1
|
Heo K, Ho TSY, Zeng X, Turnes BL, Arab M, Jayakar S, Chen K, Kimourtzis G, Condro MC, Fazzari E, Song X, Tabitha Hees J, Xu Z, Chen X, Barrett LB, Perrault L, Pandey R, Zhang K, Bhaduri A, He Z, Kornblum HI, Hubbs J, Woolf CJ. Non-muscle myosin II inhibition at the site of axon injury increases axon regeneration. Nat Commun 2025; 16:2975. [PMID: 40140393 PMCID: PMC11947156 DOI: 10.1038/s41467-025-58303-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 03/14/2025] [Indexed: 03/28/2025] Open
Abstract
Motor axon regeneration following peripheral nerve injury is critical for motor recovery but therapeutic interventions enhancing this are not available. We conduct a phenotypic screen on human motor neurons and identified blebbistatin, a non-muscle myosin II inhibitor, as the most effective neurite outgrowth promotor. Despite its efficacy in vitro, its poor bioavailability limits in vivo application. We, therefore, utilize a blebbistatin analog, NMIIi2, to explore its therapeutic potential for promoting axon regeneration. Local NMIIi2 application directly to injured axons enhances regeneration in human motor neurons. Furthermore, following a sciatic nerve crush injury in male mice, local NMIIi2 administration to the axonal injury site facilitates motor neuron regeneration, muscle reinnervation, and functional recovery. NMIIi2 also promotes axon regeneration in sensory, cortical, and retinal ganglion neurons. These findings highlight the therapeutic potential of topical NMII inhibition for treating axon damage.
Collapse
Affiliation(s)
- Keunjung Heo
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Tammy Szu-Yu Ho
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Xiangsunze Zeng
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Bruna Lenfers Turnes
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Maryam Arab
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Selwyn Jayakar
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Kuchuan Chen
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Georgios Kimourtzis
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Michael C Condro
- Intellectual and Developmental Disabilities Research Center and the Departments of Psychiatry, Pharmacology and Pediatrics, University of California, Los Angeles, CA, USA
| | - Elisa Fazzari
- Department of Biological Chemistry, University of California, Los Angeles, CA, USA
| | - Xuan Song
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA
- Department of Neurology and Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - J Tabitha Hees
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA
| | - Zhuqiu Xu
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Xirui Chen
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Lee B Barrett
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Laura Perrault
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA
| | - Roshan Pandey
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Kathleen Zhang
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA
| | - Aparna Bhaduri
- Department of Biological Chemistry, University of California, Los Angeles, CA, USA
| | - Zhigang He
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA
- Department of Neurology and Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Harley I Kornblum
- Intellectual and Developmental Disabilities Research Center and the Departments of Psychiatry, Pharmacology and Pediatrics, University of California, Los Angeles, CA, USA
| | - Jed Hubbs
- Rosamund Stone Zander Translational Neuroscience Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Clifford J Woolf
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA.
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
2
|
Zhao H, Li H, Meng L, Du P, Mo X, Gong M, Chen J, Liao Y. Disrupting heroin-associated memory reconsolidation through actin polymerization inhibition in the nucleus accumbens core. Int J Neuropsychopharmacol 2024; 28:pyae065. [PMID: 39716383 DOI: 10.1093/ijnp/pyae065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Accepted: 12/23/2024] [Indexed: 12/25/2024] Open
Abstract
BACKGROUND Understanding drug addiction as a disorder of maladaptive learning, where drug-associated or environmental cues trigger drug cravings and seeking, is crucial for developing effective treatments. Actin polymerization, a biochemical process, plays a crucial role in drug-related memory formation, particularly evident in conditioned place preference paradigms involving drugs like morphine and methamphetamine. However, the role of actin polymerization in the reconsolidation of heroin-associated memories remains understudied. METHODS This study employed a rodent model of self-administered heroin to investigate the involvement of actin polymerization in the reconsolidation of heroin-associated memories. Rats underwent ten days of intravenous heroin self-administration paired with conditioned cues. Subsequently, a 10-day extinction phase aimed to reduce heroin-seeking behaviors. Following this, rats participated in a 15-minute retrieval trial with or without cues. Immediately post-retrieval, rats received bilateral injections of the actin polymerization inhibitor Latrunculin A (Lat A) into the nucleus accumbens core (NACc), a critical brain region for memory reconsolidation. RESULTS Immediate administration of Lat A into the NACc post-retrieval significantly reduced cue-induced and heroin-primed reinstatement of heroin-seeking behavior for at least 28 days. However, administering Lat A 6-hour post-retrieval or without a retrieval trial, as well as administering Jasplakionlide prior to memory reactivation did not affect heroin-seeking behaviors. CONCLUSIONS Inhibiting actin polymerization during the reconsolidation window disrupts heroin-associated memory reconsolidation, leading to decreased heroin-seeking behavior and prevention of relapse. These effects are contingent upon the presence of a retrieval trial and exhibit temporal specificity, shedding light on addiction mechanisms and potential therapeutic interventions.
Collapse
Affiliation(s)
- Haiting Zhao
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Haoyu Li
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Li Meng
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Department of Radiology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Peng Du
- Department of Neurosurgery, The Second Affiliated Hospital, Xinjiang Medical University, Urumqi, China
| | - Xin Mo
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Mengqi Gong
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jiaxin Chen
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yiwei Liao
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
3
|
Radnai L, Young EJ, Kikuti C, Hafenbreidel M, Stremel RF, Lin L, Toth K, Pasetto P, Jin X, Patel A, Conlon M, Briggs S, Heidsieck L, Sweeney HL, Sellers J, Krieger-Burke T, Martin WH, Sisco J, Young S, Pearson P, Rumbaugh G, Araldi GL, Duddy SK, Cameron MD, Surman M, Houdusse A, Griffin PR, Kamenecka TM, Miller CA. Development of Clinically Viable Non-Muscle Myosin II Small Molecule Inhibitors with Broad Therapeutic Potential. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.07.617018. [PMID: 39416074 PMCID: PMC11482808 DOI: 10.1101/2024.10.07.617018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Non-muscle myosin II (NMII), a molecular motor that regulates critical processes such as cytokinesis and neuronal synaptic plasticity, has substantial therapeutic potential. However, translating this potential to in vivo use has been hampered by the lack of selective tools. The most prototypical non-selective inhibitor, blebbistatin inactivates both NMII and cardiac myosin II (CMII), a key regulator of heart function. Using rational drug design, we developed a series of NMII inhibitors that improve tolerability by selectively targeting NMII over CMII, including MT-228, which has excellent properties such as high brain penetration and efficacy in preclinical models of stimulant use disorder, which has no current FDA-approved therapies. The structure of MT-228 bound to myosin II provides insight into its 17-fold selectivity for NMII over CMII. MT-228's broad therapeutic window opens the door to new disease treatments and provides valuable tools for the scientific community, along with promising leads for future medication development. Highlights Research suggests numerous indications, from axon regeneration and cancer, would benefit from a small molecule inhibitor of non-muscle myosin II, a molecular motor that regulates the actin cytoskeleton. Current chemical probe options are very limited and lack sufficient safety for in vivo studies, which we show is primarily due to potent inhibition of cardiac myosin II.Rational design that focused on improving target selectivity over the pan-myosin II inhibitor, blebbistatin, led to the identification of MT-228, a small molecule inhibitor with a wide therapeutic window.High-resolution structure of MT-228 bound to myosin II reveals that selectivity results from a different positioning compared to blebbistatin and an important sequence difference between cardiac and non-muscle myosin II in the inhibitor binding pocket.A single administration of MT-228 shows long-lasting efficacy in animal models of stimulant use disorder, a current unmet and rapidly escalating need with no FDA-approved treatments.
Collapse
|
4
|
Young EJ, Radnai L, Prikhodko V, Miller CA. Novel therapeutics in development for the treatment of stimulant-use disorder. Curr Opin Neurobiol 2024; 87:102898. [PMID: 39096558 DOI: 10.1016/j.conb.2024.102898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Revised: 07/14/2024] [Accepted: 07/14/2024] [Indexed: 08/05/2024]
Abstract
Misuse and accidental overdoses attributed to stimulants are escalating rapidly. These stimulants include methamphetamine, cocaine, amphetamine, ecstasy-type drugs, and prescription stimulants such as methylphenidate. Unlike opioids and alcohol, there are no therapies approved by the US Food and Drug Administration (FDA) to treat stimulant-use disorder. The high rate of relapse among this population highlights the insufficiency of current treatment options, which are limited to abstinence support programs and behavioral modification therapies. Here, we briefly outline recent regulatory actions taken by FDA to help support the development of new stimulant use disorder treatments and highlight several new therapeutics in the clinical development pipeline.
Collapse
Affiliation(s)
- Erica J Young
- Department of Molecular Medicine, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL, USA; Department of Neuroscience, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL, USA; Myosin Therapeutics, Jupiter, FL, USA
| | - Laszlo Radnai
- Department of Molecular Medicine, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL, USA; Department of Neuroscience, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL, USA
| | | | - Courtney A Miller
- Department of Molecular Medicine, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL, USA; Department of Neuroscience, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL, USA.
| |
Collapse
|
5
|
Chinthalapudi K, Heissler SM. Structure, regulation, and mechanisms of nonmuscle myosin-2. Cell Mol Life Sci 2024; 81:263. [PMID: 38878079 PMCID: PMC11335295 DOI: 10.1007/s00018-024-05264-6] [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: 03/11/2024] [Revised: 04/24/2024] [Accepted: 04/30/2024] [Indexed: 06/23/2024]
Abstract
Members of the myosin superfamily of molecular motors are large mechanochemical ATPases that are implicated in an ever-expanding array of cellular functions. This review focuses on mammalian nonmuscle myosin-2 (NM2) paralogs, ubiquitous members of the myosin-2 family of filament-forming motors. Through the conversion of chemical energy into mechanical work, NM2 paralogs remodel and shape cells and tissues. This process is tightly controlled in time and space by numerous synergetic regulation mechanisms to meet cellular demands. We review how recent advances in structural biology together with elegant biophysical and cell biological approaches have contributed to our understanding of the shared and unique mechanisms of NM2 paralogs as they relate to their kinetics, regulation, assembly, and cellular function.
Collapse
Affiliation(s)
- Krishna Chinthalapudi
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University College of Medicine, Columbus, OH, 43210, USA
| | - Sarah M Heissler
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University College of Medicine, Columbus, OH, 43210, USA.
| |
Collapse
|
6
|
Mitra A, Deats SP, Dickson PE, Zhu J, Gardin J, Nieman BJ, Henkelman RM, Tsai NP, Chesler EJ, Zhang ZW, Kumar V. Tmod2 Is a Regulator of Cocaine Responses through Control of Striatal and Cortical Excitability and Drug-Induced Plasticity. J Neurosci 2024; 44:e1389232024. [PMID: 38508714 PMCID: PMC11063827 DOI: 10.1523/jneurosci.1389-23.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 02/12/2024] [Accepted: 02/24/2024] [Indexed: 03/22/2024] Open
Abstract
Drugs of abuse induce neuroadaptations, including synaptic plasticity, that are critical for transition to addiction, and genes and pathways that regulate these neuroadaptations are potential therapeutic targets. Tropomodulin 2 (Tmod2) is an actin-regulating gene that plays an important role in synapse maturation and dendritic arborization and has been implicated in substance abuse and intellectual disability in humans. Here, we mine the KOMP2 data and find that Tmod2 knock-out mice show emotionality phenotypes that are predictive of addiction vulnerability. Detailed addiction phenotyping shows that Tmod2 deletion does not affect the acute locomotor response to cocaine administration. However, sensitized locomotor responses are highly attenuated in these knock-outs, indicating perturbed drug-induced plasticity. In addition, Tmod2 mutant animals do not self-administer cocaine indicating lack of hedonic responses to cocaine. Whole-brain MR imaging shows differences in brain volume across multiple regions, although transcriptomic experiments did not reveal perturbations in gene coexpression networks. Detailed electrophysiological characterization of Tmod2 KO neurons showed increased spontaneous firing rate of early postnatal and adult cortical and striatal neurons. Cocaine-induced synaptic plasticity that is critical for sensitization is either missing or reciprocal in Tmod2 KO nucleus accumbens shell medium spiny neurons, providing a mechanistic explanation of the cocaine response phenotypes. Combined, these data, collected from both males and females, provide compelling evidence that Tmod2 is a major regulator of plasticity in the mesolimbic system and regulates the reinforcing and addictive properties of cocaine.
