Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Michael D. Scofield is active.

Publication


Featured researches published by Michael D. Scofield.


The Neuroscientist | 2014

Astrocytic Dysfunction and Addiction Consequences of Impaired Glutamate Homeostasis

Michael D. Scofield; Peter W. Kalivas

Addiction is characterized as a chronic relapsing disorder whereby addicted individuals persistently engage in drug seeking and use despite profound negative consequences. The results of studies using animal models of addiction and relapse indicate that drug seeking is mediated by alterations in cortico-accumbal plasticity induced by chronic drug exposure. Among the maladaptive responses to drug exposure are long-lasting alterations in the expression of proteins localized to accumbal astrocytes, which are responsible for maintaining glutamate homeostasis. These alterations engender an aberrant potentiation of glutamate transmission in the cortico-accumbens circuit that is linked to the reinstatement of drug seeking. Accordingly, pharmacological restoration of glutamate homeostasis functions as an efficient method of reversing drug-induced plasticity and inhibiting drug seeking in both rodents and humans.


Pharmacological Reviews | 2016

The Nucleus Accumbens: Mechanisms of Addiction across Drug Classes Reflect the Importance of Glutamate Homeostasis

Michael D. Scofield; Jasper A. Heinsbroek; Cassandra D. Gipson; Y. M. Kupchik; Sade Spencer; Alexander C.W. Smith; Douglas Roberts-Wolfe; Peter W. Kalivas

The nucleus accumbens is a major input structure of the basal ganglia and integrates information from cortical and limbic structures to mediate goal-directed behaviors. Chronic exposure to several classes of drugs of abuse disrupts plasticity in this region, allowing drug-associated cues to engender a pathologic motivation for drug seeking. A number of alterations in glutamatergic transmission occur within the nucleus accumbens after withdrawal from chronic drug exposure. These drug-induced neuroadaptations serve as the molecular basis for relapse vulnerability. In this review, we focus on the role that glutamate signal transduction in the nucleus accumbens plays in addiction-related behaviors. First, we explore the nucleus accumbens, including the cell types and neuronal populations present as well as afferent and efferent connections. Next we discuss rodent models of addiction and assess the viability of these models for testing candidate pharmacotherapies for the prevention of relapse. Then we provide a review of the literature describing how synaptic plasticity in the accumbens is altered after exposure to drugs of abuse and withdrawal and also how pharmacological manipulation of glutamate systems in the accumbens can inhibit drug seeking in the laboratory setting. Finally, we examine results from clinical trials in which pharmacotherapies designed to manipulate glutamate systems have been effective in treating relapse in human patients. Further elucidation of how drugs of abuse alter glutamatergic plasticity within the accumbens will be necessary for the development of new therapeutics for the treatment of addiction across all classes of addictive substances.


The Journal of Neuroscience | 2014

Synaptic Glutamate Spillover Due to Impaired Glutamate Uptake Mediates Heroin Relapse

Hao-Wei Shen; Michael D. Scofield; Heather A. Boger; Megan Hensley; Peter W. Kalivas

Reducing the enduring vulnerability to relapse is a therapeutic goal in treating drug addiction. Studies with animal models of drug addiction show a marked increase in extrasynaptic glutamate in the core subcompartment of the nucleus accumbens (NAcore) during reinstated drug seeking. However, the synaptic mechanisms linking drug-induced changes in extrasynaptic glutamate to relapse are poorly understood. Here, we discovered impaired glutamate elimination in rats extinguished from heroin self-administration that leads to spillover of synaptically released glutamate into the nonsynaptic extracellular space in NAcore and investigated whether restoration of glutamate transport prevented reinstated heroin seeking. Through multiple functional assays of glutamate uptake and analyzing NMDA receptor-mediated currents, we show that heroin self-administration produced long-lasting downregulation of glutamate uptake and surface expression of the transporter GLT-1. This downregulation was associated with spillover of synaptic glutamate to extrasynaptic NMDA receptors within the NAcore. Ceftriaxone restored glutamate uptake and prevented synaptic glutamate spillover and cue-induced heroin seeking. Ceftriaxone-induced inhibition of reinstated heroin seeking was blocked by morpholino-antisense targeting GLT-1 synthesis. These data reveal that the synaptic glutamate spillover in the NAcore results from reduced glutamate transport and is a critical pathophysiological mechanism underling reinstated drug seeking in rats extinguished from heroin self-administration.


Biological Psychiatry | 2015

Gq-DREADD Selectively Initiates Glial Glutamate Release and Inhibits Cue-induced Cocaine Seeking.

