 2021. [DOI: 10.1134/s0026893321020254] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
21
|
Wang H, Bu S, Tang J, Li Y, Liu C, Dong J. PTPN5 promotes follicle-stimulating hormone secretion through regulating intracellular calcium homeostasis. FASEB J 2021; 35:e21756. [PMID: 34270805 DOI: 10.1096/fj.202002752rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Revised: 05/16/2021] [Accepted: 06/10/2021] [Indexed: 11/11/2022]
Abstract
Protein tyrosine phosphatase non-receptor type 5 (PTPN5), also called striatal-enriched protein tyrosine phosphatase (STEP), is highly expressed in neurons of the basal ganglia, hippocampus, cortex, and related structures, also in the pituitary. Gonadotropins are the key regulator of the reproduction in mammals. In this study, PTPN5 is detected to express in murine pituitary in a developmental manner. Moreover, the expression of PTPN5 in the pituitary is heavily reduced after ovary removal. Follicle-stimulating hormone (FSH) secretion in gonadotropes is regulated by PTPN5 via binding GnRH to GnRH-R. Two parallel signaling pathways, Gs-protein kinase A (PKA)-PTPN5 and Gq-phospholipases C (PLC)-p38 MAPK-PTPN5, cooperatively regulate GnRH-induced FSH secretion. We also show that influx of Ca2+ activates the Ca2+ -dependent phosphatase calcineurin, leading to the phosphorylation and activation of PTPN5. The intracellular release of Ca2+ is reduced via TC2153. In conclusion, blocking or knocking out of PTPN5 reduces the release of FSH in whole pituitary. Mechanically, PTPN5 regulates gonadotropes' function through regulating intracellular calcium homeostasis.
Collapse
Affiliation(s)
- Hongmei Wang
- Department of Pharmacology, School of Medicine, Southeast University, Nanjing, China
| | - Siyuan Bu
- Department of Pharmacology, School of Medicine, Southeast University, Nanjing, China
| | - Jiajian Tang
- Department of Pharmacology, School of Medicine, Southeast University, Nanjing, China
| | - Yi Li
- Department of Joint Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Chunhua Liu
- Department of Physiology, Shandong First Medical University, Taian, China
| | - Junhong Dong
- Department of Biochemistry, School of Basic Medicine, Weifang Medical University, Weifang, China
| |
Collapse
|
22
|
Mahaman YAR, Huang F, Embaye KS, Wang X, Zhu F. The Implication of STEP in Synaptic Plasticity and Cognitive Impairments in Alzheimer's Disease and Other Neurological Disorders. Front Cell Dev Biol 2021; 9:680118. [PMID: 34195199 PMCID: PMC8236946 DOI: 10.3389/fcell.2021.680118] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Accepted: 05/06/2021] [Indexed: 12/31/2022] Open
Abstract
STriatal-Enriched protein tyrosine Phosphatase (STEP) is a tyrosine phosphatase that has been implicated in Alzheimer’s disease (AD), the most common form of dementia, and many other neurological diseases. The protein level and activity of STEP have been found to be elevated in most of these disorders, and specifically in AD as a result of dysregulation of different pathways including PP2B/DARPP32/PP1, PKA as well as impairments of both proteasomal and lysosomal systems. The upregulation in STEP leads to increased binding to, and dephosphorylation of, its substrates which are mainly found to be synaptic plasticity and thus learning and memory related proteins. These proteins include kinases like Fyn, Pyk2, ERK1/2 and both NMDA and AMPA receptor subunits GluN2B and GluA2. The dephosphorylation of these molecules results in inactivation of these kinases and internalization of NMDA and AMPA receptor complexes leading to synapse loss and cognitive impairments. In this study, we aim to review STEP regulation and its implications in AD as well as other neurological disorders and then summarize data on targeting STEP as therapeutic strategy in these diseases.
Collapse
Affiliation(s)
- Yacoubou Abdoul Razak Mahaman
- Cognitive Impairment Ward of Neurology Department, The Third Affiliated Hospital, Shenzhen University, Shenzhen, China.,Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fang Huang
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Kidane Siele Embaye
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaochuan Wang
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Feiqi Zhu
- Cognitive Impairment Ward of Neurology Department, The Third Affiliated Hospital, Shenzhen University, Shenzhen, China
| |
Collapse
|
23
|
Altered nociception in Alzheimer disease is associated with striatal-enriched protein tyrosine phosphatase signaling. Pain 2021; 162:1669-1680. [PMID: 33433143 DOI: 10.1097/j.pain.0000000000002180] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 12/18/2020] [Indexed: 01/16/2023]
Abstract
ABSTRACT Alzheimer disease (AD) is the most common form of dementia, accounting for approximately 60% of cases. In addition to memory loss, changes in pain sensitivity are found in a substantial proportion of patients with AD. However, the mechanism of nociception deficits in AD is still unclear. Here, we hypothesize that the nociception abnormality in AD is due to the aberrant activation of striatal-enriched protein tyrosine phosphatase (STEP) signaling, which modulates proteins related to nociception transduction. Our results indicated that the transgenic mice carrying human amyloid precursor protein (APP) gene had lower sensitivity to mechanical and thermal stimulation than the wild-type group at the ages of 6, 9, and 12 months. These APP mice exhibited elevated STEP activity and decreased phosphorylation of proteins involved in nociception transduction in hippocampi. The pharmacological inhibition of STEP activity using TC-2153 further reversed nociception and cognitive deficits in the APP mice. Moreover, the phosphorylation of nociception-related proteins in the APP mice was also rescued after STEP inhibitor treatment, indicating the key role of STEP in nociception alteration. In summary, this study identifies a mechanism for the reduced nociceptive sensitivity in an AD mouse model that could serve as a therapeutic target to improve the quality of life for patients with AD.
Collapse
|
24
|
Lambert LJ, Grotegut S, Celeridad M, Gosalia P, Backer LJSD, Bobkov AA, Salaniwal S, Chung TDY, Zeng FY, Pass I, Lombroso PJ, Cosford NDP, Tautz L. Development of a Robust High-Throughput Screening Platform for Inhibitors of the Striatal-Enriched Tyrosine Phosphatase (STEP). Int J Mol Sci 2021; 22:ijms22094417. [PMID: 33922601 PMCID: PMC8122956 DOI: 10.3390/ijms22094417] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Revised: 04/14/2021] [Accepted: 04/20/2021] [Indexed: 12/16/2022] Open
Abstract
Many human diseases are the result of abnormal expression or activation of protein tyrosine kinases (PTKs) and protein tyrosine phosphatases (PTPs). Not surprisingly, more than 30 tyrosine kinase inhibitors (TKIs) are currently in clinical use and provide unique treatment options for many patients. PTPs on the other hand have long been regarded as “undruggable” and only recently have gained increased attention in drug discovery. Striatal-enriched tyrosine phosphatase (STEP) is a neuron-specific PTP that is overactive in Alzheimer’s disease (AD) and other neurodegenerative and neuropsychiatric disorders, including Parkinson’s disease, schizophrenia, and fragile X syndrome. An emergent model suggests that the increase in STEP activity interferes with synaptic function and contributes to the characteristic cognitive and behavioral deficits present in these diseases. Prior efforts to generate STEP inhibitors with properties that warrant clinical development have largely failed. To identify novel STEP inhibitor scaffolds, we developed a biophysical, label-free high-throughput screening (HTS) platform based on the protein thermal shift (PTS) technology. In contrast to conventional HTS using STEP enzymatic assays, we found the PTS platform highly robust and capable of identifying true hits with confirmed STEP inhibitory activity and selectivity. This new platform promises to greatly advance STEP drug discovery and should be applicable to other PTP targets.