Collapse
Affiliation(s)
| | | | | | - Jiuhe Zhu
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801
| | | | - Brian J Nieman
- Mouse Imaging Centre and Translational Medicine, Hospital for Sick Children; Ontario Institute for Cancer Research; Department of Medical Biophysics, University of Toronto, Toronto, Ontario M5T 3H7, Canada
| | - R Mark Henkelman
- Mouse Imaging Centre and Translational Medicine, Hospital for Sick Children; Ontario Institute for Cancer Research; Department of Medical Biophysics, University of Toronto, Toronto, Ontario M5T 3H7, Canada
| | - Nien-Pei Tsai
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801
| | | | | | - Vivek Kumar
- The Jackson Laboratory, Bar Harbor, Maine 04609
| |
Collapse
|
7
|
Pandey S, Miller CA. Targeting the cytoskeleton as a therapeutic approach to substance use disorders. Pharmacol Res 2024; 202:107143. [PMID: 38499081 PMCID: PMC11034636 DOI: 10.1016/j.phrs.2024.107143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/01/2024] [Revised: 03/06/2024] [Accepted: 03/12/2024] [Indexed: 03/20/2024]
Abstract
Substance use disorders (SUD) are chronic relapsing disorders governed by continually shifting cycles of positive drug reward experiences and drug withdrawal-induced negative experiences. A large body of research points to plasticity within systems regulating emotional, motivational, and cognitive processes as drivers of continued compulsive pursuit and consumption of substances despite negative consequences. This plasticity is observed at all levels of analysis from molecules to networks, providing multiple avenues for intervention in SUD. The cytoskeleton and its regulatory proteins within neurons and glia are fundamental to the structural and functional integrity of brain processes and are potentially the major drivers of the morphological and behavioral plasticity associated with substance use. In this review, we discuss preclinical studies that provide support for targeting the brain cytoskeleton as a therapeutic approach to SUD. We focus on the interplay between actin cytoskeleton dynamics and exposure to cocaine, methamphetamine, alcohol, opioids, and nicotine and highlight preclinical studies pointing to a wide range of potential therapeutic targets, such as nonmuscle myosin II, Rac1, cofilin, prosapip 1, and drebrin. These studies broaden our understanding of substance-induced plasticity driving behaviors associated with SUD and provide new research directions for the development of SUD therapeutics.
Collapse
Affiliation(s)
- Surya Pandey
- Department of Molecular Medicine, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL 33458, United States; Department of Neuroscience, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL 33458, United States
| | - Courtney A Miller
- Department of Molecular Medicine, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL 33458, United States; Department of Neuroscience, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL 33458, United States.
| |
Collapse
|
8
|
Hafenbreidel M, Pandey S, Briggs SB, Arza M, Bonthu S, Fisher C, Tiller A, Hall AB, Reed S, Mayorga N, Lin L, Khan S, Cameron MD, Rumbaugh G, Miller CA. Basolateral amygdala corticotropin releasing factor receptor 2 interacts with nonmuscle myosin II to destabilize memory in males. Neurobiol Learn Mem 2023; 206:107865. [PMID: 37995804 DOI: 10.1016/j.nlm.2023.107865] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 10/24/2023] [Accepted: 11/20/2023] [Indexed: 11/25/2023]
Abstract
Preclinical studies show that inhibiting the actin motor ATPase nonmuscle myosin II (NMII) with blebbistatin (Blebb) in the basolateral amgydala (BLA) depolymerizes actin, resulting in an immediate, retrieval-independent disruption of methamphetamine (METH)-associated memory in male and female adult and adolescent rodents. The effect is highly selective, as NMII inhibition has no effect in other relevant brain regions (e.g., dorsal hippocampus [dPHC], nucleus accumbens [NAc]), nor does it interfere with associations for other aversive or appetitive stimuli, including cocaine (COC). To understand the mechanisms responsible for drug specific selectivity we began by investigating, in male mice, the pharmacokinetic differences in METH and COC brain exposure . Replicating METH's longer half-life with COC did not render the COC association susceptible to disruption by NMII inhibition. Therefore, we next assessed transcriptional differences. Comparative RNA-seq profiling in the BLA, dHPC and NAc following METH or COC conditioning identified crhr2, which encodes the corticotropin releasing factor receptor 2 (CRF2), as uniquely upregulated by METH in the BLA. CRF2 antagonism with Astressin-2B (AS2B) had no effect on METH-associated memory after consolidation, allowing for determination of CRF2 influences on NMII-based susceptibility. Pretreatment with AS2B prevented the ability of Blebb to disrupt an established METH-associated memory. Alternatively, combining CRF2 overexpression and agonist treatment, urocortin 3 (UCN3), in the BLA during conditioning rendered COC-associated memory susceptible to disruption by NMII inhibition, mimicking the Blebb-induced, retrieval-independent memory disruption seen with METH. These results suggest that BLA CRF2 receptor activation during memory formation in male mice can prevent stabilization of the actin-myosin cytoskeleton supporting the memory, rendering it vulnerable to disruption by NMII inhibition. CRF2 represents an interesting target for BLA-dependent memory destabilization via downstream effects on NMII.
Collapse
Affiliation(s)
- Madalyn Hafenbreidel
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, FL 33458, United States; Department of Neuroscience, The Scripps Research Institute, Jupiter, FL 33458, United States; The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL 33458, United States
| | - Surya Pandey
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, FL 33458, United States; Department of Neuroscience, The Scripps Research Institute, Jupiter, FL 33458, United States; The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL 33458, United States
| | - Sherri B Briggs
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, FL 33458, United States; Department of Neuroscience, The Scripps Research Institute, Jupiter, FL 33458, United States; The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL 33458, United States
| | - Meghana Arza
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, FL 33458, United States; Department of Neuroscience, The Scripps Research Institute, Jupiter, FL 33458, United States; The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL 33458, United States
| | - Shalakha Bonthu
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, FL 33458, United States; Department of Neuroscience, The Scripps Research Institute, Jupiter, FL 33458, United States; The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL 33458, United States
| | - Cadence Fisher
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, FL 33458, United States; Department of Neuroscience, The Scripps Research Institute, Jupiter, FL 33458, United States; The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL 33458, United States
| | - Annika Tiller
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, FL 33458, United States; Department of Neuroscience, The Scripps Research Institute, Jupiter, FL 33458, United States; The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL 33458, United States
| | - Alice B Hall
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, FL 33458, United States; Department of Neuroscience, The Scripps Research Institute, Jupiter, FL 33458, United States; The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL 33458, United States
| | - Shayna Reed
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, FL 33458, United States; Department of Neuroscience, The Scripps Research Institute, Jupiter, FL 33458, United States; The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL 33458, United States
| | - Natasha Mayorga
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, FL 33458, United States; Department of Neuroscience, The Scripps Research Institute, Jupiter, FL 33458, United States; The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL 33458, United States
| | - Li Lin
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, FL 33458, United States; The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL 33458, United States
| | - Susan Khan
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, FL 33458, United States; The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL 33458, United States
| | - Michael D Cameron
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, FL 33458, United States; The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL 33458, United States
| | - Gavin Rumbaugh
- Department of Neuroscience, The Scripps Research Institute, Jupiter, FL 33458, United States; The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL 33458, United States
| | - Courtney A Miller
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, FL 33458, United States; Department of Neuroscience, The Scripps Research Institute, Jupiter, FL 33458, United States; The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL 33458, United States.
| |
Collapse
|
9
|
Hafenbreidel M, Briggs SB, Arza M, Bonthu S, Fisher C, Tiller A, Hall AB, Reed S, Mayorga N, Lin L, Khan S, Cameron MD, Rumbaugh G, Miller CA. Basolateral Amygdala Corticotrophin Releasing Factor Receptor 2 Interacts with Nonmuscle Myosin II to Destabilize Memory. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.22.541732. [PMID: 37292925 PMCID: PMC10245849 DOI: 10.1101/2023.05.22.541732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Inhibiting the actin motor ATPase nonmuscle myosin II (NMII) with blebbistatin (Blebb) in the basolateral amgydala (BLA) depolymerizes actin, resulting in an immediate, retrieval-independent disruption of methamphetamine (METH)-associated memory. The effect is highly selective, as NMII inhibition has no effect in other relevant brain regions (e.g. dorsal hippocampus [dPHC], nucleus accumbens [NAc]), nor does it interfere with associations for other aversive or appetitive stimuli, including cocaine (COC). To investigate a potential source of this specificity, pharmacokinetic differences in METH and COC brain exposure were examined. Replicating METH's longer half-life with COC did not render the COC association susceptible to disruption by NMII inhibition. Therefore, transcriptional differences were next assessed. Comparative RNA-seq profiling in the BLA, dHPC and NAc following METH or COC conditioning identified crhr2, which encodes the corticotrophin releasing factor receptor 2 (CRF2), as uniquely upregulated by METH in the BLA. CRF2 antagonism with Astressin-2B (AS2B) had no effect on METH-associated memory after consolidation, allowing for determination of CRF2 influences on NMII-based susceptibility after METH conditioning. Pretreatment with AS2B occluded the ability of Blebb to disrupt an established METH-associated memory. Alternatively, the Blebb-induced, retrieval-independent memory disruption seen with METH was mimicked for COC when combined with CRF2 overexpression in the BLA and its ligand, UCN3 during conditioning. These results indicate that BLA CRF2 receptor activation during learning can prevent stabilization of the actin-myosin cytoskeleton supporting the memory, rendering it vulnerable to disruption via NMII inhibition. CRF2 represents an interesting target for BLA-dependent memory destabilization via downstream effects on NMII.
Collapse
Affiliation(s)
- Madalyn Hafenbreidel
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, FL, 33458
- Department of Neuroscience, The Scripps Research Institute, Jupiter, FL, 33458
- Present address: Department of Molecular Medicine, Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL, 33458
- Present address: Department of Neuroscience, Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology Jupiter, FL, 33458
| | - Sherri B Briggs
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, FL, 33458
- Department of Neuroscience, The Scripps Research Institute, Jupiter, FL, 33458
- Present address: Department of Molecular Medicine, Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL, 33458
- Present address: Department of Neuroscience, Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology Jupiter, FL, 33458
| | - Meghana Arza
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, FL, 33458
- Department of Neuroscience, The Scripps Research Institute, Jupiter, FL, 33458
- Present address: Department of Molecular Medicine, Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL, 33458
- Present address: Department of Neuroscience, Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology Jupiter, FL, 33458
| | - Shalakha Bonthu
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, FL, 33458
- Department of Neuroscience, The Scripps Research Institute, Jupiter, FL, 33458
- Present address: Department of Molecular Medicine, Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL, 33458
- Present address: Department of Neuroscience, Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology Jupiter, FL, 33458
| | - Cadence Fisher
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, FL, 33458
- Department of Neuroscience, The Scripps Research Institute, Jupiter, FL, 33458
- Present address: Department of Molecular Medicine, Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL, 33458
- Present address: Department of Neuroscience, Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology Jupiter, FL, 33458
| | - Annika Tiller
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, FL, 33458
- Department of Neuroscience, The Scripps Research Institute, Jupiter, FL, 33458
- Present address: Department of Molecular Medicine, Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL, 33458
- Present address: Department of Neuroscience, Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology Jupiter, FL, 33458
- Present address: Department of Physiology and Neuroscience, Medical University of South Carolina, Charleston, SC, 29464
| | - Alice B Hall
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, FL, 33458
- Department of Neuroscience, The Scripps Research Institute, Jupiter, FL, 33458
- Present address: Department of Molecular Medicine, Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL, 33458
- Present address: Department of Neuroscience, Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology Jupiter, FL, 33458
| | - Shayna Reed
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, FL, 33458
- Department of Neuroscience, The Scripps Research Institute, Jupiter, FL, 33458
- Present address: Department of Molecular Medicine, Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL, 33458
- Present address: Department of Neuroscience, Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology Jupiter, FL, 33458
| | - Natasha Mayorga
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, FL, 33458
- Department of Neuroscience, The Scripps Research Institute, Jupiter, FL, 33458
- Present address: Department of Molecular Medicine, Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL, 33458
- Present address: Department of Neuroscience, Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology Jupiter, FL, 33458
| | - Li Lin
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, FL, 33458
- Present address: Department of Molecular Medicine, Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL, 33458
| | - Susan Khan
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, FL, 33458
- Present address: Department of Molecular Medicine, Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL, 33458
| | - Michael D Cameron
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, FL, 33458
- Present address: Department of Molecular Medicine, Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL, 33458
| | - Gavin Rumbaugh
- Department of Neuroscience, The Scripps Research Institute, Jupiter, FL, 33458
- Present address: Department of Neuroscience, Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology Jupiter, FL, 33458
| | - Courtney A Miller
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, FL, 33458
- Department of Neuroscience, The Scripps Research Institute, Jupiter, FL, 33458
- Present address: Department of Molecular Medicine, Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL, 33458
- Present address: Department of Neuroscience, Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology Jupiter, FL, 33458
| |
Collapse
|
10
|
Motz CT, Kabat V, Saxena T, Bellamkonda RV, Zhu C. Neuromechanobiology: An Expanding Field Driven by the Force of Greater Focus. Adv Healthc Mater 2021; 10:e2100102. [PMID: 34342167 PMCID: PMC8497434 DOI: 10.1002/adhm.202100102] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Revised: 07/06/2021] [Indexed: 12/14/2022]
Abstract
The brain processes information by transmitting signals through highly connected and dynamic networks of neurons. Neurons use specific cellular structures, including axons, dendrites and synapses, and specific molecules, including cell adhesion molecules, ion channels and chemical receptors to form, maintain and communicate among cells in the networks. These cellular and molecular processes take place in environments rich of mechanical cues, thus offering ample opportunities for mechanical regulation of neural development and function. Recent studies have suggested the importance of mechanical cues and their potential regulatory roles in the development and maintenance of these neuronal structures. Also suggested are the importance of mechanical cues and their potential regulatory roles in the interaction and function of molecules mediating the interneuronal communications. In this review, the current understanding is integrated and promising future directions of neuromechanobiology are suggested at the cellular and molecular levels. Several neuronal processes where mechanics likely plays a role are examined and how forces affect ligand binding, conformational change, and signal induction of molecules key to these neuronal processes are indicated, especially at the synapse. The disease relevance of neuromechanobiology as well as therapies and engineering solutions to neurological disorders stemmed from this emergent field of study are also discussed.