Michael D. Scofield; Heather A. Boger; Rachel J. Smith; Hao Li; Philip G. Haydon; Peter W. Kalivas

BACKGROUND Glial cells of the central nervous system directly influence neuronal activity by releasing neuroactive small molecules, including glutamate. Long-lasting cocaine-induced reductions in extracellular glutamate in the nucleus accumbens core (NAcore) affect synaptic plasticity responsible for relapse vulnerability. METHODS We transduced NAcore astrocytes with an adeno-associated virus vector expressing hM3D designer receptor exclusively activated by a designer drug (DREADD) under control of the glial fibrillary acidic protein promoter in 62 male Sprague Dawley rats, 4 dominant-negative soluble N-ethylmaleimide-sensitive factor attachment protein receptor mice, and 4 wild-type littermates. Using glutamate biosensors, we measured NAcore glutamate levels following intracranial or systemic administration of clozapine N-oxide (CNO) and tested the ability of systemic CNO to inhibit reinstated cocaine or sucrose seeking following self-administration and extinction training. RESULTS Administration of CNO in glial fibrillary acidic protein-hM3D-DREADD transfected animals increased NAcore extracellular glutamate levels in vivo. The glial origin of released glutamate was validated by an absence of CNO-mediated release in mice expressing a dominant-negative soluble N-ethylmaleimide-sensitive factor attachment protein receptor variant in glia. Also, CNO-mediated release was relatively insensitive to N-type calcium channel blockade. Systemic administration of CNO inhibited cue-induced reinstatement of cocaine seeking in rats extinguished from cocaine but not sucrose self-administration. The capacity to inhibit reinstated cocaine seeking was prevented by systemic administration of the group II metabotropic glutamate receptor antagonist LY341495. CONCLUSIONS DREADD-mediated glutamate gliotransmission inhibited cue-induced reinstatement of cocaine seeking by stimulating release-regulating group II metabotropic glutamate receptor autoreceptors to inhibit cue-induced synaptic glutamate spillover.


Addiction Biology | 2015

Glutamate transporter GLT‐1 mediates N‐acetylcysteine inhibition of cocaine reinstatement

Kathryn J. Reissner; Cassandra D. Gipson; Phuong K. Tran; Lori A. Knackstedt; Michael D. Scofield; Peter W. Kalivas

Both pre‐clinical and clinical studies indicate that N‐acetylcysteine (NAC) may be useful in treating relapse to addictive drug use. Cocaine self‐administration in rats reduces both cystine‐glutamate exchange and glutamate transport via GLT‐1 in the nucleus accumbens, and NAC treatment normalizes these two glial processes critical for maintaining glutamate homeostasis. However, it is not known if one or both of these actions by NAC is needed to inhibit relapse to cocaine seeking. To determine whether the restoration of GLT‐1 and/or cystine‐glutamate exchange is required for NAC to inhibit cue‐induced reinstatement of cocaine seeking, we utilized the rat self‐administration/extinction/reinstatement model of cocaine relapse. Rats were pre‐treated in the nucleus accumbens with vivo‐morpholino antisense oligomers targeting either GLT‐1 or xCT (catalytic subunit of the cystine‐glutamate exchanger) overlapping with daily NAC administration during extinction (100 mg/kg, i.p. for the last 5 days). Rats then underwent cue‐induced reinstatement of active lever pressing in the absence of NAC, to determine if preventing NAC‐induced restoration of one or the other protein was sufficient to block the capacity of chronic NAC to inhibit reinstatement. The vivo‐morpholino suppression of xCT reduced cystine‐glutamate exchange but did not affect NAC‐induced reduction of reinstated cocaine seeking. In contrast, suppressing NAC‐induced restoration of GLT‐1 not only prevented NAC from inhibiting reinstatement, but augmented the capacity of cues to reinstate cocaine seeking. We hypothesized that the increased reinstatement after inhibiting NAC induction of GLT‐1 resulted from increased extracellular glutamate, and show that augmented reinstatement is prevented by blocking mGluR5. Restoring GLT‐1, not cystine‐glutamate exchange, is a key mechanism whereby daily NAC reduces cue‐induced cocaine reinstatement.