Collapse
Affiliation(s)
- Lester J Lambert
- Sanford Burnham Prebys Medical Discovery Institute, NCI-Designated Cancer Center, 10901 N Torrey Pines Rd, La Jolla, CA 92037, USA; (L.J.L.); (M.C.); (L.J.D.B.); (N.D.C.)
| | - Stefan Grotegut
- Sanford Burnham Prebys Medical Discovery Institute, Conrad Prebys Center for Chemical Genomics, 10901 N Torrey Pines Rd, La Jolla, CA 92037, USA; (S.G.); (P.G.); (A.A.B.); (S.S.); (T.D.C.); (F.-Y.Z.); (I.P.)
| | - Maria Celeridad
- Sanford Burnham Prebys Medical Discovery Institute, NCI-Designated Cancer Center, 10901 N Torrey Pines Rd, La Jolla, CA 92037, USA; (L.J.L.); (M.C.); (L.J.D.B.); (N.D.C.)
| | - Palak Gosalia
- Sanford Burnham Prebys Medical Discovery Institute, Conrad Prebys Center for Chemical Genomics, 10901 N Torrey Pines Rd, La Jolla, CA 92037, USA; (S.G.); (P.G.); (A.A.B.); (S.S.); (T.D.C.); (F.-Y.Z.); (I.P.)
| | - Laurent JS De Backer
- Sanford Burnham Prebys Medical Discovery Institute, NCI-Designated Cancer Center, 10901 N Torrey Pines Rd, La Jolla, CA 92037, USA; (L.J.L.); (M.C.); (L.J.D.B.); (N.D.C.)
| | - Andrey A Bobkov
- Sanford Burnham Prebys Medical Discovery Institute, Conrad Prebys Center for Chemical Genomics, 10901 N Torrey Pines Rd, La Jolla, CA 92037, USA; (S.G.); (P.G.); (A.A.B.); (S.S.); (T.D.C.); (F.-Y.Z.); (I.P.)
| | - Sumeet Salaniwal
- Sanford Burnham Prebys Medical Discovery Institute, Conrad Prebys Center for Chemical Genomics, 10901 N Torrey Pines Rd, La Jolla, CA 92037, USA; (S.G.); (P.G.); (A.A.B.); (S.S.); (T.D.C.); (F.-Y.Z.); (I.P.)
| | - Thomas DY Chung
- Sanford Burnham Prebys Medical Discovery Institute, Conrad Prebys Center for Chemical Genomics, 10901 N Torrey Pines Rd, La Jolla, CA 92037, USA; (S.G.); (P.G.); (A.A.B.); (S.S.); (T.D.C.); (F.-Y.Z.); (I.P.)
| | - Fu-Yue Zeng
- Sanford Burnham Prebys Medical Discovery Institute, Conrad Prebys Center for Chemical Genomics, 10901 N Torrey Pines Rd, La Jolla, CA 92037, USA; (S.G.); (P.G.); (A.A.B.); (S.S.); (T.D.C.); (F.-Y.Z.); (I.P.)
| | - Ian Pass
- Sanford Burnham Prebys Medical Discovery Institute, Conrad Prebys Center for Chemical Genomics, 10901 N Torrey Pines Rd, La Jolla, CA 92037, USA; (S.G.); (P.G.); (A.A.B.); (S.S.); (T.D.C.); (F.-Y.Z.); (I.P.)
| | - Paul J Lombroso
- Child Study Center, Departments of Psychiatry and Departments of Neurobiology, Yale University, 230 South Frontage Rd, New Haven, CT 06520, USA;
| | - Nicholas DP Cosford
- Sanford Burnham Prebys Medical Discovery Institute, NCI-Designated Cancer Center, 10901 N Torrey Pines Rd, La Jolla, CA 92037, USA; (L.J.L.); (M.C.); (L.J.D.B.); (N.D.C.)
| | - Lutz Tautz
- Sanford Burnham Prebys Medical Discovery Institute, NCI-Designated Cancer Center, 10901 N Torrey Pines Rd, La Jolla, CA 92037, USA; (L.J.L.); (M.C.); (L.J.D.B.); (N.D.C.)
- Correspondence:
| |
Collapse
|
25
|
Domenici MR, Mallozzi C, Pepponi R, Casella I, Chiodi V, Ferrante A, Popoli P. Insight into the Role of the STriatal-Enriched Protein Tyrosine Phosphatase (STEP) in A 2A Receptor-Mediated Effects in the Central Nervous System. Front Pharmacol 2021; 12:647742. [PMID: 33953681 PMCID: PMC8090931 DOI: 10.3389/fphar.2021.647742] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Accepted: 02/26/2021] [Indexed: 12/18/2022] Open
Abstract
The STriatal-Enriched protein tyrosine phosphatase STEP is a brain-specific tyrosine phosphatase that plays a pivotal role in the mechanisms of learning and memory, and it has been demonstrated to be involved in several neuropsychiatric diseases. Recently, we found a functional interaction between STEP and adenosine A2A receptor (A2AR), a subtype of the adenosine receptor family widely expressed in the central nervous system, where it regulates motor behavior and cognition, and plays a role in cell survival and neurodegeneration. Specifically, we demonstrated the involvement of STEP in A2AR-mediated cocaine effects in the striatum and, more recently, we found that in the rat striatum and hippocampus, as well as in a neuroblastoma cell line, the overexpression of the A2AR, or its stimulation, results in an increase in STEP activity. In the present article we will discuss the functional implication of this interaction, trying to examine the possible mechanisms involved in this relation between STEP and A2ARs.
Collapse
Affiliation(s)
- Maria Rosaria Domenici
- National Centre for Drug Research and Evaluation, Istituto Superiore di Sanitá, Rome, Italy
| | - Cinzia Mallozzi
- Department of Neuroscience, Istituto Superiore di Sanità, Rome, Italy
| | - Rita Pepponi
- National Centre for Drug Research and Evaluation, Istituto Superiore di Sanitá, Rome, Italy
| | - Ida Casella
- National Centre for Drug Research and Evaluation, Istituto Superiore di Sanitá, Rome, Italy
| | - Valentina Chiodi
- National Centre for Drug Research and Evaluation, Istituto Superiore di Sanitá, Rome, Italy
| | - Antonella Ferrante
- National Centre for Drug Research and Evaluation, Istituto Superiore di Sanitá, Rome, Italy
| | - Patrizia Popoli
- National Centre for Drug Research and Evaluation, Istituto Superiore di Sanitá, Rome, Italy
| |
Collapse
|
26
|
Bowie D. Neurotransmitter-gated ion channels, still front and centre stage. J Physiol 2021; 599:389-395. [PMID: 33448020 DOI: 10.1113/jp280800] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Affiliation(s)
- Derek Bowie
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec, H3G 1Y6, Canada
| |
Collapse
|
27
|
Zelek-Molik A, Bobula B, Gądek-Michalska A, Chorązka K, Bielawski A, Kuśmierczyk J, Siwiec M, Wilczkowski M, Hess G, Nalepa I. Psychosocial Crowding Stress-Induced Changes in Synaptic Transmission and Glutamate Receptor Expression in the Rat Frontal Cortex. Biomolecules 2021; 11:biom11020294. [PMID: 33669305 PMCID: PMC7920072 DOI: 10.3390/biom11020294] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 02/11/2021] [Accepted: 02/12/2021] [Indexed: 11/16/2022] Open
Abstract
This study demonstrates how exposure to psychosocial crowding stress (CS) for 3, 7, and 14 days affects glutamate synapse functioning and signal transduction in the frontal cortex (FC) of rats. CS effects on synaptic activity were evaluated in FC slices of the primary motor cortex (M1) by measuring field potential (FP) amplitude, paired-pulse ratio (PPR), and long-term potentiation (LTP). Protein expression of GluA1, GluN2B mGluR1a/5, VGLUT1, and VGLUT2 was assessed in FC by western blot. The body’s response to CS was evaluated by measuring body weight and the plasma level of plasma corticosterone (CORT), adrenocorticotropic hormone (ACTH), and interleukin 1 beta (IL1B). CS 3 14d increased FP and attenuated LTP in M1, while PPR was augmented in CS 14d. The expression of GluA1, GluN2B, and mGluR1a/5 was up-regulated in CS 3d and downregulated in CS 14d. VGLUTs expression tended to increase in CS 7d. The failure to blunt the effects of chronic CS on FP and LTP in M1 suggests the impairment of habituation mechanisms by psychosocial stressors. PPR augmented by chronic CS with increased VGLUTs level in the CS 7d indicates that prolonged CS exposure changed presynaptic signaling within the FC. The CS bidirectional profile of changes in glutamate receptors’ expression seems to be a common mechanism evoked by stress in the FC.