Collapse
Affiliation(s)
- Cara T Motz
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, 30332-0363, USA
- Parker H. Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA, 30332-0363, USA
| | - Victoria Kabat
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, 30332-0363, USA
| | - Tarun Saxena
- Department of Biomedical Engineering, Duke University, Durham, NC, 27709, USA
| | - Ravi V Bellamkonda
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC, 27708, USA
| | - Cheng Zhu
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, 30332-0363, USA
- Parker H. Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA, 30332-0363, USA
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, 30332-0363, USA
| |
Collapse
|
11
|
Chen L, Yan H, Wang Y, He Z, Leng Q, Huang S, Wu F, Feng X, Yan J. The Mechanisms and Boundary Conditions of Drug Memory Reconsolidation. Front Neurosci 2021; 15:717956. [PMID: 34421529 PMCID: PMC8377231 DOI: 10.3389/fnins.2021.717956] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 07/20/2021] [Indexed: 11/13/2022] Open
Abstract
Drug addiction can be seen as a disorder of maladaptive learning characterized by relapse. Therefore, disrupting drug-related memories could be an approach to improving therapies for addiction. Pioneering studies over the last two decades have revealed that consolidated memories are not static, but can be reconsolidated after retrieval, thereby providing candidate pathways for the treatment of addiction. The limbic-corticostriatal system is known to play a vital role in encoding the drug memory engram. Specific structures within this system contribute differently to the process of memory reconsolidation, making it a potential target for preventing relapse. In addition, as molecular processes are also active during memory reconsolidation, amnestic agents can be used to attenuate drug memory. In this review, we focus primarily on the brain structures involved in storing the drug memory engram, as well as the molecular processes involved in drug memory reconsolidation. Notably, we describe reports regarding boundary conditions constraining the therapeutic potential of memory reconsolidation. Furthermore, we discuss the principles that could be employed to modify stored memories. Finally, we emphasize the challenge of reconsolidation-based strategies, but end with an optimistic view on the development of reconsolidation theory for drug relapse prevention.
Collapse
Affiliation(s)
- Liangpei Chen
- Department of Forensic Science, School of Basic Medical Science, Central South University, Changsha, China
| | - He Yan
- Department of Forensic Science, School of Basic Medical Science, Central South University, Changsha, China
| | - Yufang Wang
- Department of Forensic Science, School of Basic Medical Science, Central South University, Changsha, China
| | - Ziping He
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Qihao Leng
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Shihao Huang
- Key Laboratory of Molecular Epidemiology of Hunan Province, School of Medicine, Hunan Normal University, Changsha, China
| | - Feilong Wu
- Key Laboratory of Molecular Epidemiology of Hunan Province, School of Medicine, Hunan Normal University, Changsha, China
| | - Xiangyang Feng
- Department of Forensic Science, School of Basic Medical Science, Xinjiang Medical University, Urumqi, China
| | - Jie Yan
- Department of Forensic Science, School of Basic Medical Science, Central South University, Changsha, China.,Department of Forensic Science, School of Basic Medical Science, Xinjiang Medical University, Urumqi, China
| |
Collapse
|
12
|
Oyigeya M. Reflex memory theory of acquired involuntary motor and sensory disorders. THE EGYPTIAN JOURNAL OF NEUROLOGY, PSYCHIATRY AND NEUROSURGERY 2021. [DOI: 10.1186/s41983-021-00307-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Explicit and implicit memories are conserved but flexible biological tools that nature uses to regulate the daily behaviors of human beings. An aberrant form of the implicit memory is presumed to exist and may be contributory to the pathophysiology of disorders such as tardive syndromes, phantom phenomena, flashback, posttraumatic stress disorders (PTSD), and related disorders. These disorders have posed significant clinical problems for both patients and physicians for centuries. All extant pathophysiological theories of these disorders have failed to provide basis for effective treatment.
Objective
The objective of this article is to propose an alternative pathophysiological theory that will hopefully lead to new treatment approaches.
Methods
The author sourced over 60 journal articles that treated topics on memory, and involuntary motor and sensory disorders, from open access journals using Google Scholar, and reviewed them and this helped in the formulation of this theory.
Results
From the reviews, the author thinks physical or chemical insult to the nervous system can cause defective circuit remodeling, leading to generation of a variant of implicit (automatic) memory, herein called “reflex memory” and this is encoded interoceptively to contribute to these phenomena states.
Conclusion
Acquired involuntary motor and sensory disorders are caused by defective circuit remodeling involving multiple neural mechanisms. Dysregulation of excitatory neurotransmitters, calcium overload, homeostatic failure, and neurotoxicity are implicated in the process. Sustained effects of these defective mechanisms are encoded interoceptively as abnormal memory in the neurons and the conscious manifestations are these disorders. Extant theories failed to recognize this possibility.
Collapse
|
13
|
Gyimesi M, Rauscher AÁ, Suthar SK, Hamow KÁ, Oravecz K, Lőrincz I, Borhegyi Z, Déri MT, Kiss ÁF, Monostory K, Szabó PT, Nag S, Tomasic I, Krans J, Tierney PJ, Kovács M, Kornya L, Málnási-Csizmadia A. Improved Inhibitory and Absorption, Distribution, Metabolism, Excretion, and Toxicology (ADMET) Properties of Blebbistatin Derivatives Indicate That Blebbistatin Scaffold Is Ideal for drug Development Targeting Myosin-2. J Pharmacol Exp Ther 2021; 376:358-373. [PMID: 33468641 DOI: 10.1124/jpet.120.000167] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 10/07/2020] [Indexed: 11/22/2022] Open
Abstract
Blebbistatin, para-nitroblebbistatin (NBleb), and para-aminoblebbistatin (AmBleb) are highly useful tool compounds as they selectively inhibit the ATPase activity of myosin-2 family proteins. Despite the medical importance of the myosin-2 family as drug targets, chemical optimization has not yet provided a promising lead for drug development because previous structure-activity-relationship studies were limited to a single myosin-2 isoform. Here we evaluated the potential of blebbistatin scaffold for drug development and found that D-ring substitutions can fine-tune isoform specificity, absorption-distribution-metabolism-excretion, and toxicological properties. We defined the inhibitory properties of NBleb and AmBleb on seven different myosin-2 isoforms, which revealed an unexpected potential for isoform specific inhibition. We also found that NBleb metabolizes six times slower than blebbistatin and AmBleb in rats, whereas AmBleb metabolizes two times slower than blebbistatin and NBleb in human, and that AmBleb accumulates in muscle tissues. Moreover, mutagenicity was also greatly reduced in case of AmBleb. These results demonstrate that small substitutions have beneficial functional and pharmacological consequences, which highlight the potential of the blebbistatin scaffold for drug development targeting myosin-2 family proteins and delineate a route for defining the chemical properties of further derivatives to be developed. SIGNIFICANCE STATEMENT: Small substitutions on the blebbistatin scaffold have beneficial functional and pharmacological consequences, highlighting their potential in drug development targeting myosin-2 family proteins.
Collapse
Affiliation(s)
- Máté Gyimesi
- Department of Biochemistry, Eötvös Loránd University, Budapest and Martonvásár, Hungary (M.G., K.O., I.L., Z.B., M.K., A.M.-C.); MTA-ELTE Motor Pharmacology Research Group, Budapest, Hungary (M.G., M.K., A.M.-C.); Motorharma Ltd., Budapest, Hungary (A.Á.R.); Printnet Ltd., Budapest, Hungary (S.K.S., I.L.); Plant Protection Institute, Centre for Agricultural Research, Martonvásár, Hungary (K.Á.H.); Metabolic Drug Interactions Research Group, Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary (M.T.D., Á.F.K., K.M.); Research Centre for Natural Sciences, Instrumentation Center, MS Metabolomic Research Laboratory, Budapest, Hungary (P.T.S.); Department of Biology, MyoKardia Inc., Brisbane, California (S.N., I.T.); Department of Neuroscience, Western New England University, Springfield, Massachusetts (J.K., P.J.T.); and Central Hospital of Southern Pest, National Institute of Hematology and Infectious Diseases, Budapest, Hungary (L.K.)
| | - Anna Á Rauscher
- Department of Biochemistry, Eötvös Loránd University, Budapest and Martonvásár, Hungary (M.G., K.O., I.L., Z.B., M.K., A.M.-C.); MTA-ELTE Motor Pharmacology Research Group, Budapest, Hungary (M.G., M.K., A.M.-C.); Motorharma Ltd., Budapest, Hungary (A.Á.R.); Printnet Ltd., Budapest, Hungary (S.K.S., I.L.); Plant Protection Institute, Centre for Agricultural Research, Martonvásár, Hungary (K.Á.H.); Metabolic Drug Interactions Research Group, Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary (M.T.D., Á.F.K., K.M.); Research Centre for Natural Sciences, Instrumentation Center, MS Metabolomic Research Laboratory, Budapest, Hungary (P.T.S.); Department of Biology, MyoKardia Inc., Brisbane, California (S.N., I.T.); Department of Neuroscience, Western New England University, Springfield, Massachusetts (J.K., P.J.T.); and Central Hospital of Southern Pest, National Institute of Hematology and Infectious Diseases, Budapest, Hungary (L.K.)
| | - Sharad Kumar Suthar
- Department of Biochemistry, Eötvös Loránd University, Budapest and Martonvásár, Hungary (M.G., K.O., I.L., Z.B., M.K., A.M.-C.); MTA-ELTE Motor Pharmacology Research Group, Budapest, Hungary (M.G., M.K., A.M.-C.); Motorharma Ltd., Budapest, Hungary (A.Á.R.); Printnet Ltd., Budapest, Hungary (S.K.S., I.L.); Plant Protection Institute, Centre for Agricultural Research, Martonvásár, Hungary (K.Á.H.); Metabolic Drug Interactions Research Group, Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary (M.T.D., Á.F.K., K.M.); Research Centre for Natural Sciences, Instrumentation Center, MS Metabolomic Research Laboratory, Budapest, Hungary (P.T.S.); Department of Biology, MyoKardia Inc., Brisbane, California (S.N., I.T.); Department of Neuroscience, Western New England University, Springfield, Massachusetts (J.K., P.J.T.); and Central Hospital of Southern Pest, National Institute of Hematology and Infectious Diseases, Budapest, Hungary (L.K.)
| | - Kamirán Á Hamow
- Department of Biochemistry, Eötvös Loránd University, Budapest and Martonvásár, Hungary (M.G., K.O., I.L., Z.B., M.K., A.M.-C.); MTA-ELTE Motor Pharmacology Research Group, Budapest, Hungary (M.G., M.K., A.M.-C.); Motorharma Ltd., Budapest, Hungary (A.Á.R.); Printnet Ltd., Budapest, Hungary (S.K.S., I.L.); Plant Protection Institute, Centre for Agricultural Research, Martonvásár, Hungary (K.Á.H.); Metabolic Drug Interactions Research Group, Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary (M.T.D., Á.F.K., K.M.); Research Centre for Natural Sciences, Instrumentation Center, MS Metabolomic Research Laboratory, Budapest, Hungary (P.T.S.); Department of Biology, MyoKardia Inc., Brisbane, California (S.N., I.T.); Department of Neuroscience, Western New England University, Springfield, Massachusetts (J.K., P.J.T.); and Central Hospital of Southern Pest, National Institute of Hematology and Infectious Diseases, Budapest, Hungary (L.K.)
| | - Kinga Oravecz
- Department of Biochemistry, Eötvös Loránd University, Budapest and Martonvásár, Hungary (M.G., K.O., I.L., Z.B., M.K., A.M.-C.); MTA-ELTE Motor Pharmacology Research Group, Budapest, Hungary (M.G., M.K., A.M.-C.); Motorharma Ltd., Budapest, Hungary (A.Á.R.); Printnet Ltd., Budapest, Hungary (S.K.S., I.L.); Plant Protection Institute, Centre for Agricultural Research, Martonvásár, Hungary (K.Á.H.); Metabolic Drug Interactions Research Group, Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary (M.T.D., Á.F.K., K.M.); Research Centre for Natural Sciences, Instrumentation Center, MS Metabolomic Research Laboratory, Budapest, Hungary (P.T.S.); Department of Biology, MyoKardia Inc., Brisbane, California (S.N., I.T.); Department of Neuroscience, Western New England University, Springfield, Massachusetts (J.K., P.J.T.); and Central Hospital of Southern Pest, National Institute of Hematology and Infectious Diseases, Budapest, Hungary (L.K.)
| | - István Lőrincz
- Department of Biochemistry, Eötvös Loránd University, Budapest and Martonvásár, Hungary (M.G., K.O., I.L., Z.B., M.K., A.M.-C.); MTA-ELTE Motor Pharmacology Research Group, Budapest, Hungary (M.G., M.K., A.M.-C.); Motorharma Ltd., Budapest, Hungary (A.Á.R.); Printnet Ltd., Budapest, Hungary (S.K.S., I.L.); Plant Protection Institute, Centre for Agricultural Research, Martonvásár, Hungary (K.Á.H.); Metabolic Drug Interactions Research Group, Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary (M.T.D., Á.F.K., K.M.); Research Centre for Natural Sciences, Instrumentation Center, MS Metabolomic Research Laboratory, Budapest, Hungary (P.T.S.); Department of Biology, MyoKardia Inc., Brisbane, California (S.N., I.T.); Department of Neuroscience, Western New England University, Springfield, Massachusetts (J.K., P.J.T.); and Central Hospital of Southern Pest, National Institute of Hematology and Infectious Diseases, Budapest, Hungary (L.K.)