Collapse
Affiliation(s)
- Agnieszka Zelek-Molik
- Department of Brain Biochemistry, Maj Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, 31-343 Krakow, Poland; (K.C.); (A.B.); (J.K.); (M.W.); (I.N.)
- Correspondence: ; Tel.: +48-12-6623335
| | - Bartosz Bobula
- Department of Physiology, Maj Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, 31-343 Krakow, Poland; (B.B.); (A.G.-M.); (M.S.); (G.H.)
| | - Anna Gądek-Michalska
- Department of Physiology, Maj Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, 31-343 Krakow, Poland; (B.B.); (A.G.-M.); (M.S.); (G.H.)
| | - Katarzyna Chorązka
- Department of Brain Biochemistry, Maj Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, 31-343 Krakow, Poland; (K.C.); (A.B.); (J.K.); (M.W.); (I.N.)
| | - Adam Bielawski
- Department of Brain Biochemistry, Maj Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, 31-343 Krakow, Poland; (K.C.); (A.B.); (J.K.); (M.W.); (I.N.)
| | - Justyna Kuśmierczyk
- Department of Brain Biochemistry, Maj Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, 31-343 Krakow, Poland; (K.C.); (A.B.); (J.K.); (M.W.); (I.N.)
| | - Marcin Siwiec
- Department of Physiology, Maj Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, 31-343 Krakow, Poland; (B.B.); (A.G.-M.); (M.S.); (G.H.)
| | - Michał Wilczkowski
- Department of Brain Biochemistry, Maj Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, 31-343 Krakow, Poland; (K.C.); (A.B.); (J.K.); (M.W.); (I.N.)
| | - Grzegorz Hess
- Department of Physiology, Maj Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, 31-343 Krakow, Poland; (B.B.); (A.G.-M.); (M.S.); (G.H.)
| | - Irena Nalepa
- Department of Brain Biochemistry, Maj Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, 31-343 Krakow, Poland; (K.C.); (A.B.); (J.K.); (M.W.); (I.N.)
| |
Collapse
|
28
|
Adenosine A 2A receptor inhibition reduces synaptic and cognitive hippocampal alterations in Fmr1 KO mice. Transl Psychiatry 2021; 11:112. [PMID: 33547274 PMCID: PMC7864914 DOI: 10.1038/s41398-021-01238-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 12/28/2020] [Accepted: 01/18/2021] [Indexed: 02/07/2023] Open
Abstract
In fragile X syndrome (FXS) the lack of the fragile X mental retardation protein (FMRP) leads to exacerbated signaling through the metabotropic glutamate receptors 5 (mGlu5Rs). The adenosine A2A receptors (A2ARs), modulators of neuronal damage, could play a role in FXS. A synaptic colocalization and a strong permissive interaction between A2A and mGlu5 receptors in the hippocampus have been previously reported, suggesting that blocking A2ARs might normalize the mGlu5R-mediated effects of FXS. To study the cross-talk between A2A and mGlu5 receptors in the absence of FMRP, we performed extracellular electrophysiology experiments in hippocampal slices of Fmr1 KO mouse. The depression of field excitatory postsynaptic potential (fEPSPs) slope induced by the mGlu5R agonist CHPG was completely blocked by the A2AR antagonist ZM241385 and strongly potentiated by the A2AR agonist CGS21680, suggesting that the functional synergistic coupling between the two receptors could be increased in FXS. To verify if chronic A2AR blockade could reverse the FXS phenotypes, we treated the Fmr1 KO mice with istradefylline, an A2AR antagonist. We found that hippocampal DHPG-induced long-term depression (LTD), which is abnormally increased in FXS mice, was restored to the WT level. Furthermore, istradefylline corrected aberrant dendritic spine density, specific behavioral alterations, and overactive mTOR, TrkB, and STEP signaling in Fmr1 KO mice. Finally, we identified A2AR mRNA as a target of FMRP. Our results show that the pharmacological blockade of A2ARs partially restores some of the phenotypes of Fmr1 KO mice, both by reducing mGlu5R functioning and by acting on other A2AR-related downstream targets.
Collapse
|
29
|
Won S, Roche KW. Regulation of glutamate receptors by striatal-enriched tyrosine phosphatase 61 (STEP 61 ). J Physiol 2021; 599:443-451. [PMID: 32170729 PMCID: PMC11526339 DOI: 10.1113/jp278703] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 02/25/2020] [Indexed: 12/26/2022] Open
Abstract
Phosphorylation regulates glutamate receptor trafficking. The cytosolic C-terminal domains of both NMDA receptors (NMDARs) and AMPA receptors (AMPARs) have distinct motifs, which are substrates for serine/threonine and tyrosine phosphorylation. Decades of research have shown how phosphorylation of glutamate receptors mediates protein binding and receptor trafficking, ultimately controlling synaptic transmission and plasticity. STEP is a protein tyrosine phosphatase (also known as PTPN5), with several isoforms resulting from alternative splicing. Targets of STEP include a variety of important synaptic substrates, among which are the tyrosine kinase Fyn and glutamate receptors. In particular, STEP61 , the longest isoform, dephosphorylates the NMDAR subunit GluN2B and strongly regulates the expression of NMDARs at synapses. This interplay between STEP, Fyn and GluN2B-containing NMDARs has been characterized by multiple groups. More recently, STEP61 was shown to bind to AMPARs in a subunit-specific manner and differentially regulate synaptic NMDARs and AMPARs. Because of its many effects on synaptic proteins, STEP has been implicated in regulating excitatory synapses during plasticity and playing a role in synaptic dysfunction in a variety of neurological disorders. In this review, we will highlight the ways in which STEP61 differentially regulates NMDARs and AMPARs, as well as its role in plasticity and disease.
Collapse
Affiliation(s)
- Sehoon Won
- Receptor Biology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Katherine W Roche
- Receptor Biology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, 20892, USA
| |
Collapse
|
30
|
Ehinger Y, Morisot N, Phamluong K, Sakhai SA, Soneja D, Adrover MF, Alvarez VA, Ron D. cAMP-Fyn signaling in the dorsomedial striatum direct pathway drives excessive alcohol use. Neuropsychopharmacology 2021; 46:334-342. [PMID: 32417851 PMCID: PMC7852539 DOI: 10.1038/s41386-020-0712-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Accepted: 05/11/2020] [Indexed: 12/19/2022]
Abstract
Fyn kinase in the dorsomedial striatum (DMS) of rodents plays a central role in mechanisms underlying excessive alcohol intake. The DMS is comprised of medium spiny neurons (MSNs) that project directly (dMSNs) or indirectly (iMSNs) to the substantia nigra. Here, we examined the cell-type specificity of Fyn's actions in alcohol use. First, we knocked down Fyn selectively in DMS dMSNs or iMSNs of mice and measured the level of alcohol consumption. We found that downregulation of Fyn in dMSNs, but not in iMSNs, reduces excessive alcohol but not saccharin intake. D1Rs are coupled to Gαs/olf, which activate cAMP signaling. To examine whether Fyn's actions are mediated through cAMP signaling, DMS dMSNs were infected with GαsDREADD, and the activation of Fyn signaling was measured following CNO treatment. We found that remote stimulation of cAMP signaling in DMS dMSNs activates Fyn and promotes the phosphorylation of the Fyn substrate, GluN2B. In contract, remote activation of GαsDREADD in DLS dMSNs did not alter Fyn signaling. We then tested whether activation of GαsDREADD in DMS dMSNs or iMSNs alters alcohol intake and observed that CNO-dependent activation of GαsDREADD in DMS dMSNs but not iMSNs increases alcohol but not saccharin intake. Finally, we examined the contribution of Fyn to GαsDREADD-dependent increase in alcohol intake, and found that systemic administration of the Fyn inhibitor, AZD0503 blocks GαsDREADD-dependent increase in alcohol consumption. Our results suggest that the cAMP-Fyn axis in the DMS dMSNs is a molecular transducer of mechanisms underlying the development of excessive alcohol consumption.
Collapse
Affiliation(s)
- Yann Ehinger
- Department of Neurology, University of California San Francisco (UCSF), San Francisco, CA, 94143, USA
| | - Nadege Morisot
- Department of Neurology, University of California San Francisco (UCSF), San Francisco, CA, 94143, USA
- Nkarta Therapeutics, San Francisco, CA, USA
| | - Khanhky Phamluong
- Department of Neurology, University of California San Francisco (UCSF), San Francisco, CA, 94143, USA
| | - Samuel A Sakhai
- Department of Neurology, University of California San Francisco (UCSF), San Francisco, CA, 94143, USA
- Sage Therapeutics, San Francisco, CA, USA
| | - Drishti Soneja
- Department of Neurology, University of California San Francisco (UCSF), San Francisco, CA, 94143, USA
| | - Martin F Adrover
- National Institutes of Alcohol Abuse and Alcoholism, National Institute of Health, Bethesda, MD, 20892, USA
- INGEBI, CONICET, Buenos Aires, Argentina
| | - Veronica A Alvarez
- National Institutes of Alcohol Abuse and Alcoholism, National Institute of Health, Bethesda, MD, 20892, USA
- Center on Compulsive Behaviors, Intramural Research Program, National Institute of Health, Bethesda, MD, 20892, USA
| | - Dorit Ron
- Department of Neurology, University of California San Francisco (UCSF), San Francisco, CA, 94143, USA.
| |
Collapse
|
31
|
Zhao X, Zhou Z. Expression and Regulation of the GABA A Receptor/STEP61 Signaling Pathway in Cerebral Cortical Neurons Treated with Emulsified Isoflurane In Vitro. ACS Chem Neurosci 2020; 11:4329-4335. [PMID: 33232128 DOI: 10.1021/acschemneuro.0c00586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Emulsified isoflurane (EISO) is an intravenous anesthetic. However, researchers have not clearly determined how emulsified isoflurane affects the central nervous system during the process of anesthesia. The aim of this study was to explore changes in the gamma-aminobutyric acid type A receptor subunit (GABAA), 61 kD isoform of striatal-enriched protein phosphatase (STEP61) signaling pathway, and epigenetic regulation in cortical neurons after treatment with emulsified isoflurane. After immunological identification, the isolated neurons were randomly divided into three groups: the blank group (Con), intralipid treatment group (FE), and emulsified isoflurane treatment group (EISO). Neuron viability was assayed using cell counting kit-8 (CCK-8). The expression levels of target nucleic acids, proteins, and corresponding ligands were detected. Using real-time polymerase chain reaction (PCR) to assess the promoter methylation of ion channel proteins in the cerebral cortex of rats anesthetized with EISO, we observed changes in promoter methylation of the genes encoding gamma-aminobutyric acid type A receptor α1 subunit (GABAAα1), N-methyl-d-aspartate receptor subunit 1 (NMDAR1), and mu opioid receptor 1 (OPRM1), accompanied by changes in the levels of their messenger ribonucleic acids (mRNAs) and proteins. The levels of ligands for these receptors were also altered. EISO altered the methylation rate of the promoter region of channel protein-coding genes involved in the GABAA/STEP61 signaling pathway in cerebral cortical neurons to regulate gene expression. The ligands for the receptors were also changed.
Collapse
Affiliation(s)
- Xingkai Zhao
- Department of Veterinary Clinical Science, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, P. R. China
| | - Zhenlei Zhou
- Department of Veterinary Clinical Science, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, P. R. China
| |
Collapse
|
32
|
Chatterjee M, Singh P, Xu J, Lombroso PJ, Kurup PK. Inhibition of striatal-enriched protein tyrosine phosphatase (STEP) activity reverses behavioral deficits in a rodent model of autism. Behav Brain Res 2020; 391:112713. [PMID: 32461127 PMCID: PMC7346720 DOI: 10.1016/j.bbr.2020.112713] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Revised: 05/09/2020] [Accepted: 05/15/2020] [Indexed: 02/06/2023]
Abstract
Autism spectrum disorders (ASDs) are highly prevalent childhood illnesses characterized by impairments in communication, social behavior, and repetitive behaviors. Studies have found aberrant synaptic plasticity and neuronal connectivity during the early stages of brain development and have suggested that these contribute to an increased risk for ASD. STEP is a protein tyrosine phosphatase that regulates synaptic plasticity and is implicated in several cognitive disorders. Here we test the hypothesis that STEP may contribute to some of the aberrant behaviors present in the VPA-induced mouse model of ASD. In utero VPA exposure of pregnant dams results in autistic-like behavior in the pups, which is associated with a significant increase in the STEP expression in the prefrontal cortex. The elevated STEP protein levels are correlated with increased dephosphorylation of STEP substrates GluN2B, Pyk2 and ERK, suggesting upregulated STEP activity. Moreover, pharmacological inhibition of STEP rescues the sociability, repetitive and abnormal anxiety phenotypes commonly associated with ASD. These data suggest that STEP may play a role in the VPA model of ASD and STEP inhibition may have a potential therapeutic benefit in this model.
Collapse
Affiliation(s)
- Manavi Chatterjee
- Child Study Center, Yale University, 230 South Frontage Rd, New Haven, CT 06520, United States; Department of Pharmacology, Yale University, 333 Cedar Street, New Haven, CT 06520, United States.
| | - Priya Singh
- Child Study Center, Yale University, 230 South Frontage Rd, New Haven, CT 06520, United States
| | - Jian Xu
- Child Study Center, Yale University, 230 South Frontage Rd, New Haven, CT 06520, United States; Department of Psychiatry, Yale University, 333 Cedar Street, New Haven, CT 06520, United States
| | - Paul J Lombroso
- Child Study Center, Yale University, 230 South Frontage Rd, New Haven, CT 06520, United States; Department of Psychiatry, Yale University, 333 Cedar Street, New Haven, CT 06520, United States; Department of Neuroscience, Yale University, 333 Cedar Street, New Haven, CT 06520, United States
| | - Pradeep K Kurup
- Child Study Center, Yale University, 230 South Frontage Rd, New Haven, CT 06520, United States; Department of Surgery, University of Alabama at Birmingham, 1900 University Blvd, Birmingham, AL 35233, United States.
| |
Collapse
|
33
|
Zhao X, Chang G, Cheng Y, Zhou Z. GABA A Receptor/STEP61 Signaling Pathway May Be Involved in Emulsified Isoflurane Anesthesia in Rats. Int J Mol Sci 2020; 21:E4078. [PMID: 32517358 PMCID: PMC7312199 DOI: 10.3390/ijms21114078] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2020] [Revised: 06/04/2020] [Accepted: 06/05/2020] [Indexed: 11/17/2022] Open
Abstract
(1) Background: Emulsified isoflurane (EISO) is a type of intravenous anesthetic. How emulsified isoflurane works in the brain is still unclear. The aim of this study was to explore whether epigenetic mechanisms affect anesthesia and to evaluate the anesthetic effects of emulsified isoflurane in rats. (2) Methods: Rats were randomly divided into four groups (n = 8/group): The tail vein was injected with normal saline 0.1 mL·kg-1·min-1for the control (Con) group, with intralipid for the fat emulsion (FE) group, with EISO at 60 mg·kg-1·min-1 for the high-concentration (HD) group, and 45 mg·kg-1·min-1 for the low-concentration (LD) group. The consciousness state, motor function of limbs, and response to nociceptive stimulus were observed after drug administration. (3) Results: Using real-time polymerase chain reaction (PCR) to assess the promoter methylation of ion channel proteins in the cerebral cortex of rats anesthetized by EISO, we demonstrated that the change in the promoters' methylation of the coding genes for gamma-aminobutyric acid A receptor α1 subunit (GABAAα1), N-methyl-D-aspartate receptor subunit 1 (NMDAR1), and mu opioid receptor 1 (OPRM1) was accompanied by the change in messenger ribonucleic acid (mRNA) and protein expression by these genes. (4) Conclusion: These data suggest that the epigenetic factors' modulation might offer a novel approach to explore the anesthetic mechanism of EISO.