| | - Zsolt Borhegyi
- Department of Biochemistry, Eötvös Loránd University, Budapest and Martonvásár, Hungary (M.G., K.O., I.L., Z.B., M.K., A.M.-C.); MTA-ELTE Motor Pharmacology Research Group, Budapest, Hungary (M.G., M.K., A.M.-C.); Motorharma Ltd., Budapest, Hungary (A.Á.R.); Printnet Ltd., Budapest, Hungary (S.K.S., I.L.); Plant Protection Institute, Centre for Agricultural Research, Martonvásár, Hungary (K.Á.H.); Metabolic Drug Interactions Research Group, Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary (M.T.D., Á.F.K., K.M.); Research Centre for Natural Sciences, Instrumentation Center, MS Metabolomic Research Laboratory, Budapest, Hungary (P.T.S.); Department of Biology, MyoKardia Inc., Brisbane, California (S.N., I.T.); Department of Neuroscience, Western New England University, Springfield, Massachusetts (J.K., P.J.T.); and Central Hospital of Southern Pest, National Institute of Hematology and Infectious Diseases, Budapest, Hungary (L.K.)
| | - Máté T Déri
- Department of Biochemistry, Eötvös Loránd University, Budapest and Martonvásár, Hungary (M.G., K.O., I.L., Z.B., M.K., A.M.-C.); MTA-ELTE Motor Pharmacology Research Group, Budapest, Hungary (M.G., M.K., A.M.-C.); Motorharma Ltd., Budapest, Hungary (A.Á.R.); Printnet Ltd., Budapest, Hungary (S.K.S., I.L.); Plant Protection Institute, Centre for Agricultural Research, Martonvásár, Hungary (K.Á.H.); Metabolic Drug Interactions Research Group, Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary (M.T.D., Á.F.K., K.M.); Research Centre for Natural Sciences, Instrumentation Center, MS Metabolomic Research Laboratory, Budapest, Hungary (P.T.S.); Department of Biology, MyoKardia Inc., Brisbane, California (S.N., I.T.); Department of Neuroscience, Western New England University, Springfield, Massachusetts (J.K., P.J.T.); and Central Hospital of Southern Pest, National Institute of Hematology and Infectious Diseases, Budapest, Hungary (L.K.)
| | - Ádám F Kiss
- Department of Biochemistry, Eötvös Loránd University, Budapest and Martonvásár, Hungary (M.G., K.O., I.L., Z.B., M.K., A.M.-C.); MTA-ELTE Motor Pharmacology Research Group, Budapest, Hungary (M.G., M.K., A.M.-C.); Motorharma Ltd., Budapest, Hungary (A.Á.R.); Printnet Ltd., Budapest, Hungary (S.K.S., I.L.); Plant Protection Institute, Centre for Agricultural Research, Martonvásár, Hungary (K.Á.H.); Metabolic Drug Interactions Research Group, Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary (M.T.D., Á.F.K., K.M.); Research Centre for Natural Sciences, Instrumentation Center, MS Metabolomic Research Laboratory, Budapest, Hungary (P.T.S.); Department of Biology, MyoKardia Inc., Brisbane, California (S.N., I.T.); Department of Neuroscience, Western New England University, Springfield, Massachusetts (J.K., P.J.T.); and Central Hospital of Southern Pest, National Institute of Hematology and Infectious Diseases, Budapest, Hungary (L.K.)
| | - Katalin Monostory
- Department of Biochemistry, Eötvös Loránd University, Budapest and Martonvásár, Hungary (M.G., K.O., I.L., Z.B., M.K., A.M.-C.); MTA-ELTE Motor Pharmacology Research Group, Budapest, Hungary (M.G., M.K., A.M.-C.); Motorharma Ltd., Budapest, Hungary (A.Á.R.); Printnet Ltd., Budapest, Hungary (S.K.S., I.L.); Plant Protection Institute, Centre for Agricultural Research, Martonvásár, Hungary (K.Á.H.); Metabolic Drug Interactions Research Group, Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary (M.T.D., Á.F.K., K.M.); Research Centre for Natural Sciences, Instrumentation Center, MS Metabolomic Research Laboratory, Budapest, Hungary (P.T.S.); Department of Biology, MyoKardia Inc., Brisbane, California (S.N., I.T.); Department of Neuroscience, Western New England University, Springfield, Massachusetts (J.K., P.J.T.); and Central Hospital of Southern Pest, National Institute of Hematology and Infectious Diseases, Budapest, Hungary (L.K.)
| | - Pál Tamás Szabó
- Department of Biochemistry, Eötvös Loránd University, Budapest and Martonvásár, Hungary (M.G., K.O., I.L., Z.B., M.K., A.M.-C.); MTA-ELTE Motor Pharmacology Research Group, Budapest, Hungary (M.G., M.K., A.M.-C.); Motorharma Ltd., Budapest, Hungary (A.Á.R.); Printnet Ltd., Budapest, Hungary (S.K.S., I.L.); Plant Protection Institute, Centre for Agricultural Research, Martonvásár, Hungary (K.Á.H.); Metabolic Drug Interactions Research Group, Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary (M.T.D., Á.F.K., K.M.); Research Centre for Natural Sciences, Instrumentation Center, MS Metabolomic Research Laboratory, Budapest, Hungary (P.T.S.); Department of Biology, MyoKardia Inc., Brisbane, California (S.N., I.T.); Department of Neuroscience, Western New England University, Springfield, Massachusetts (J.K., P.J.T.); and Central Hospital of Southern Pest, National Institute of Hematology and Infectious Diseases, Budapest, Hungary (L.K.)
| | - Suman Nag
- Department of Biochemistry, Eötvös Loránd University, Budapest and Martonvásár, Hungary (M.G., K.O., I.L., Z.B., M.K., A.M.-C.); MTA-ELTE Motor Pharmacology Research Group, Budapest, Hungary (M.G., M.K., A.M.-C.); Motorharma Ltd., Budapest, Hungary (A.Á.R.); Printnet Ltd., Budapest, Hungary (S.K.S., I.L.); Plant Protection Institute, Centre for Agricultural Research, Martonvásár, Hungary (K.Á.H.); Metabolic Drug Interactions Research Group, Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary (M.T.D., Á.F.K., K.M.); Research Centre for Natural Sciences, Instrumentation Center, MS Metabolomic Research Laboratory, Budapest, Hungary (P.T.S.); Department of Biology, MyoKardia Inc., Brisbane, California (S.N., I.T.); Department of Neuroscience, Western New England University, Springfield, Massachusetts (J.K., P.J.T.); and Central Hospital of Southern Pest, National Institute of Hematology and Infectious Diseases, Budapest, Hungary (L.K.)
| | - Ivan Tomasic
- Department of Biochemistry, Eötvös Loránd University, Budapest and Martonvásár, Hungary (M.G., K.O., I.L., Z.B., M.K., A.M.-C.); MTA-ELTE Motor Pharmacology Research Group, Budapest, Hungary (M.G., M.K., A.M.-C.); Motorharma Ltd., Budapest, Hungary (A.Á.R.); Printnet Ltd., Budapest, Hungary (S.K.S., I.L.); Plant Protection Institute, Centre for Agricultural Research, Martonvásár, Hungary (K.Á.H.); Metabolic Drug Interactions Research Group, Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary (M.T.D., Á.F.K., K.M.); Research Centre for Natural Sciences, Instrumentation Center, MS Metabolomic Research Laboratory, Budapest, Hungary (P.T.S.); Department of Biology, MyoKardia Inc., Brisbane, California (S.N., I.T.); Department of Neuroscience, Western New England University, Springfield, Massachusetts (J.K., P.J.T.); and Central Hospital of Southern Pest, National Institute of Hematology and Infectious Diseases, Budapest, Hungary (L.K.)
| | - Jacob Krans
- Department of Biochemistry, Eötvös Loránd University, Budapest and Martonvásár, Hungary (M.G., K.O., I.L., Z.B., M.K., A.M.-C.); MTA-ELTE Motor Pharmacology Research Group, Budapest, Hungary (M.G., M.K., A.M.-C.); Motorharma Ltd., Budapest, Hungary (A.Á.R.); Printnet Ltd., Budapest, Hungary (S.K.S., I.L.); Plant Protection Institute, Centre for Agricultural Research, Martonvásár, Hungary (K.Á.H.); Metabolic Drug Interactions Research Group, Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary (M.T.D., Á.F.K., K.M.); Research Centre for Natural Sciences, Instrumentation Center, MS Metabolomic Research Laboratory, Budapest, Hungary (P.T.S.); Department of Biology, MyoKardia Inc., Brisbane, California (S.N., I.T.); Department of Neuroscience, Western New England University, Springfield, Massachusetts (J.K., P.J.T.); and Central Hospital of Southern Pest, National Institute of Hematology and Infectious Diseases, Budapest, Hungary (L.K.)
| | - Patrick J Tierney
- Department of Biochemistry, Eötvös Loránd University, Budapest and Martonvásár, Hungary (M.G., K.O., I.L., Z.B., M.K., A.M.-C.); MTA-ELTE Motor Pharmacology Research Group, Budapest, Hungary (M.G., M.K., A.M.-C.); Motorharma Ltd., Budapest, Hungary (A.Á.R.); Printnet Ltd., Budapest, Hungary (S.K.S., I.L.); Plant Protection Institute, Centre for Agricultural Research, Martonvásár, Hungary (K.Á.H.); Metabolic Drug Interactions Research Group, Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary (M.T.D., Á.F.K., K.M.); Research Centre for Natural Sciences, Instrumentation Center, MS Metabolomic Research Laboratory, Budapest, Hungary (P.T.S.); Department of Biology, MyoKardia Inc., Brisbane, California (S.N., I.T.); Department of Neuroscience, Western New England University, Springfield, Massachusetts (J.K., P.J.T.); and Central Hospital of Southern Pest, National Institute of Hematology and Infectious Diseases, Budapest, Hungary (L.K.)
| | - Mihály Kovács
- Department of Biochemistry, Eötvös Loránd University, Budapest and Martonvásár, Hungary (M.G., K.O., I.L., Z.B., M.K., A.M.-C.); MTA-ELTE Motor Pharmacology Research Group, Budapest, Hungary (M.G., M.K., A.M.-C.); Motorharma Ltd., Budapest, Hungary (A.Á.R.); Printnet Ltd., Budapest, Hungary (S.K.S., I.L.); Plant Protection Institute, Centre for Agricultural Research, Martonvásár, Hungary (K.Á.H.); Metabolic Drug Interactions Research Group, Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary (M.T.D., Á.F.K., K.M.); Research Centre for Natural Sciences, Instrumentation Center, MS Metabolomic Research Laboratory, Budapest, Hungary (P.T.S.); Department of Biology, MyoKardia Inc., Brisbane, California (S.N., I.T.); Department of Neuroscience, Western New England University, Springfield, Massachusetts (J.K., P.J.T.); and Central Hospital of Southern Pest, National Institute of Hematology and Infectious Diseases, Budapest, Hungary (L.K.)
| | - László Kornya
- Department of Biochemistry, Eötvös Loránd University, Budapest and Martonvásár, Hungary (M.G., K.O., I.L., Z.B., M.K., A.M.-C.); MTA-ELTE Motor Pharmacology Research Group, Budapest, Hungary (M.G., M.K., A.M.-C.); Motorharma Ltd., Budapest, Hungary (A.Á.R.); Printnet Ltd., Budapest, Hungary (S.K.S., I.L.); Plant Protection Institute, Centre for Agricultural Research, Martonvásár, Hungary (K.Á.H.); Metabolic Drug Interactions Research Group, Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary (M.T.D., Á.F.K., K.M.); Research Centre for Natural Sciences, Instrumentation Center, MS Metabolomic Research Laboratory, Budapest, Hungary (P.T.S.); Department of Biology, MyoKardia Inc., Brisbane, California (S.N., I.T.); Department of Neuroscience, Western New England University, Springfield, Massachusetts (J.K., P.J.T.); and Central Hospital of Southern Pest, National Institute of Hematology and Infectious Diseases, Budapest, Hungary (L.K.)
| | - András Málnási-Csizmadia
- Department of Biochemistry, Eötvös Loránd University, Budapest and Martonvásár, Hungary (M.G., K.O., I.L., Z.B., M.K., A.M.-C.); MTA-ELTE Motor Pharmacology Research Group, Budapest, Hungary (M.G., M.K., A.M.-C.); Motorharma Ltd., Budapest, Hungary (A.Á.R.); Printnet Ltd., Budapest, Hungary (S.K.S., I.L.); Plant Protection Institute, Centre for Agricultural Research, Martonvásár, Hungary (K.Á.H.); Metabolic Drug Interactions Research Group, Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary (M.T.D., Á.F.K., K.M.); Research Centre for Natural Sciences, Instrumentation Center, MS Metabolomic Research Laboratory, Budapest, Hungary (P.T.S.); Department of Biology, MyoKardia Inc., Brisbane, California (S.N., I.T.); Department of Neuroscience, Western New England University, Springfield, Massachusetts (J.K., P.J.T.); and Central Hospital of Southern Pest, National Institute of Hematology and Infectious Diseases, Budapest, Hungary (L.K.)
| |
Collapse
|
14
|
Zhao Q, Mao Y, Li X, Zheng Y. Drug use characteristics and correlates of multiple risk behaviors among synthetic drug users. JOURNAL OF SUBSTANCE USE 2020. [DOI: 10.1080/14659891.2020.1846804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Affiliation(s)
- Qun Zhao
- Research Institute for Environment and Health, School of Law and Public Affairs, Nanjing University of Information Science and Technology, Nanjing, Jiangsu, China
| | - Yuchen Mao
- Department of Statistics, University of South Carolina, Columbia, South Carolina, USA
| | - Xiaoming Li
- Research Institute for Environment and Health, School of Law and Public Affairs, Nanjing University of Information Science and Technology, Nanjing, Jiangsu, China
- South Carolina SmartState Center for Healthcare Quality (CHQ), University of South Carolina Arnold School of Public Health, Columbia, South Carolina, USA
| | - Yu Zheng
- Center for Global Health, Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
15
|
Radnai L, Stremel RF, Vaissiere T, Lin L, Cameron M, Martin WH, Rumbaugh G, Kamenecka TM, Griffin PR, Miller CA. A simple and robust cell-based assay for the discovery of novel cytokinesis inhibitors. J Biol Methods 2020; 7:e136. [PMID: 33204739 PMCID: PMC7666332 DOI: 10.14440/jbm.2020.335] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 05/31/2020] [Accepted: 06/28/2020] [Indexed: 12/12/2022] Open
Abstract
Cytokinesis is the last step of mitotic cell division that separates the cytoplasm of dividing cells. Small molecule inhibitors targeting either the elements of the regulatory pathways controlling cytokinesis, or the terminal effectors have been of interest as potential drug candidates for the treatment of various diseases. Here we present a detailed protocol for a cell-based cytokinesis assay that can be used for the discovery of novel cytokinesis inhibitors. The assay is performed in a 96-well plate format in 48 h. Living cells, nuclei and nuclei of dead cells are identified by a single staining step using three fluorescent dyes, followed by rapid live cell imaging. The primary signal is the nuclei-to-cell ratio (NCR). In the presence of cytokinesis inhibitors, this ratio increases over time, as the ratio of multinucleated cells increases in the population. The ratio of dead nuclei to total nuclei provides a simultaneous measure of cytotoxicity. A screening window coefficient (Z`) of 0.65 indicates that the assay is suitable for screening purposes, as the positive and negative controls are well-separated. EC50 values can be reliably determined in a single 96-well plate by using only six different compound concentrations, enabling the testing of 4 compounds per plate. An excellent test-retest reliability (R2 = 0.998) was found for EC50 values covering a ~1500-fold range of potencies. Established small molecule inhibitors of cytokinesis operating via direct action on actin dynamics or nonmuscle myosin II are used to demonstrate the robustness, simplicity and flexibility of the assay.
Collapse
Affiliation(s)
- Laszlo Radnai
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, FL 33458, USA.,Department of Neuroscience, The Scripps Research Institute, Jupiter, FL 33458, USA
| | - Rebecca F Stremel
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, FL 33458, USA.,Department of Neuroscience, The Scripps Research Institute, Jupiter, FL 33458, USA
| | - Thomas Vaissiere
- Department of Neuroscience, The Scripps Research Institute, Jupiter, FL 33458, USA
| | - Li Lin
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, FL 33458, USA
| | - Michael Cameron
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, FL 33458, USA
| | | | - Gavin Rumbaugh
- Department of Neuroscience, The Scripps Research Institute, Jupiter, FL 33458, USA
| | - Theodore M Kamenecka
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, FL 33458, USA
| | - Patrick R Griffin
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, FL 33458, USA
| | - Courtney A Miller
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, FL 33458, USA.,Department of Neuroscience, The Scripps Research Institute, Jupiter, FL 33458, USA
| |
Collapse
|
16
|
Conventional and Non-Conventional Roles of Non-Muscle Myosin II-Actin in Neuronal Development and Degeneration. Cells 2020; 9:cells9091926. [PMID: 32825197 PMCID: PMC7566000 DOI: 10.3390/cells9091926] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 08/12/2020] [Accepted: 08/13/2020] [Indexed: 12/13/2022] Open
Abstract
Myosins are motor proteins that use chemical energy to produce mechanical forces driving actin cytoskeletal dynamics. In the brain, the conventional non-muscle myosin II (NMII) regulates actin filament cytoskeletal assembly and contractile forces during structural remodeling of axons and dendrites, contributing to morphology, polarization, and migration of neurons during brain development. NMII isoforms also participate in neurotransmission and synaptic plasticity by driving actin cytoskeletal dynamics during synaptic vesicle release and retrieval, and formation, maturation, and remodeling of dendritic spines. NMIIs are expressed differentially in cerebral non-neuronal cells, such as microglia, astrocytes, and endothelial cells, wherein they play key functions in inflammation, myelination, and repair. Besides major efforts to understand the physiological functions and regulatory mechanisms of NMIIs in the nervous system, their contributions to brain pathologies are still largely unclear. Nonetheless, genetic mutations or deregulation of NMII and its regulatory effectors are linked to autism, schizophrenia, intellectual disability, and neurodegeneration, indicating non-conventional roles of NMIIs in cellular mechanisms underlying neurodevelopmental and neurodegenerative disorders. Here, we summarize the emerging biological roles of NMIIs in the brain, and discuss how actomyosin signaling contributes to dysfunction of neurons and glial cells in the context of neurological disorders. This knowledge is relevant for a deep understanding of NMIIs on the pathogenesis and therapeutics of neuropsychiatric and neurodegenerative diseases.
Collapse
|
17
|
Mucke HA. Patent highlights December 2019-January 2020. Pharm Pat Anal 2020; 9:67-74. [PMID: 32539539 DOI: 10.4155/ppa-2020-0005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 04/02/2020] [Indexed: 11/17/2022]
Abstract
A snapshot of noteworthy recent developments in the patent literature of relevance to pharmaceutical and medical research and development.
Collapse
|
18
|
Daws SE, Jamieson S, de Nijs L, Jones M, Snijders C, Klengel T, Joseph NF, Krauskopf J, Kleinjans J, Vinkers CH, Boks MPM, Geuze E, Vermetten E, Berretta S, Ressler KJ, Rutten BPF, Rumbaugh G, Miller CA. MicroRNA regulation of persistent stress-enhanced memory. Mol Psychiatry 2020; 25:965-976. [PMID: 31142820 PMCID: PMC6883139 DOI: 10.1038/s41380-019-0432-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 03/04/2019] [Accepted: 04/11/2019] [Indexed: 12/28/2022]
Abstract
Disruption of persistent, stress-associated memories is relevant for treating posttraumatic stress disorder (PTSD) and related syndromes, which develop in a subset of individuals following a traumatic event. We previously developed a stress-enhanced fear learning (SEFL) paradigm in inbred mice that produces PTSD-like characteristics in a subset of mice, including persistently enhanced memory and heightened cFos in the basolateral amygdala complex (BLC) with retrieval of the remote (30-day-old) stress memory. Here, the contribution of BLC microRNAs (miRNAs) to stress-enhanced memory was investigated because of the molecular complexity they achieve through their ability to regulate multiple targets simultaneously. We performed small-RNA sequencing (smRNA-Seq) and quantitative proteomics on BLC tissue collected from mice 1 month after SEFL and identified persistently changed microRNAs, including mir-135b-5p, and proteins associated with PTSD-like heightened fear expression. Viral-mediated overexpression of mir-135b-5p in the BLC of stress-resilient animals enhanced remote fear memory expression and promoted spontaneous renewal 14 days after extinction. Conversely, inhibition of BLC mir-135b-5p in stress-susceptible animals had the opposite effect, promoting a resilient-like phenotype. mir-135b-5p is highly conserved across mammals and was detected in post mortem human amygdala, as well as human serum samples. The mir-135b passenger strand, mir-135b-3p, was significantly elevated in serum from PTSD military veterans, relative to combat-exposed control subjects. Thus, miR-135b-5p may be an important therapeutic target for dampening persistent, stress-enhanced memory and its passenger strand a potential biomarker for responsivity to a mir-135-based therapeutic.
Collapse
Affiliation(s)
- Stephanie E Daws
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, FL, USA
- Department of Neuroscience, The Scripps Research Institute, Jupiter, FL, USA
| | - Sarah Jamieson
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, FL, USA
- Department of Neuroscience, The Scripps Research Institute, Jupiter, FL, USA
| | - Laurence de Nijs
- School for Mental Health and Neuroscience, Department of Psychiatry and Neuropsychology, Maastricht University, Maastricht, The Netherlands
| | - Meghan Jones
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, FL, USA
- Department of Neuroscience, The Scripps Research Institute, Jupiter, FL, USA
| | - Clara Snijders
- School for Mental Health and Neuroscience, Department of Psychiatry and Neuropsychology, Maastricht University, Maastricht, The Netherlands
| | - Torsten Klengel
- Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA, USA
| | - Nadine F Joseph
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, FL, USA
- Department of Neuroscience, The Scripps Research Institute, Jupiter, FL, USA
| | - Julian Krauskopf
- Department of Toxicogenomics, Maastricht University, Maastricht, The Netherlands
| | - Jos Kleinjans
- Department of Toxicogenomics, Maastricht University, Maastricht, The Netherlands
| | - Christiaan H Vinkers
- Department of Psychiatry, Amsterdam UMC (location VUmc), Amsterdam, The Netherlands
- Department of Anatomy and Neurosciences, Amsterdam UMC (location VUmc), Amsterdam, The Netherlands
| | - Marco P M Boks
- Brain Center Rudolf Magnus, Department of Psychiatry, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Elbert Geuze
- Brain Center Rudolf Magnus, Department of Psychiatry, University Medical Center Utrecht, Utrecht, The Netherlands
- Research Centre for Military Mental Healthcare, Ministry of Defence, Utrecht, The Netherlands
| | - Eric Vermetten
- Brain Center Rudolf Magnus, Department of Psychiatry, University Medical Center Utrecht, Utrecht, The Netherlands
- Research Centre for Military Mental Healthcare, Ministry of Defence, Utrecht, The Netherlands
- Department of Psychiatry, Leiden University Medical Center, Leiden, The Netherlands
| | - Sabina Berretta
- Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA, USA
| | - Kerry J Ressler
- Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA, USA
| | - Bart P F Rutten
- School for Mental Health and Neuroscience, Department of Psychiatry and Neuropsychology, Maastricht University, Maastricht, The Netherlands
| | - Gavin Rumbaugh
- Department of Neuroscience, The Scripps Research Institute, Jupiter, FL, USA
| | - Courtney A Miller
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, FL, USA.
- Department of Neuroscience, The Scripps Research Institute, Jupiter, FL, USA.
| |
Collapse
|
19
|
Methamphetamine Learning Induces Persistent and Selective Nonmuscle Myosin II-Dependent Spine Motility in the Basolateral Amygdala. J Neurosci 2020; 40:2695-2707. [PMID: 32066582 DOI: 10.1523/jneurosci.2182-19.2020] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Revised: 01/17/2020] [Accepted: 02/04/2020] [Indexed: 12/19/2022] Open
Abstract
Nonmuscle myosin II inhibition (NMIIi) in the basolateral amygdala (BLA), but not dorsal hippocampus (CA1), selectively disrupts memories associated with methamphetamine (METH) days after learning, without retrieval. However, the molecular mechanisms underlying this selective vulnerability remain poorly understood. A known function of NMII is to transiently activate synaptic actin dynamics with learning. Therefore, we hypothesized that METH-associated learning perpetuates NMII-driven actin dynamics in synapses, leading to an extended window of vulnerability for memory disruption. We used time-lapse two-photon imaging of dendritic spine motility in acutely prepared brain slices from female and male mice following METH-associated learning as a readout of actin-myosin dynamics. Spine motility was persistently increased in the BLA, but not in CA1. Consistent with the memory disrupting effect of intra-BLA NMII inhibition, METH-induced changes to BLA spine dynamics were reversed by a single systemic injection of an NMII inhibitor. Intra-CA1 NMII inhibition, on the other hand, did not disrupt METH-associated memory. Thus, we report identification of a previously unknown ability for spine actin dynamics to persist days after stimulation and that this is under the control of NMII. Further, these perpetual NMII-driven spine actin dynamics in BLA neurons may contribute to the unique susceptibility of METH-associated memories.SIGNIFICANCE STATEMENT There are no Food and Drug Administration-approved pharmacotherapies to prevent relapse to the use of stimulants, such as methamphetamine (METH). Environmental cues become associated with drug use, such that the memories can elicit strong motivation to seek the drug during abstinence. We previously reported that the storage of METH-associated memories is uniquely vulnerable to immediate, retrieval-independent, and lasting disruption by direct actin depolymerization or by inhibiting the actin driver nonmuscle myosin II (NMII) in the BLA or systemically. Here we report a potential structural mechanism responsible for the unique vulnerability of METH-associated memories and METH-seeking behavior to NMII inhibition within the BLA.
Collapse
|
20
|
Jones ME, Sillivan SE, Jamieson S, Rumbaugh G, Miller CA. microRNA mir-598-3p mediates susceptibility to stress enhancement of remote fear memory. ACTA ACUST UNITED AC 2019; 26:363-372. [PMID: 31416909 PMCID: PMC6699414 DOI: 10.1101/lm.048827.118] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Accepted: 06/10/2019] [Indexed: 12/26/2022]
Abstract
microRNAs (miRNAs) have emerged as potent regulators of learning, recent memory, and extinction. However, our understanding of miRNAs directly involved in regulating complex psychiatric conditions perpetuated by aberrant memory, such as in posttraumatic stress disorder (PTSD), remains limited. To begin to address the role of miRNAs in persistent memories, we performed small-RNA sequencing on basolateral amygdala (BLA) tissue and identified miRNAs altered by auditory fear conditioning (FC) one month after training. mir-598-3p, a highly conserved miRNA previously unstudied in the brain, was down-regulated in the BLA. Further decreasing BLA mir-598-3p levels did not increase strength of the remote fear memory. Given that stress is a critical component in PTSD, we next assessed the impact of stress and stress-enhanced fear learning (SEFL) on mir-598-3p levels, finding the miRNA is elevated in the BLA of male, but not female, mice susceptible to the effects of stress in SEFL. Accordingly, intra-BLA inhibition of mir-598-3p interfered with expression and extinction of the remote fear memory in male, but not female, mice. This effect could not be attributed to an anxiolytic effect of miRNA inhibition. Finally, bioinformatic analysis following quantitative proteomics on BLA tissue collected 30 d post-SEFL training identified putative mir-598-3p targets and related pathways mediating the differential susceptibility, with evidence for regulation of the actin cytoskeleton, the core mediator of structural plasticity. Taken together, the results suggest BLA mir-598-3p may be recruited by stress to mediate a critical switch from a salient remote fear memory to one that is enhanced and extinction-resistant.