Collapse
Affiliation(s)
| | | | | | - Zhenlei Zhou
- Department of Veterinary Clinical Science, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; (X.Z.); (G.C.); (Y.C.)
| |
Collapse
|
34
|
Quan C, Wang S, Duan K, Ma J, Yu H, Yang M, Hu N, Long G, Zeng G, Huang Z. The role of kynurenine pathway and kynurenic aminotransferase alleles in postpartum depression following cesarean section in Chinese women. Brain Behav 2020; 10:e01566. [PMID: 32101387 PMCID: PMC7177593 DOI: 10.1002/brb3.1566] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 01/08/2020] [Accepted: 01/13/2020] [Indexed: 02/03/2023] Open
Abstract
OBJECTIVES A growing body of data indicates that the kynurenine pathway may play a role in the pathogenesis of postpartum depressive symptoms (PDS). Kynurenic aminotransferase (KAT) is an important kynurenine pathway enzyme, catalyzing kynurenine (KYN) into kynurenic acid (KYNA). This study investigated as to whether genetic variations in KAT are associated with PDS. METHODS A cohort of 360 Chinese women scheduled to undergo cesarean delivery was enrolled into this study. PDS was determined by an Edinburgh Postnatal Depression Scale (EPDS) score ≥ 13. A total of eight KAT single nucleotide polymorphisms (SNPs) were genotyped and their association with PDS investigated. Serum concentrations of KYN, KYNA, and quinolinic acid (QUIN) in women with or without PDS were also measured. This allowed the determination of the KYNA/KYN ratio, which is reflective of KAT activity. RESULTS Postpartum depressive symptoms incidence was 7.2%. Advanced maternal age, lower education, antenatal depression, and postpartum blues were risk factors for PDS (p < .05). Women with PDS, versus non-PDS, had heightened KYN levels one day prior to surgery (ante-d1) (p < .05), as well as having significantly lower KYNA and higher QUIN levels at postnatal day three (post-d3) (p < .05). Women with, versus without, PDS also had a significantly higher QUIN/KYNA ratio at post-d3 (p < .05). KAT activity was significantly lower in women with, versus without, PDS at ante-d3 (p < .05). No significant association was evident between the KAT SNPs and PDS. CONCLUSION Our data support a role for alterations in the kynurenine pathway in the pathogenesis of PDS, although no significant association was found for the eight tested KAT SNPs with PDS.
Collapse
Affiliation(s)
- Chengxuan Quan
- Department of Anesthesia, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Saiying Wang
- Department of Anesthesia, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Kaiming Duan
- Department of Anesthesia, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Jiahui Ma
- Department of Anesthesia, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Heya Yu
- Department of Anesthesia, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Mi Yang
- Department of Anesthesia, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Na Hu
- Department of Anesthesia, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Ge Long
- Department of Anesthesia, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Guang Zeng
- Department of Anesthesia, Changsha Taihe Hospital, Changsha, China
| | - Zhendong Huang
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
35
|
Kulikova EA, Fursenko DV, Bazhenova EY, Kulikov AV. Pargyline and р-Chlorophenylalanine Decrease Expression of Ptpn5 Encoding Striatal-Enriched Protein Tyrosine Phosphatase (STEP) in the Mouse Striatum. Mol Biol 2020. [DOI: 10.1134/s0026893320020090] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
36
|
Mossa A, Manzini MC. Molecular causes of sex-specific deficits in rodent models of neurodevelopmental disorders. J Neurosci Res 2019; 99:37-56. [PMID: 31872500 PMCID: PMC7754327 DOI: 10.1002/jnr.24577] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Revised: 11/02/2019] [Accepted: 12/09/2019] [Indexed: 12/12/2022]
Abstract
Neurodevelopmental disorders (NDDs) such as intellectual disability and autism spectrum disorder consistently show a male bias in prevalence, but it remains unclear why males and females are affected with different frequency. While many behavioral studies of transgenic NDD models have focused only on males, the requirement by the National Institutes of Health to consider sex as a biological variable has promoted the comparison of male and female performance in wild-type and mutant animals. Here, we review examples of rodent models of NDDs in which sex-specific deficits were identified in molecular, physiological, and/or behavioral responses, showing sex differences in susceptibility to disruption of genes mutated in NDDs. Haploinsufficiency in genes involved in mechanisms such as synaptic function (GABRB3 and NRXN1), chromatin remodeling (CHD8, EMHT1, and ADNP), and intracellular signaling (CC2D1A and ERK1) lead to more severe behavioral outcomes in males. However, in the absence of behavioral deficits, females can still present with cellular and electrophysiological changes that could be due to compensatory mechanisms or differential allocation of molecular and cellular functions in the two sexes. By contrasting these findings with mouse models where females are more severely affected (MTHFR and AMBRA1), we propose a framework to approach the study of sex-specific deficits possibly leading to sex bias in NDDs.
Collapse
Affiliation(s)
- Adele Mossa
- Child Health Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - M Chiara Manzini
- Child Health Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA.,Department of Neuroscience and Cell Biology, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| |
Collapse
|
37
|
Yasuda RP. Adenosine STEPs on synaptic function: An Editorial for 'The activity of the STriatal-enriched protein tyrosine phosphatase in neuronal cells is modulated by adenosine A2A receptor on' page 284. J Neurochem 2019; 152:270-272. [PMID: 31724181 DOI: 10.1111/jnc.14901] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 10/16/2019] [Accepted: 10/19/2019] [Indexed: 12/13/2022]
Abstract
This is an Editorial Highlight of a manuscript by Mallozzi et al. (2019) in the current issue of the Journal of Neurochemistry, in which the authors detail the biochemical pathway that leads to synaptic depression by cocaine. This pathway requires the adenosine A2A receptor and STEP phosphatases. Activation of the adenosine A2A receptor leads to an increase in intracellular calcium, activation of STEP by dephosphorylation, inhibition of excitatory ionotropic glutamate receptors by dephosphorylation of phospho-tyrosine residues and subsequent internalization of the ionotropic glutamate receptors. This adenosine A2A receptor pathway could lead to potential drug targets for neurologic and neuropsychiatric disorders.
Collapse
Affiliation(s)
- Robert P Yasuda
- Department of Pharmacology & Physiology, Georgetown University, Washington, DC, USA
| |
Collapse
|
38
|
Kulikov A, Sinyakova N, Kulikova E, Khomenko T, Salakhutdinov N, Kulikov V, Volcho K. Effects of Acute and Chronic Treatment of Novel Psychotropic Drug, 8- (Trifluoromethyl)-1, 2, 3, 4, 5-benzopentathiepin-6-amine Hydrochloride (TC-2153), on the Behavior of Zebrafish (Danio Rerio): A Comparison with Fluoxetine. LETT DRUG DES DISCOV 2019. [DOI: 10.2174/1570180816666190221162952] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Background:
Striatal-enriched Tyrosine Phosphatase (STEP) plays a key role in the
mechanisms of neuronal signaling and is a potential molecular target for new generation of
psychotropic drugs. STEP inhibitor, 8-(trifluoromethyl-1,2,3,4,5-benzopentathiepin-6-amine
hydrochloride (TC-2153), shows anxiolytic effect on mice. Zebrafish (Danio rerio) is a suitable
model for the study of anxiety pharmacology.
Objective:
The objective of this study is to investigate the effects of acute and chronic TC-2153
treatment on zebrafish anxiety-related behavior.
Methods:
The effects of acute (0.125 and 0.25 mg/l, 3 h) and chronic (0.125 mg/l, 14 days)
administration of TC-2153 on locomotion and anxiety-related behavior (time spent near the bottom
and mean distance from the bottom) of adult zebrafish in the Novel Tank (NT) test were compared
with those of the same doses of fluoxetine chosen as a positive control.