Collapse
Affiliation(s)
- Meghan E Jones
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, Florida 33458, USA.,Department of Neuroscience, The Scripps Research Institute, Jupiter, Florida 33458, USA
| | - Stephanie E Sillivan
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, Florida 33458, USA.,Department of Neuroscience, The Scripps Research Institute, Jupiter, Florida 33458, USA
| | - Sarah Jamieson
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, Florida 33458, USA.,Department of Neuroscience, The Scripps Research Institute, Jupiter, Florida 33458, USA
| | - Gavin Rumbaugh
- Department of Neuroscience, The Scripps Research Institute, Jupiter, Florida 33458, USA
| | - Courtney A Miller
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, Florida 33458, USA.,Department of Neuroscience, The Scripps Research Institute, Jupiter, Florida 33458, USA
| |
Collapse
|
21
|
Wróbel A, Nowakowski Ł, Doboszewska U, Rechberger E, Bańczerowska-Górska M, Wlaźlak E, Zakrocka I, Wlaź P, Semczuk A, Dudka J, Poleszak E. Blebbistatin reveals beneficial effects on the cystometric parameters in an animal model of detrusor overactivity. Naunyn Schmiedebergs Arch Pharmacol 2019; 392:843-850. [PMID: 30852655 PMCID: PMC7260150 DOI: 10.1007/s00210-019-01640-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2018] [Accepted: 02/25/2019] [Indexed: 11/29/2022]
Abstract
The aims of the study were to determine the effectiveness of blebbistatin (BLEB) on detrusor overactivity (DO) in an animal model induced by retinyl acetate (RA) and, because of potential urothelial permeability, to evaluate the degenerative impact of BLEB on the urothelium. Three days after RA instillation into the urinary bladder, BLEB was administered into the bladder and immediately after cystometric assessment was performed. Furthermore, Evans Blue extravasation into bladder tissue and urothelium thickness were measured. Sixty female Wistar rats were used and randomly assigned to one of four groups (n = 15 in each group): (1) control, (2) RA, (3) BLEB, and (4) RA + BLEB. RA administration induced changes in cystometric parameters reflecting DO, as previously reported. Treatment with BLEB did not significantly alter cystometric parameters in rats which did not receive RA. Administration of BLEB to rats pretreated with RA reversed changes in cystometric parameters induced by RA in basal pressure, threshold pressure, detrusor overactivity index, amplitude of nonvoiding contractions, frequency of nonvoiding contractions, voided volume, volume threshold, intercontraction interval, bladder compliance, and volume threshold to elicit nonvoiding contractions. There were no significant differences in Evans Blue extravasation into bladder tissue or urothelium thickness between the groups. The current research provides new data on the possible utility of blebbistatin in the pharmacotherapy of DO, which is an important feature of overactive bladder (OAB). Further studies in human patients with DO/OAB are warranted to confirm these preclinical results.
Collapse
Affiliation(s)
- Andrzej Wróbel
- Second Department of Gynecology, Medical University of Lublin, Jaczewskiego 8, 20-954, Lublin, Poland.
| | - Łukasz Nowakowski
- Second Department of Gynecology, Medical University of Lublin, Jaczewskiego 8, 20-954, Lublin, Poland
| | - Urszula Doboszewska
- Department of Animal Physiology, Institute of Biology and Biochemistry, Faculty of Biology and Biotechnology, Maria Curie-Sklodowska University, Akademicka 19, 20-033, Lublin, Poland.
| | - Ewa Rechberger
- Second Department of Gynecology, Medical University of Lublin, Jaczewskiego 8, 20-954, Lublin, Poland
| | | | - Edyta Wlaźlak
- Clinic of Operative Gynecology and Gynecologic Oncology, 1st Department of Gynecology and Obstetrics, Medical University of Lodz, Wileńska 37, Łódź, 94-029, Poland
| | - Izabela Zakrocka
- Chair and Department of Nephrology, Medical University of Lublin, Jaczewskiego 8, 20-954, Lublin, Poland
| | - Piotr Wlaź
- Department of Animal Physiology, Institute of Biology and Biochemistry, Faculty of Biology and Biotechnology, Maria Curie-Sklodowska University, Akademicka 19, 20-033, Lublin, Poland
| | - Andrzej Semczuk
- Second Department of Gynecology, Medical University of Lublin, Jaczewskiego 8, 20-954, Lublin, Poland
| | - Jarosław Dudka
- Chair and Department of Toxicology, Medical University of Lublin, Chodźki 8, 20-093, Lublin, Poland
| | - Ewa Poleszak
- Chair and Department of Applied Pharmacy, Medical University of Lublin, Chodźki 1, 20-093, Lublin, Poland
| |
Collapse
|
22
|
Belcher AM, Ferré S, Martinez PE, Colloca L. Role of placebo effects in pain and neuropsychiatric disorders. Prog Neuropsychopharmacol Biol Psychiatry 2018; 87:298-306. [PMID: 28595945 PMCID: PMC5722709 DOI: 10.1016/j.pnpbp.2017.06.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Revised: 06/01/2017] [Accepted: 06/03/2017] [Indexed: 02/06/2023]
Abstract
The placebo (and the nocebo) effect is a powerful determinant of health outcomes in clinical disease treatment and management. Efforts to completely eradicate placebo effects have shifted dynamically, as increasingly more researchers are tuned to the potentially beneficial effects of incorporating those uncontrollable placebo effects into clinical therapeutic strategies. In this review, we highlight the major findings from placebo research, elucidating the main neurobiological systems and candidate determinants of the placebo phenomenon, and illustrate a perspective that can effectively frame future research on the topic. Finally, we issue a call for increased research on the efficacy of therapeutic strategies that incorporate placebo "tools," and argue that clinical trials of the placebo response in neuropsychiatric diseases and disorders has important and far-reaching translational and clinical relevance.
Collapse
Affiliation(s)
- Annabelle M Belcher
- Department of Psychiatry, School of Medicine, University of Maryland, Baltimore, USA
| | - Sergi Ferré
- National Institute on Drug Abuse, National Institutes of Health, Bethesda, USA
| | - Pedro E Martinez
- National Institute of Mental Health, National Institutes of Health, Bethesda, USA
| | - Luana Colloca
- Department of Psychiatry, School of Medicine, University of Maryland, Baltimore, USA; Department of Pain and Translational Symptom Science, School of Nursing, University of Maryland, Baltimore, USA; Department of Anesthesiology and Psychiatry, School of Medicine, University of Maryland, Baltimore, USA; Center to Advance Chronic Pain Research, University of Maryland, Baltimore, USA.
| |
Collapse
|
23
|
Briggs SB, Hafenbreidel M, Young EJ, Rumbaugh G, Miller CA. The role of nonmuscle myosin II in polydrug memories and memory reconsolidation. ACTA ACUST UNITED AC 2018; 25:391-398. [PMID: 30115760 PMCID: PMC6097765 DOI: 10.1101/lm.046763.117] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Accepted: 06/21/2018] [Indexed: 12/13/2022]
Abstract
Using pharmacologic and genetic approaches targeting actin or the actin-driving molecular motor, nonmuscle myosin II (NMII), we previously discovered an immediate, retrieval-independent, and long-lasting disruption of methamphetamine- (METH-) and amphetamine-associated memories. A single intrabasolateral amygdala complex infusion or systemic administration of the NMII inhibitor Blebbistatin (Blebb) is sufficient to produce this disruption, which is selective, having no retrieval-independent effect on memories for fear, food reward, cocaine, or morphine. However, it was unclear if Blebb treatment would disrupt memories of other stimulants and amphetamine class drugs, such as nicotine (NIC) or mephedrone (MEPH; bath salts). Moreover, many individuals abuse multiple drugs, but it was unknown if Blebb could disrupt polydrug memories, or if the inclusion of another substance would render Blebb no longer able to disrupt METH-associated memories. Therefore, the present study had two primary goals: (1) to determine the ability of Blebb to disrupt NIC- or MEPH-associated memories, and (2) to determine the ability of METH to modify other unconditioned stimulus (US) associations’ susceptibility to Blebb. To this end, using the conditional place preference model, mice were conditioned to NIC and MEPH alone or METH in combination with NIC, morphine, or foot shock. We report that, unlike METH, there was no retrieval-independent effect of Blebb on NIC- or MEPH-associated memories. However, similar to cocaine, reconsolidation of the memory for both drugs was disrupted. Further, when combined with METH administration, NIC- and morphine-, but not fear-, associated memories were rendered susceptible to disruption by Blebb. Given the high rate of polydrug use and the resurgence of METH use, these results have important implications for the treatment of substance use disorder.
Collapse
Affiliation(s)
- Sherri B Briggs
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, Florida 33458, USA.,Department of Neuroscience, The Scripps Research Institute, Jupiter, Florida 33458, USA
| | - Madalyn Hafenbreidel
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, Florida 33458, USA.,Department of Neuroscience, The Scripps Research Institute, Jupiter, Florida 33458, USA
| | - Erica J Young
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, Florida 33458, USA.,Department of Neuroscience, The Scripps Research Institute, Jupiter, Florida 33458, USA
| | - Gavin Rumbaugh
- Department of Neuroscience, The Scripps Research Institute, Jupiter, Florida 33458, USA
| | - Courtney A Miller
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, Florida 33458, USA.,Department of Neuroscience, The Scripps Research Institute, Jupiter, Florida 33458, USA
| |
Collapse
|
24
|
Doncheck EM, Hafenbreidel M, Ruder SA, Fitzgerald MK, Torres L, Mueller D. bFGF expression is differentially regulated by cocaine seeking versus extinction in learning-related brain regions. Learn Mem 2018; 25:361-368. [PMID: 30012881 PMCID: PMC6049391 DOI: 10.1101/lm.047530.118] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Accepted: 06/11/2018] [Indexed: 11/24/2022]
Abstract
In cocaine use disorder, relapse can be elicited by drug-associated cues despite long periods of abstinence. The persistence of drug-associated cues in eliciting drug seeking suggests enduring changes in structural and functional plasticity, which may be mediated by basic fibroblast growth factor (bFGF, FGF2). Stimulant drug use increases bFGF expression in reward- and learning-related brain regions, such as the infralimbic medial-prefrontal cortex (IL-mPFC), and we previously found that this increase was reversed by extinction. However, whether bFGF expression is similarly modified in other brain regions is unknown. Therefore, we used the conditioned place preference (CPP) paradigm to assess bFGF expression following cocaine-associated CPP or extinction of that CPP within the mPFC, nucleus accumbens (NAc), hippocampus (Hipp), and basolateral amygdala (BLA). bFGF expression was increased in IL-mPFC and NAc-Core and -Shell following a cocaine-associated CPP, an effect reversed by extinction. Conversely, bFGF expression was increased in BLA following extinction, but no significant changes were observed in PL-mPFC or either dorsal or ventral Hipp. These results demonstrate differential regulation of bFGF following cocaine-associated CPP or extinction of that CPP in discrete brain regions. Changes in bFGF expression may regulate long-lasting drug-induced plasticity that underlies persistent drug-associated memories, and therefore present potential prophylactic targets.
Collapse
Affiliation(s)
- Elizabeth M Doncheck
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, Wisconsin 53201-0413, USA
| | - Madalyn Hafenbreidel
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, Wisconsin 53201-0413, USA
| | - Sarah A Ruder
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, Wisconsin 53201-0413, USA
| | - Michael K Fitzgerald
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, Wisconsin 53201-0413, USA
| | - Lilith Torres
- Department of Basic Sciences, Ponce Health Sciences University-School of Medicine/Ponce Research Institute, Ponce, Puerto Rico, 00732, USA
| | - Devin Mueller
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, Wisconsin 53201-0413, USA
- Department of Basic Sciences, Ponce Health Sciences University-School of Medicine/Ponce Research Institute, Ponce, Puerto Rico, 00732, USA
| |
Collapse
|
25
|
Rauscher AÁ, Gyimesi M, Kovács M, Málnási-Csizmadia A. Targeting Myosin by Blebbistatin Derivatives: Optimization and Pharmacological Potential. Trends Biochem Sci 2018; 43:700-713. [PMID: 30057142 DOI: 10.1016/j.tibs.2018.06.006] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Revised: 06/08/2018] [Accepted: 06/11/2018] [Indexed: 11/28/2022]
Abstract
Blebbistatin is a widely used inhibitor of myosin 2 that enables the study of a broad range of cytoskeleton-related processes. However, blebbistatin has several limitations hindering its applicability: it is fluorescent, poorly water soluble, cytotoxic, and prone to (photo)degradation. Despite these adverse effects, being the only available myosin 2-specific inhibitor, blebbistatin is rather a choice of necessity. Blebbistatin has been modified to improve its properties and some of the new compounds have proven to be useful replacements of the original molecule. This review summarizes recent results on blebbistatin development. We also discuss the pharmacological perspectives of these efforts, as myosins are becoming promising drug target candidates for a variety of conditions ranging from neurodegeneration to muscle disease, wound healing, and cancer metastasis.
Collapse
Affiliation(s)
- Anna Á Rauscher
- Motor Pharmacology Research Group, Department of Biochemistry, Eötvös Loránd University, H-1117 Budapest, Hungary
| | - Máté Gyimesi
- Motor Pharmacology Research Group, Department of Biochemistry, Eötvös Loránd University, H-1117 Budapest, Hungary
| | - Mihály Kovács
- Motor Pharmacology Research Group, Department of Biochemistry, Eötvös Loránd University, H-1117 Budapest, Hungary.
| | - András Málnási-Csizmadia
- Motor Pharmacology Research Group, Department of Biochemistry, Eötvös Loránd University, H-1117 Budapest, Hungary.
| |
Collapse
|
26
|
Roman BI, Verhasselt S, Mangodt CW, De Wever O, Stevens CV. Synthesis of C-ring-modified blebbistatin derivatives and evaluation of their myosin II ATPase inhibitory potency. Bioorg Med Chem Lett 2018; 28:2261-2264. [DOI: 10.1016/j.bmcl.2018.05.041] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Revised: 05/15/2018] [Accepted: 05/21/2018] [Indexed: 10/16/2022]
|
27
|
Do Actomyosin Single-Molecule Mechanics Data Predict Mechanics of Contracting Muscle? Int J Mol Sci 2018; 19:ijms19071863. [PMID: 29941816 PMCID: PMC6073448 DOI: 10.3390/ijms19071863] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Revised: 06/19/2018] [Accepted: 06/20/2018] [Indexed: 12/15/2022] Open
Abstract
In muscle, but not in single-molecule mechanics studies, actin, myosin and accessory proteins are incorporated into a highly ordered myofilament lattice. In view of this difference we compare results from single-molecule studies and muscle mechanics and analyze to what degree data from the two types of studies agree with each other. There is reasonable correspondence in estimates of the cross-bridge power-stroke distance (7–13 nm), cross-bridge stiffness (~2 pN/nm) and average isometric force per cross-bridge (6–9 pN). Furthermore, models defined on the basis of single-molecule mechanics and solution biochemistry give good fits to experimental data from muscle. This suggests that the ordered myofilament lattice, accessory proteins and emergent effects of the sarcomere organization have only minor modulatory roles. However, such factors may be of greater importance under e.g., disease conditions. We also identify areas where single-molecule and muscle data are conflicting: (1) whether force generation is an Eyring or Kramers process with just one major power-stroke or several sub-strokes; (2) whether the myofilaments and the cross-bridges have Hookean or non-linear elasticity; (3) if individual myosin heads slip between actin sites under certain conditions, e.g., in lengthening; or (4) if the two heads of myosin cooperate.
Collapse
|
28
|
Abstract
( S)-Blebbistatin, a chiral tetrahydropyrroloquinolinone, is a widely used and well-characterized ATPase inhibitor selective for myosin II. The central role of myosin II in many normal and pathological biological processes has been revealed with the aid of this small molecule. The first part of this manuscript provides a summary of myosin II and ( S)-blebbistatin literature from a medicinal chemist's perspective. The second part of this perspective deals with the physicochemical deficiencies that trouble the use of ( S)-blebbistatin in advanced biological settings: low potency and solubility, fluorescence interference, (photo)toxicity, and stability issues. A large toolbox of analogues has been developed in which particular shortcomings have been addressed. This perspective provides a necessary overview of these developments and presents guidelines for selecting the best available analogue for a given application. As the unmet need for high-potency analogues remains, we also propose starting points for medicinal chemists in search of nanomolar myosin II inhibitors.
Collapse
|
29
|
Roman BI, Guedes RC, Stevens CV, García-Sosa AT. Recovering Actives in Multi-Antitarget and Target Design of Analogs of the Myosin II Inhibitor Blebbistatin. Front Chem 2018; 6:179. [PMID: 29881723 PMCID: PMC5976736 DOI: 10.3389/fchem.2018.00179] [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: 02/28/2018] [Accepted: 05/04/2018] [Indexed: 11/18/2022] Open
Abstract
In multitarget drug design, it is critical to identify active and inactive compounds against a variety of targets and antitargets. Multitarget strategies thus test the limits of available technology, be that in screening large databases of compounds vs. a large number of targets, or in using in silico methods for understanding and reliably predicting these pharmacological outcomes. In this paper, we have evaluated the potential of several in silico approaches to predict the target, antitarget and physicochemical profile of (S)-blebbistatin, the best-known myosin II ATPase inhibitor, and a series of analogs thereof. Standard and augmented structure-based design techniques could not recover the observed activity profiles. A ligand-based method using molecular fingerprints was, however, able to select actives for myosin II inhibition. Using further ligand- and structure-based methods, we also evaluated toxicity through androgen receptor binding, affinity for an array of antitargets and the ADME profile (including assay-interfering compounds) of the series. In conclusion, in the search for (S)-blebbistatin analogs, the dissimilarity distance of molecular fingerprints to known actives and the computed antitarget and physicochemical profile of the molecules can be used for compound design for molecules with potential as tools for modulating myosin II and motility-related diseases.
Collapse
Affiliation(s)
- Bart I Roman
- Research Group SynBioC, Department of Green Chemistry and Technology, Faculty of Bioscience Engineering, Ghent University Ghent, Belgium.,Cancer Research Institute Ghent Ghent, Belgium
| | - Rita C Guedes
- Department of Medicinal Chemistry, Faculty of Pharmacy, Research Institute for Medicines (iMed.ULisboa), Universidade de Lisboa Lisbon, Portugal
| | - Christian V Stevens
- Research Group SynBioC, Department of Green Chemistry and Technology, Faculty of Bioscience Engineering, Ghent University Ghent, Belgium.,Cancer Research Institute Ghent Ghent, Belgium
| | - Alfonso T García-Sosa
- Department of Molecular Technology, Institute of Chemistry, University of Tartu Tartu, Estonia
| |
Collapse
|
30
|
Verhasselt S, Roman BI, De Wever O, Van Hecke K, Van Deun R, Bracke ME, Stevens CV. Discovery of (S)-3'-hydroxyblebbistatin and (S)-3'-aminoblebbistatin: polar myosin II inhibitors with superior research tool properties. Org Biomol Chem 2018; 15:2104-2118. [PMID: 28220174 DOI: 10.1039/c7ob00006e] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
In search of myosin II inhibitors with superior research tool properties, a chemical optimization campaign of the blebbistatin scaffold was conducted in this paper. (S)-Blebbistatin is the best known small-molecule inhibitor of myosin II ATPase activity. Unfortunately, as a research tool this compound has several deficiencies: it is photolabile and (photo)toxic, has low water solubility, and its (fluorescent) precipitates interfere in (fluorescence) readouts. In view of obtaining tool compounds with improved properties, both enantiomers of a series of D-ring modified polar analogs were prepared. We identified (S)-3'-hydroxyblebbistatin (S)-2 and (S)-3'-aminoblebbistatin (S)-3 as two myosin II inhibitors with a 30-fold higher water solubility than (S)-blebbistatin. These molecules furthermore do not cause interference in (fluorescence) readouts. (S)-2 and (S)-3 thus are superior alternatives to (S)-blebbistatin as research tools to study myosin II.
Collapse
Affiliation(s)
- Sigrid Verhasselt
- SynBioC Research Group, Department of Sustainable Organic Chemistry and Technology, Ghent University, Coupure Links 653, 9000 Ghent, Belgium.
| | - Bart I Roman
- SynBioC Research Group, Department of Sustainable Organic Chemistry and Technology, Ghent University, Coupure Links 653, 9000 Ghent, Belgium.
| | - Olivier De Wever
- Laboratory of Experimental Cancer Research, Department of Radiation Oncology and Experimental Cancer Research, Ghent University, De Pintelaan 185, 9000 Ghent, Belgium
| | - Kristof Van Hecke
- Department of Inorganic and Physical Chemistry, Ghent University, Krijgslaan 281 S3, 9000 Ghent, Belgium
| | - Rik Van Deun
- Department of Inorganic and Physical Chemistry, Ghent University, Krijgslaan 281 S3, 9000 Ghent, Belgium
| | - Marc E Bracke
- Laboratory of Experimental Cancer Research, Department of Radiation Oncology and Experimental Cancer Research, Ghent University, De Pintelaan 185, 9000 Ghent, Belgium
| | - Christian V Stevens
- SynBioC Research Group, Department of Sustainable Organic Chemistry and Technology, Ghent University, Coupure Links 653, 9000 Ghent, Belgium.
| |
Collapse
|
31
|
Swanson AM, DePoy LM, Gourley SL. Inhibiting Rho kinase promotes goal-directed decision making and blocks habitual responding for cocaine. Nat Commun 2017; 8:1861. [PMID: 29187752 PMCID: PMC5707361 DOI: 10.1038/s41467-017-01915-4] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2017] [Accepted: 10/25/2017] [Indexed: 01/04/2023] Open
Abstract
The prelimbic prefrontal cortex is necessary for associating actions with their consequences, enabling goal-directed decision making. We find that the strength of action–outcome conditioning correlates with dendritic spine density in prelimbic cortex, suggesting that new action–outcome learning involves dendritic spine plasticity. To test this, we inhibited the cytoskeletal regulatory factor Rho kinase. We find that the inhibitor fasudil enhances action–outcome memory, resulting in goal-directed behavior in mice that would otherwise express stimulus-response habits. Fasudil transiently reduces prelimbic cortical dendritic spine densities during a period of presumed memory consolidation, but only when paired with new learning. Fasudil also blocks habitual responding for cocaine, an effect that persists over time, across multiple contexts, and depends on actin polymerization. We suggest that Rho kinase inhibition promotes goal-oriented action selection by augmenting the plasticity of prelimbic cortical dendritic spines during the formation of new action–outcome memories. Action-outcome learning requires the prelimbic prefrontal cortex. Here the authors report that fasudil, a Rho kinase inhibitor, reduces dendritic spine densities on prelimbic neurons in an activity-dependent manner, stimulating goal-directed actions, and reducing habitual responding for cocaine.
Collapse
Affiliation(s)
- Andrew M Swanson
- Departments of Pediatrics and Psychiatry, Emory University School of Medicine, 954 Gatewood Road NE, Atlanta, GA, 30329, USA.,Yerkes National Primate Research Center, Graduate Program in Neuroscience, Emory University, 954 Gatewood Road NE, Atlanta, GA, 30329, USA
| | - Lauren M DePoy
- Departments of Pediatrics and Psychiatry, Emory University School of Medicine, 954 Gatewood Road NE, Atlanta, GA, 30329, USA.,Yerkes National Primate Research Center, Graduate Program in Neuroscience, Emory University, 954 Gatewood Road NE, Atlanta, GA, 30329, USA
| | - Shannon L Gourley
- Departments of Pediatrics and Psychiatry, Emory University School of Medicine, 954 Gatewood Road NE, Atlanta, GA, 30329, USA. .,Yerkes National Primate Research Center, Graduate Program in Neuroscience, Emory University, 954 Gatewood Road NE, Atlanta, GA, 30329, USA.
| |
Collapse
|
32
|
M344 promotes nonamyloidogenic amyloid precursor protein processing while normalizing Alzheimer's disease genes and improving memory. Proc Natl Acad Sci U S A 2017; 114:E9135-E9144. [PMID: 29073110 PMCID: PMC5664514 DOI: 10.1073/pnas.1707544114] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Hundreds of failed clinical trials with Alzheimer’s disease (AD) patients over the last fifteen years demonstrate that the one-target–one-disease approach is not effective in AD. In silico, structure-based, multitarget drug design approaches to treat multifactorial diseases have not been successful in the context of AD either. Here, we show that M344, an inhibitor of class I and IIB histone deacetylases, affects multiple AD-related genes, including those related to both early- and late-onset AD. We also show that M344 improves memory in the 3xTg AD mouse model. This work endorses a shift to a multitargeted approach to the treatment of AD, supporting the therapeutic potential of a single small molecule with an epigenetic mechanism of action. Alzheimer’s disease (AD) comprises multifactorial ailments for which current therapeutic strategies remain insufficient to broadly address the underlying pathophysiology. Epigenetic gene regulation relies upon multifactorial processes that regulate multiple gene and protein pathways, including those involved in AD. We therefore took an epigenetic approach where a single drug would simultaneously affect the expression of a number of defined AD-related targets. We show that the small-molecule histone deacetylase inhibitor M344 reduces beta-amyloid (Aβ), reduces tau Ser396 phosphorylation, and decreases both β-secretase (BACE) and APOEε4 gene expression. M344 increases the expression of AD-relevant genes: BDNF, α-secretase (ADAM10), MINT2, FE65, REST, SIRT1, BIN1, and ABCA7, among others. M344 increases sAPPα and CTFα APP metabolite production, both cleavage products of ADAM10, concordant with increased ADAM10 gene expression. M344 also increases levels of immature APP, supporting an effect on APP trafficking, concurrent with the observed increase in MINT2 and FE65, both shown to increase immature APP in the early secretory pathway. Chronic i.p. treatment of the triple transgenic (APPsw/PS1M146V/TauP301L) mice with M344, at doses as low as 3 mg/kg, significantly prevented cognitive decline evaluated by Y-maze spontaneous alternation, novel object recognition, and Barnes maze spatial memory tests. M344 displays short brain exposure, indicating that brief pulses of daily drug treatment may be sufficient for long-term efficacy. Together, these data show that M344 normalizes several disparate pathogenic pathways related to AD. M344 therefore serves as an example of how a multitargeting compound could be used to address the polygenic nature of multifactorial diseases.