Results:
Acute treatment with 0.125 mg/l and 0.25 mg/l of TC-2153 or fluoxetine decreased time
spent near the bottom, increased time spent near the surface and increased mean distance from the
bottom of tank. Chronic treatment with 0.125 mg/l of TC-2153 reduced only time spent near the
tank bottom without any effect on time spent near the surface and mean distance from the bottom,
while chronic administration of 0.125 mg/l of fluoxetine altered these three indices of anxiety.
Conclusion:
Both acute and chronic TC-2153 produces anxiety-like effect indicating STEP
involved in the mechanism of anxiety-related behavior in zebrafish. At the same time, chronic
treatment with TC-2153 reduced locomotor activity. Zebrafish is a promising laboratory object to
study the role of STEP in the nervous system.
Collapse
Affiliation(s)
- Alexander Kulikov
- Federal Research Center Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, Novosibirsk 630090, Russian Federation
| | - Nadezhda Sinyakova
- Federal Research Center Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, Novosibirsk 630090, Russian Federation
| | - Elizabeth Kulikova
- Federal Research Center Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, Novosibirsk 630090, Russian Federation
| | - Tatyana Khomenko
- Novosibirsk Institute of Organic Chemistry, Siberian Branch of Russian Academy of Sciences, Novosibirsk 630090, Russian Federation
| | - Nariman Salakhutdinov
- Novosibirsk Institute of Organic Chemistry, Siberian Branch of Russian Academy of Sciences, Novosibirsk 630090, Russian Federation
| | - Victor Kulikov
- Institute of Automation and Electrometry, Siberian Branch of Russian Academy of Sciences, Novosibirsk 630090, Russian Federation
| | - Konstantin Volcho
- Novosibirsk Institute of Organic Chemistry, Siberian Branch of Russian Academy of Sciences, Novosibirsk 630090, Russian Federation
| |
Collapse
|
39
|
Zong MM, Yuan HM, He X, Zhou ZQ, Qiu XD, Yang JJ, Ji MH. Disruption of Striatal-Enriched Protein Tyrosine Phosphatase Signaling Might Contribute to Memory Impairment in a Mouse Model of Sepsis-Associated Encephalopathy. Neurochem Res 2019; 44:2832-2842. [DOI: 10.1007/s11064-019-02905-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2019] [Revised: 04/01/2019] [Accepted: 10/29/2019] [Indexed: 02/07/2023]
|
40
|
Mallozzi C, Pepponi R, Visentin S, Chiodi V, Lombroso PJ, Bader M, Popoli P, Domenici MR. The activity of the Striatal-enriched protein tyrosine phosphatase in neuronal cells is modulated by adenosine A 2A receptor. J Neurochem 2019; 152:284-298. [PMID: 31520531 DOI: 10.1111/jnc.14866] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Revised: 07/30/2019] [Accepted: 09/01/2019] [Indexed: 12/15/2022]
Abstract
We recently demonstrated that a tonic activation of adenosine A2A receptors (A2A Rs) is required for cocaine-induced synaptic depression and increase in the activity of STriatal-Enriched protein tyrosine Phosphatase (STEP). In this study, we elaborated on the relationship between A2A R and STEP using genetic, pharmacological, and cellular tools. We found that the activities of protein tyrosine phosphatases (PTPs), and in particular of STEP, are significantly increased in the striatum and hippocampus of a transgenic rat strain over-expressing the neuronal A2A R (NSEA2A ) with respect to wild-type (WT) rats. Moreover the selective A2A R agonist 4-[2-[[6-Amino-9-(N-ethyl-β-d-ribofuranuronamidosyl)-9H-purin-2-yl]amino]ethyl]benzenepropanoic acid hydrochloride up-regulates PTPs and STEP activities in WT but not in NSEA2A rats, while the selective A2A R antagonist 4-(-2-[7-amino-2-{2-furyl}{1,2,4}triazolo{2,3-a} {1,3,5}triazin-5-yl-amino]ethyl)phenol restores the tyrosine phosphatase activities in NSEA2A , having no effects in WT rats. In addition, while cocaine induced the activation of PTP and STEP in WT rats, it failed to increase phosphatase activity in NSEA2A rats. A2A Rs modulate STEP activity also in the SH-SY5Y neuroblastoma cell line, where a calcium-dependent calcineurin/PP1 pathway was found to play a major role. In summary, the present study identified a novel interaction between A2A R and STEP that could have important clinical implications, since STEP has emerged as key regulator of signaling pathways involved in neurodegenerative and neuropsychiatric diseases and A2A Rs are considered a promising target for the development of therapeutic strategies for different diseases of the central nervous system. Read the Editorial Highlight for this article on page 270.
Collapse
Affiliation(s)
- Cinzia Mallozzi
- Department of Neuroscience, Istituto Superiore di Sanità, Rome, Italy
| | - Rita Pepponi
- National Centre for Drug Research and Evaluation, Istituto Superiore di Sanità, Rome, Italy
| | - Sergio Visentin
- National Centre for Drug Research and Evaluation, Istituto Superiore di Sanità, Rome, Italy
| | - Valentina Chiodi
- National Centre for Drug Research and Evaluation, Istituto Superiore di Sanità, Rome, Italy
| | - Paul J Lombroso
- Child Study Center, Yale School of Medicine, New Haven, CT, USA
| | - Michael Bader
- Max-Delbrűck-Center for Molecular Medicine, Berlin, Germany
| | - Patrizia Popoli
- National Centre for Drug Research and Evaluation, Istituto Superiore di Sanità, Rome, Italy
| | - Maria Rosaria Domenici
- National Centre for Drug Research and Evaluation, Istituto Superiore di Sanità, Rome, Italy
| |
Collapse
|
41
|
Adenosine A2A receptor as potential therapeutic target in neuropsychiatric disorders. Pharmacol Res 2019; 147:104338. [DOI: 10.1016/j.phrs.2019.104338] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 06/26/2019] [Accepted: 07/01/2019] [Indexed: 01/20/2023]
|
42
|
Zafarullah M, Tassone F. Molecular Biomarkers in Fragile X Syndrome. Brain Sci 2019; 9:E96. [PMID: 31035599 PMCID: PMC6562871 DOI: 10.3390/brainsci9050096] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Revised: 04/22/2019] [Accepted: 04/24/2019] [Indexed: 01/01/2023] Open
Abstract
Fragile X syndrome (FXS) is the most common inherited form of intellectual disability (ID) and a known monogenic cause of autism spectrum disorder (ASD). It is a trinucleotide repeat disorder, in which more than 200 CGG repeats in the 5' untranslated region (UTR) of the fragile X mental retardation 1 (FMR1) gene causes methylation of the promoter with consequent silencing of the gene, ultimately leading to the loss of the encoded fragile X mental retardation 1 protein, FMRP. FMRP is an RNA binding protein that plays a primary role as a repressor of translation of various mRNAs, many of which are involved in the maintenance and development of neuronal synaptic function and plasticity. In addition to intellectual disability, patients with FXS face several behavioral challenges, including anxiety, hyperactivity, seizures, repetitive behavior, and problems with executive and language performance. Currently, there is no cure or approved medication for the treatment of the underlying causes of FXS, but in the past few years, our knowledge about the proteins and pathways that are dysregulated by the loss of FMRP has increased, leading to clinical trials and to the path of developing molecular biomarkers for identifying potential targets for therapies. In this paper, we review candidate molecular biomarkers that have been identified in preclinical studies in the FXS mouse animal model and are now under validation for human applications or have already made their way to clinical trials.
Collapse
Affiliation(s)
- Marwa Zafarullah
- Department of Biochemistry and Molecular Medicine, University of California Davis, School of Medicine, Sacramento, 95817 CA, USA.
| | - Flora Tassone
- Department of Biochemistry and Molecular Medicine, University of California Davis, School of Medicine, Sacramento, 95817 CA, USA.
- MIND Institute, University of California Davis Medical Center, Sacramento, 95817 CA, USA.
| |
Collapse
|
43
|
The STEP 61 interactome reveals subunit-specific AMPA receptor binding and synaptic regulation. Proc Natl Acad Sci U S A 2019; 116:8028-8037. [PMID: 30936304 DOI: 10.1073/pnas.1900878116] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Striatal-enriched protein tyrosine phosphatase (STEP) is a brain-specific protein phosphatase that regulates a variety of synaptic proteins, including NMDA receptors (NAMDRs). To better understand STEP's effect on other receptors, we used mass spectrometry to identify the STEP61 interactome. We identified a number of known interactors, but also ones including the GluA2 subunit of AMPA receptors (AMPARs). We show that STEP61 binds to the C termini of GluA2 and GluA3 as well as endogenous AMPARs in hippocampus. The synaptic expression of GluA2 and GluA3 is increased in STEP-KO mouse brain, and STEP knockdown in hippocampal slices increases AMPAR-mediated synaptic currents. Interestingly, STEP61 overexpression reduces the synaptic expression and synaptic currents of both AMPARs and NMDARs. Furthermore, STEP61 regulation of synaptic AMPARs is mediated by lysosomal degradation. Thus, we report a comprehensive list of STEP61 binding partners, including AMPARs, and reveal a central role for STEP61 in differentially organizing synaptic AMPARs and NMDARs.
Collapse
|
44
|
Khotskin NV, Plyusnina AV, Kulikova EA, Bazhenova EY, Fursenko DV, Sorokin IE, Kolotygin I, Mormede P, Terenina EE, Shevelev OB, Kulikov AV. On association of the lethal yellow (A) mutation in the agouti gene with the alterations in mouse brain and behavior. Behav Brain Res 2019; 359:446-456. [DOI: 10.1016/j.bbr.2018.11.013] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Revised: 11/08/2018] [Accepted: 11/08/2018] [Indexed: 10/27/2022]
|
45
|
Blázquez G, Castañé A, Saavedra A, Masana M, Alberch J, Pérez-Navarro E. Social Memory and Social Patterns Alterations in the Absence of STriatal-Enriched Protein Tyrosine Phosphatase. Front Behav Neurosci 2019; 12:317. [PMID: 30760987 PMCID: PMC6362413 DOI: 10.3389/fnbeh.2018.00317] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Accepted: 12/04/2018] [Indexed: 01/23/2023] Open
Abstract
STriatal-Enriched protein tyrosine Phosphatase (STEP) is a neural-specific protein that opposes the development of synaptic strengthening and whose levels are altered in several neurodegenerative and psychiatric disorders. Since STEP is expressed in brain regions implicated in social behavior, namely the striatum, the CA2 region of the hippocampus, cortex and amygdala, here we investigated whether social memory and social patterns were altered in STEP knockout (KO) mice. Our data robustly demonstrated that STEP KO mice presented specific social memory impairment as indicated by the three-chamber sociability test, the social discrimination test, the 11-trial habituation/dishabituation social recognition test, and the novel object recognition test (NORT). This affectation was not related to deficiencies in the detection of social olfactory cues, altered sociability or anxiety levels. However, STEP KO mice showed lower exploratory activity, reduced interaction time with an intruder, less dominant behavior and higher immobility time in the tail suspension test than controls, suggesting alterations in motivation. Moreover, the extracellular levels of dopamine (DA), but not serotonin (5-HT), were increased in the dorsal striatum of STEP KO mice. Overall, our results indicate that STEP deficiency disrupts social memory and other social behaviors as well as DA homeostasis in the dorsal striatum.
Collapse
Affiliation(s)
- Gloria Blázquez
- Departament de Biomedicina, Facultat de Medicina i Ciències de la Salut, Institut de Neurociències, Universitat de Barcelona, Barcelona, Spain.,Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Barcelona, Spain
| | - Anna Castañé
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.,Department of Neurochemistry and Neuropharmacology, CSIC-Institut d'Investigacions Biomèdiques de Barcelona (IIBB), Barcelona, Spain.,Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Barcelona, Spain
| | - Ana Saavedra
- Departament de Biomedicina, Facultat de Medicina i Ciències de la Salut, Institut de Neurociències, Universitat de Barcelona, Barcelona, Spain.,Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Barcelona, Spain
| | - Mercè Masana
- Departament de Biomedicina, Facultat de Medicina i Ciències de la Salut, Institut de Neurociències, Universitat de Barcelona, Barcelona, Spain.,Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Barcelona, Spain
| | - Jordi Alberch
- Departament de Biomedicina, Facultat de Medicina i Ciències de la Salut, Institut de Neurociències, Universitat de Barcelona, Barcelona, Spain.,Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Barcelona, Spain
| | - Esther Pérez-Navarro
- Departament de Biomedicina, Facultat de Medicina i Ciències de la Salut, Institut de Neurociències, Universitat de Barcelona, Barcelona, Spain.,Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Barcelona, Spain
| |
Collapse
|
46
|
Tautermann CS, Binder F, Büttner FH, Eickmeier C, Fiegen D, Gross U, Grundl MA, Heilker R, Hobson S, Hoerer S, Luippold A, Mack V, Montel F, Peters S, Bhattacharya S, Vaidehi N, Schnapp G, Thamm S, Zeeb M. Allosteric Activation of Striatal-Enriched Protein Tyrosine Phosphatase (STEP, PTPN5) by a Fragment-like Molecule. J Med Chem 2018; 62:306-316. [PMID: 30207464 DOI: 10.1021/acs.jmedchem.8b00857] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Protein tyrosine phosphatase non-receptor type 5 (PTPN5, STEP) is a brain specific phosphatase that regulates synaptic function and plasticity by modulation of N-methyl-d-aspartate receptor (NMDAR) and α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor (AMPAR) trafficking. Dysregulation of STEP has been linked to neurodegenerative and neuropsychiatric diseases, highlighting this enzyme as an attractive therapeutic target for drug discovery. Selective targeting of STEP with small molecules has been hampered by high conservation of the active site among protein tyrosine phosphatases. We report the discovery of the first small molecule allosteric activator for STEP that binds to the phosphatase domain. Allosteric binding is confirmed by both X-ray and 15N NMR experiments, and specificity has been demonstrated by an enzymatic test cascade. Molecular dynamics simulations indicate stimulation of enzymatic activity by a long-range allosteric mechanism. To allow the scientific community to make use of this tool, we offer to provide the compound in the course of an open innovation initiative.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | - Supriyo Bhattacharya
- Department of Molecular Immunology , Beckman Research Institute of the City of Hope , 1500, E. Duarte Road , Duarte , California 91010 , United States
| | - Nagarajan Vaidehi
- Department of Molecular Immunology , Beckman Research Institute of the City of Hope , 1500, E. Duarte Road , Duarte , California 91010 , United States
| | | | | | | |
Collapse
|
47
|
Activation of Phosphotyrosine-Mediated Signaling Pathways in the Cortex and Spinal Cord of SOD1 G93A, a Mouse Model of Familial Amyotrophic Lateral Sclerosis. Neural Plast 2018; 2018:2430193. [PMID: 30154836 PMCID: PMC6098854 DOI: 10.1155/2018/2430193] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Revised: 06/06/2018] [Accepted: 07/05/2018] [Indexed: 12/12/2022] Open
Abstract
Degeneration of cortical and spinal motor neurons is the typical feature of amyotrophic lateral sclerosis (ALS), a progressive neurodegenerative disease for which a pathogenetic role for the Cu/Zn superoxide dismutase (SOD1) has been demonstrated. Mice overexpressing a mutated form of the SOD1 gene (SOD1G93A) develop a syndrome that closely resembles the human disease. The SOD1 mutations confer to this enzyme a “gain-of-function,” leading to increased production of reactive oxygen species. Several oxidants induce tyrosine phosphorylation through direct stimulation of kinases and/or phosphatases. In this study, we analyzed the activities of src and fyn tyrosine kinases and of protein tyrosine phosphatases in synaptosomal fractions prepared from the motor cortex and spinal cord of transgenic mice expressing SOD1G93A. We found that (i) protein phosphotyrosine level is increased, (ii) src and fyn activities are upregulated, and (iii) the activity of tyrosine phosphatases, including the striatal-enriched tyrosine phosphatase (STEP), is significantly decreased. Moreover, the NMDA receptor (NMDAR) subunit GluN2B tyrosine phosphorylation was upregulated in SOD1G93A. Tyrosine phosphorylation of GluN2B subunits regulates the NMDAR function and the recruitment of downstream signaling molecules. Indeed, we found that proline-rich tyrosine kinase 2 (Pyk2) and ERK1/2 kinase are upregulated in SOD1G93A mice. These results point out an involvement of tyrosine kinases and phosphatases in the pathogenesis of ALS.