Collapse
|
33
|
Otis JM, Mueller D. Reversal of Cocaine-Associated Synaptic Plasticity in Medial Prefrontal Cortex Parallels Elimination of Memory Retrieval. Neuropsychopharmacology 2017; 42:2000-2010. [PMID: 28466871 PMCID: PMC5561348 DOI: 10.1038/npp.2017.90] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Revised: 04/26/2017] [Accepted: 04/27/2017] [Indexed: 01/24/2023]
Abstract
Addiction is characterized by abnormalities in prefrontal cortex that are thought to allow drug-associated cues to drive compulsive drug seeking and taking. Identification and reversal of these pathologic neuroadaptations are therefore critical for treatment of addiction. Previous studies using rodents reveal that drugs of abuse cause dendritic spine plasticity in prelimbic medial prefrontal cortex (PL-mPFC) pyramidal neurons, a phenomenon that correlates with the strength of drug-associated memories in vivo. Thus, we hypothesized that cocaine-evoked plasticity in PL-mPFC may underlie cocaine-associated memory retrieval, and therefore disruption of this plasticity would prevent retrieval. Indeed, using patch clamp electrophysiology we find that cocaine place conditioning increases excitatory presynaptic and postsynaptic transmission in rat PL-mPFC pyramidal neurons. This was accounted for by increases in excitatory presynaptic release, paired-pulse facilitation, and increased AMPA receptor transmission. Noradrenergic signaling is known to maintain glutamatergic plasticity upon reactivation of modified circuits, and we therefore next determined whether inhibition of noradrenergic signaling during memory reactivation would reverse the cocaine-evoked plasticity and/or disrupt the cocaine-associated memory. We find that administration of the β-adrenergic receptor antagonist propranolol before memory retrieval, but not after (during memory reconsolidation), reverses the cocaine-evoked presynaptic and postsynaptic modifications in PL-mPFC and causes long-lasting memory impairments. Taken together, these data reveal that cocaine-evoked synaptic plasticity in PL-mPFC is reversible in vivo, and suggest a novel strategy that would allow normalization of prefrontal circuitry in addiction.
Collapse
Affiliation(s)
- James M Otis
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, WI, USA,Department of Psychiatry, University of North Carolina-Chapel Hill, Chapel Hill, NC, USA
| | - Devin Mueller
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, WI, USA,Department of Basic Sciences, Neuroscience Division, Ponce Health Sciences University-School of Medicine, Ponce Research Institute, Ponce, Puerto Rico,Department of Basic Sciences, Neuroscience Division, Ponce Health Sciences University-School of Medicine, Ponce Research Institute, P.O. Box 7004, Ponce 00732-7004, Puerto Rico, Tel: +1 787 840 2575 Ext. 2588, Fax: +1 787 844 1980, E-mail:
| |
Collapse
|
34
|
Verhasselt S, Stevens CV, Van den broecke T, Bracke ME, Roman BI. Insights into the myosin II inhibitory potency of A-ring-modified ( S )-blebbistatin analogs. Bioorg Med Chem Lett 2017; 27:2986-2989. [DOI: 10.1016/j.bmcl.2017.05.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2017] [Revised: 05/02/2017] [Accepted: 05/03/2017] [Indexed: 11/30/2022]
|
35
|
Mohamed RMP, Kumar J, Yap E, Mohamed IN, Sidi H, Adam RL, Das S. Try to Remember: Interplay between Memory and Substance Use Disorder. Curr Drug Targets 2017. [PMID: 28641520 DOI: 10.2174/1389450118666170622092824] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Memories associated with substance use disorders, or substance-associated cues increase the likelihood of craving and relapse during abstinence. There is a growing consensus that manipulation of synaptic plasticity may reduce the strength of substance abuse-related memories. On the biological front, there are new insights that suggest memories associated with substance use disorder may follow unique neurobiological pathways that render them more accessible to pharmacological intervention. In parallel to this, research in neurochemistry has identified several potential candidate molecules that could influence the formation and maintenance of long-term memory. Drugs that target these molecules (blebbistatin, isradipine and zeta inhibitory peptide) have shown promise at the preclinical stage. In this review, we shall provide an overview of the evolving understanding on the biochemical mechanisms involved in memory formation and expound on the premise that substance use disorder is a learning disorder.
Collapse
Affiliation(s)
- Rashidi Mohamed Pakri Mohamed
- Department of Psychological Medicine, Faculty of Medicine, University of Malaya, Lembah Pantai, 59100 Kuala Lumpur, Malaysia
| | - Jaya Kumar
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, 56000 Cheras, Kuala Lumpur, Malaysia
| | - Ernie Yap
- Department of Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, 56000 Cheras, Kuala Lumpur, Malaysia
| | - Isa Naina Mohamed
- Department of Pharmacology, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, 56000 Cheras, Kuala Lumpur, Malaysia
| | - Hatta Sidi
- Department of Psychiatry, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, 56000 Cheras, Kuala Lumpur, Malaysia
| | - Raja Lope Adam
- Department of Psychiatry, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, 56000 Cheras, Kuala Lumpur, Malaysia
| | - Srijit Das
- Department of Anatomy, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, 56000 Cheras, Kuala Lumpur, Malaysia
| |
Collapse
|
36
|
Briggs SB, Blouin AM, Young EJ, Rumbaugh G, Miller CA. Memory disrupting effects of nonmuscle myosin II inhibition depend on the class of abused drug and brain region. ACTA ACUST UNITED AC 2017; 24:70-75. [PMID: 28096495 PMCID: PMC5238718 DOI: 10.1101/lm.043976.116] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Accepted: 11/23/2016] [Indexed: 11/25/2022]
Abstract
Depolymerizing actin in the amygdala through nonmuscle myosin II inhibition (NMIIi) produces a selective, lasting, and retrieval-independent disruption of the storage of methamphetamine-associated memories. Here we report a similar disruption of memories associated with amphetamine, but not cocaine or morphine, by NMIIi. Reconsolidation appeared to be disrupted with cocaine. Unlike in the amygdala, methamphetamine-associated memory storage was not disrupted by NMIIi in the hippocampus, nucleus accumbens, or orbitofrontal cortex. NMIIi in the hippocampus did appear to disrupt reconsolidation. Identification of the unique mechanisms responsible for NMII-mediated, amygdala-dependent disruption of memory storage associated with the amphetamine class may enable induction of retrieval-independent vulnerability to other pathological memories.
Collapse
Affiliation(s)
- Sherri B Briggs
- Department of Metabolism & Aging, The Scripps Research Institute, Jupiter, Florida 33458, USA.,Department of Neuroscience, The Scripps Research Institute, Jupiter, Florida 33458, USA
| | - Ashley M Blouin
- Department of Metabolism & Aging, The Scripps Research Institute, Jupiter, Florida 33458, USA.,Department of Neuroscience, The Scripps Research Institute, Jupiter, Florida 33458, USA
| | - Erica J Young
- Department of Metabolism & Aging, The Scripps Research Institute, Jupiter, Florida 33458, USA.,Department of Neuroscience, The Scripps Research Institute, Jupiter, Florida 33458, USA
| | - Gavin Rumbaugh
- Department of Neuroscience, The Scripps Research Institute, Jupiter, Florida 33458, USA
| | - Courtney A Miller
- Department of Metabolism & Aging, The Scripps Research Institute, Jupiter, Florida 33458, USA.,Department of Neuroscience, The Scripps Research Institute, Jupiter, Florida 33458, USA
| |
Collapse
|
37
|
Young EJ, Briggs SB, Rumbaugh G, Miller CA. Nonmuscle myosin II inhibition disrupts methamphetamine-associated memory in females and adolescents. Neurobiol Learn Mem 2017; 139:109-116. [PMID: 28082169 DOI: 10.1016/j.nlm.2017.01.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Revised: 12/30/2016] [Accepted: 01/02/2017] [Indexed: 12/31/2022]
Abstract
Memories associated with drug use can trigger strong motivation for the drug, which increases relapse vulnerability in substance use disorder (SUD). Currently there are no treatments for relapse to abuse of psychostimulants, such as methamphetamine (METH). We previously reported that storage of memories associated with METH, but not those for fear or food reward, and the concomitant spine density increase are disrupted in a retrieval-independent manner by depolymerizing actin in the basolateral amygdala complex (BLC) of adult male rats and mice. Similar results are achieved in males through intra-BLC or systemic inhibition of nonmuscle myosin II (NMII), a molecular motor that directly drives actin polymerization. Given the substantial differences in physiology between genders, we sought to determine if this immediate and selective disruption of METH-associated memory extends to adult females. A single intra-BLC infusion of the NMII inhibitor Blebbistatin (Blebb) produced a long-lasting disruption of context-induced drug seeking for at least 30days in female rats that mirrored our prior results in males. Furthermore, a single systemic injection of Blebb prior to testing disrupted METH-associated memory and the concomitant increase in BLC spine density in females. Importantly, as in males, the same manipulation had no effect on an auditory fear memory or associated BLC spine density. In addition, we established that the NMII-based disruption of METH-associated memory extends to both male and female adolescents. These findings provide further support that small molecular inhibitors of NMII have strong therapeutic potential for the prevention of relapse to METH abuse triggered by associative memories.
Collapse
Affiliation(s)
- Erica J Young
- Department of Metabolism & Aging, The Scripps Research Institute, Jupiter, FL, USA; Department of Neuroscience, The Scripps Research Institute, Jupiter, FL, USA
| | - Sherri B Briggs
- Department of Metabolism & Aging, The Scripps Research Institute, Jupiter, FL, USA; Department of Neuroscience, The Scripps Research Institute, Jupiter, FL, USA
| | - Gavin Rumbaugh
- Department of Metabolism & Aging, The Scripps Research Institute, Jupiter, FL, USA; Department of Neuroscience, The Scripps Research Institute, Jupiter, FL, USA
| | - Courtney A Miller
- Department of Metabolism & Aging, The Scripps Research Institute, Jupiter, FL, USA; Department of Neuroscience, The Scripps Research Institute, Jupiter, FL, USA.
| |
Collapse
|
38
|
Gipson CD, Olive MF. Structural and functional plasticity of dendritic spines - root or result of behavior? GENES BRAIN AND BEHAVIOR 2016; 16:101-117. [PMID: 27561549 DOI: 10.1111/gbb.12324] [Citation(s) in RCA: 130] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Revised: 08/19/2016] [Accepted: 08/24/2016] [Indexed: 02/06/2023]
Abstract
Dendritic spines are multifunctional integrative units of the nervous system and are highly diverse and dynamic in nature. Both internal and external stimuli influence dendritic spine density and morphology on the order of minutes. It is clear that the structural plasticity of dendritic spines is related to changes in synaptic efficacy, learning and memory and other cognitive processes. However, it is currently unclear whether structural changes in dendritic spines are primary instigators of changes in specific behaviors, a consequence of behavioral changes, or both. In this review, we first examine the basic structure and function of dendritic spines in the brain, as well as laboratory methods to characterize and quantify morphological changes in dendritic spines. We then discuss the existing literature on the temporal and functional relationship between changes in dendritic spines in specific brain regions and changes in specific behaviors mediated by those regions. Although technological advancements have allowed us to better understand the functional relevance of structural changes in dendritic spines that are influenced by environmental stimuli, the role of spine dynamics as an underlying driver or consequence of behavior still remains elusive. We conclude that while it is likely that structural changes in dendritic spines are both instigators and results of behavioral changes, improved research tools and methods are needed to experimentally and directly manipulate spine dynamics in order to more empirically delineate the relationship between spine structure and behavior.
Collapse
Affiliation(s)
- C D Gipson
- Department of Psychology, Arizona State University, Tempe, AZ, USA
| | - M F Olive
- Department of Psychology, Arizona State University, Tempe, AZ, USA
| |
Collapse
|
39
|
Inhibition of actin polymerization in the NAc shell inhibits morphine-induced CPP by disrupting its reconsolidation. Sci Rep 2015; 5:16283. [PMID: 26538334 PMCID: PMC4633728 DOI: 10.1038/srep16283] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Accepted: 10/12/2015] [Indexed: 12/18/2022] Open
Abstract
Drug-associated contextual cues contribute to drug craving and relapse after abstinence, which is a major challenge to drug addiction treatment. Previous studies showed that disrupting memory reconsolidation impairs drug reward memory. However, the underlying mechanisms remain elusive. Although actin polymerization is involved in memory formation, its role in the reconsolidation of drug reward memory is unknown. In addition, the specific brain areas responsible for drug memory have not been fully identified. In the present study, we found that inhibiting actin polymerization in the nucleus accumbens (NAc) shell, but not the NAc core, abolishes morphine-induced conditioned place preference (CPP) by disrupting its reconsolidation in rats. Moreover, this effect persists for more than 2 weeks by a single injection of the actin polymerization inhibitor, which is not reversed by a morphine-priming injection. Furthermore, the application of actin polymerization inhibitor outside the reconsolidation window has no effect on morphine-associated contextual memory. Taken together, our findings first demonstrate that inhibiting actin polymerization erases morphine-induced CPP by disrupting its reconsolidation. Our study suggests that inhibition of actin polymerization during drug memory reconsolidation may be a potential approach to prevent drug relapse.
Collapse
|