Collapse
|
48
|
Yang L, Bai HH, Zhang ZY, Liu JP, Suo ZW, Yang X, Hu XD. Disruption of SHP1/NMDA receptor signaling in spinal cord dorsal horn alleviated inflammatory pain. Neuropharmacology 2018; 137:104-113. [DOI: 10.1016/j.neuropharm.2018.04.029] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Revised: 04/08/2018] [Accepted: 04/27/2018] [Indexed: 10/17/2022]
|
49
|
Saavedra A, Ballesteros JJ, Tyebji S, Martínez-Torres S, Blázquez G, López-Hidalgo R, Azkona G, Alberch J, Martín ED, Pérez-Navarro E. Proteolytic Degradation of Hippocampal STEP 61 in LTP and Learning. Mol Neurobiol 2018; 56:1475-1487. [PMID: 29948948 DOI: 10.1007/s12035-018-1170-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Accepted: 05/31/2018] [Indexed: 10/14/2022]
Abstract
Striatal-enriched protein tyrosine phosphatase (STEP) modulates key signaling molecules involved in synaptic plasticity and neuronal function. It is postulated that STEP opposes the development of long-term potentiation (LTP) and that it exerts a restraint on long-term memory (LTM). Here, we examined whether STEP61 levels are regulated during hippocampal LTP and after training in hippocampal-dependent tasks. We found that after inducing LTP by high frequency stimulation or theta-burst stimulation STEP61 levels were significantly reduced, with a concomitant increase of STEP33 levels, a product of calpain cleavage. Importantly, inhibition of STEP with TC-2153 improved LTP in hippocampal slices. Moreover, we observed that after training in the passive avoidance and the T-maze spontaneous alternation task, hippocampal STEP61 levels were significantly reduced, but STEP33 levels were unchanged. Yet, hippocampal BDNF content and TrkB levels were increased in trained mice, and it is known that BDNF promotes STEP degradation through the proteasome. Accordingly, hippocampal pTrkBTyr816, pPLCγTyr783, and protein ubiquitination levels were increased in T-SAT trained mice. Remarkably, injection of the TrkB antagonist ANA-12 (2 mg/Kg, but not 0.5 mg/Kg) elicited LTM deficits and promoted STEP61 accumulation in the hippocampus. Also, STEP knockout mice outperformed wild-type animals in an age- and test-dependent manner. Summarizing, STEP61 undergoes proteolytic degradation in conditions leading to synaptic strengthening and memory formation, thus highlighting its role as a molecular constrain, which is removed to enable the activation of pathways important for plasticity processes.
Collapse
Affiliation(s)
- Ana Saavedra
- Departament de Biomedicina, Facultat de Medicina i Ciències de la Salut, Institut de Neurociències, Universitat de Barcelona, C/ Casanova, 143 08036, Barcelona, Catalonia, Spain.,Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Catalonia, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Jesús J Ballesteros
- Institute for Research in Neurological Disabilities (IDINE), University of Castilla-La Mancha, Albacete, Spain.,Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.,The Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Shiraz Tyebji
- Departament de Biomedicina, Facultat de Medicina i Ciències de la Salut, Institut de Neurociències, Universitat de Barcelona, C/ Casanova, 143 08036, Barcelona, Catalonia, Spain.,Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Catalonia, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.,Infection and Immunity Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Melbourne, Australia
| | - Sara Martínez-Torres
- Departament de Biomedicina, Facultat de Medicina i Ciències de la Salut, Institut de Neurociències, Universitat de Barcelona, C/ Casanova, 143 08036, Barcelona, Catalonia, Spain.,Departament de Ciències Experimentals i de la Salut, Universitat Pompeu Fabra, Barcelona, Catalonia, Spain
| | - Gloria Blázquez
- Departament de Biomedicina, Facultat de Medicina i Ciències de la Salut, Institut de Neurociències, Universitat de Barcelona, C/ Casanova, 143 08036, Barcelona, Catalonia, Spain.,Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Catalonia, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Rosa López-Hidalgo
- Institute for Research in Neurological Disabilities (IDINE), University of Castilla-La Mancha, Albacete, Spain.,Grupo de Patología Celular y Molecular del Alcohol, Centro de Investigación Príncipe Felipe, Valencia, Spain
| | - Garikoitz Azkona
- Departament de Biomedicina, Facultat de Medicina i Ciències de la Salut, Institut de Neurociències, Universitat de Barcelona, C/ Casanova, 143 08036, Barcelona, Catalonia, Spain.,Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Catalonia, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Jordi Alberch
- Departament de Biomedicina, Facultat de Medicina i Ciències de la Salut, Institut de Neurociències, Universitat de Barcelona, C/ Casanova, 143 08036, Barcelona, Catalonia, Spain.,Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Catalonia, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Eduardo D Martín
- Institute for Research in Neurological Disabilities (IDINE), University of Castilla-La Mancha, Albacete, Spain.,Instituto Cajal, CSIC, Madrid, Spain
| | - Esther Pérez-Navarro
- Departament de Biomedicina, Facultat de Medicina i Ciències de la Salut, Institut de Neurociències, Universitat de Barcelona, C/ Casanova, 143 08036, Barcelona, Catalonia, Spain. .,Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Catalonia, Spain. .,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.
| |
Collapse
|
50
|
Duan KM, Ma JH, Wang SY, Huang Z, Zhou Y, Yu H. The role of tryptophan metabolism in postpartum depression. Metab Brain Dis 2018; 33:647-660. [PMID: 29307018 DOI: 10.1007/s11011-017-0178-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Accepted: 12/26/2017] [Indexed: 01/01/2023]
Abstract
The Postpartum depression (PPD) is the most common postpartum psychiatric disorder, afflicting approximately 10%-20% of new mothers. Clinical symptoms of the PPD include depressive disorder, agitation, insomnia, anxiety and confusion, resulting in an increase in suicidal tendencies, thereby having significant impacts on the puerpera, newborn and their family. A growing body of data indicate a role for alterations in tryptophan metabolism in the PPD. The metabolism of tryptophan produces an array of crucial factors that can differentially regulate key physiological processes linked to the PPD. Importantly, an increase in stress hormones and immune-inflammatory activity drives tryptophan to the production of neuroregulatory kynurenine pathway products and away from the serotonin and melatonin pathways. This links the PPD to other disorders of depressed mood, which are classically associated with decreased serotonin and melatonin, coupled to increases in kynurenine pathway products. Several kynurenine pathway products, such as kynurenic acid and quinolinic acid, can have neuroregulatory effects, with consequences pathological underpinnings of the PPD. The current article reviews the role of alterations in tryptophan metabolism in the PPD.
Collapse
Affiliation(s)
- Kai-Ming Duan
- Department of Anesthesiology, Third Xiangya Hospital of Central South University, Changsha, 410013, People's Republic of China
| | - Jia-Hui Ma
- Department of Anesthesiology, Third Xiangya Hospital of Central South University, Changsha, 410013, People's Republic of China
| | - Sai-Ying Wang
- Department of Anesthesiology, Third Xiangya Hospital of Central South University, Changsha, 410013, People's Republic of China.
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, 410008, People's Republic of China.
- Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha, 410078, People's Republic of China.
- Hunan Province Cooperation Innovation Center for Molecular Target New Drug Study, Hengyang, 421001, People's Republic of China.
| | - ZhengDong Huang
- Department of Anesthesiology, Third Xiangya Hospital of Central South University, Changsha, 410013, People's Republic of China
| | - YingYong Zhou
- Department of Anesthesiology, Third Xiangya Hospital of Central South University, Changsha, 410013, People's Republic of China
| | - HeYa Yu
- Department of Anesthesiology, Third Xiangya Hospital of Central South University, Changsha, 410013, People's Republic of China
| |
Collapse
|