Nature Neuroscience | 2014

Synaptic plasticity mediating cocaine relapse requires matrix metalloproteinases

Alexander C.W. Smith; Yonatan M. Kupchik; Michael D. Scofield; Cassandra D. Gipson; Armina Wiggins; Charles A. Thomas; Peter W. Kalivas

Relapse to cocaine use necessitates remodeling excitatory synapses in the nucleus accumbens and synaptic reorganization requires matrix metalloproteinase (MMP) degradation of the extracellular matrix proteins. We found enduring increases in MMP-2 activity in rats after withdrawal from self-administered cocaine and transient increases in MMP-9 during cue-induced cocaine relapse. Cue-induced heroin and nicotine relapse increased MMP activity, and increased MMP activity was required for both cocaine relapse and relapse-associated synaptic plasticity.


Biological Psychiatry | 2016

Cocaine Self-Administration and Extinction Leads to Reduced Glial Fibrillary Acidic Protein Expression and Morphometric Features of Astrocytes in the Nucleus Accumbens Core

Michael D. Scofield; Hao Li; Benjamin M. Siemsen; Kati L. Healey; Phuong K. Tran; Nicholas Woronoff; Heather A. Boger; Peter W. Kalivas; Kathryn J. Reissner

BACKGROUND As a more detailed picture of nervous system function emerges, diversity of astrocyte function becomes more widely appreciated. While it has been shown that cocaine experience impairs astroglial glutamate uptake and release in the nucleus accumbens (NAc), few studies have explored effects of self-administration on the structure and physiology of astrocytes. We investigated the effects of extinction from daily cocaine self-administration on astrocyte characteristics including glial fibrillary acidic protein (GFAP) expression, surface area, volume, and colocalization with a synaptic marker. METHODS Cocaine or saline self-administration and extinction were paired with GFAP Westerns, immunohistochemistry, and fluorescent imaging of NAc core astrocytes (30 saline-administering and 36 cocaine-administering male Sprague Dawley rats were employed). Imaging was performed using a membrane-tagged lymphocyte protein tyrosine kinase-green fluorescent protein (Lck-GFP) driven by the GFAP promoter, coupled with synapsin I immunohistochemistry. RESULTS GFAP expression was significantly reduced in the NAc core following cocaine self-administration and extinction. Similarly, we observed an overall smaller surface area and volume of astrocytes, as well as reduced colocalization with synapsin I, in cocaine-administering animals. Cocaine-mediated reductions in synaptic contact were reversed by the β-lactam antibiotic ceftriaxone. CONCLUSIONS Multiple lines of investigation indicate that NAc core astrocytes exist in a hyporeactive state following cocaine self-administration and extinction. Decreased association with synaptic elements may be particularly meaningful, as cessation of chronic cocaine use is associated with changes in synaptic strength and resistance to the induction of synaptic plasticity. We hypothesize that the reduced synaptic colocalization of astrocytes represents an important maladaptive cellular response to cocaine and the mechanisms underlying relapse vulnerability.


Brain Research | 2015

The tetrapartite synapse: Extracellular matrix remodeling contributes to corticoaccumbens plasticity underlying drug addiction.

Alexander C.W. Smith; Michael D. Scofield; Peter W. Kalivas

Synaptic plasticity has long been known to involve three key elements of neuropil, the presynapse, the postsynapse and adjacent glia. Here we review the role of the extracellular matrix in synaptic plasticity as a necessary component forming the tetrapartite synapse. We describe the role of matrix metalloproteinases as enzymes sculpting extracellular proteins and thereby creating an extracellular signaling domain required for synaptic plasticity. Specifically we focus on the role of the tetrapartite synapse in mediating the effects of addictive drugs at cortico-striatal synapses, and conclude that the extracellular signaling domain and its regulation by matrix metalloproteinases is critical for developing and expressing drug seeking behaviors.


Addiction Biology | 2014

Rapid, transient potentiation of dendritic spines in context-induced relapse to cocaine seeking

Neringa M. Stankeviciute; Michael D. Scofield; Peter W. Kalivas; Cassandra D. Gipson

Addiction to cocaine produces long‐lasting, stable changes in brain synaptic physiology that might contribute to the vulnerability to relapse. In humans, exposure to environmental contexts previously paired with drug use precipitates relapse, but the neurobiological mechanisms mediating this process are unknown. Initiation of cocaine relapse via re‐exposure to a drug‐associated context elicited reinstatement of cocaine seeking as well as rapid, transient synaptic plasticity in the nucleus accumbens core (NAcore), measured as an increase in dendritic spine diameter. These results show that rapid context‐evoked synaptic potentiation in the NAcore may underpin relapse to cocaine use.


The Journal of Neuroscience | 2014

Cocaine Dysregulates Opioid Gating of GABA Neurotransmission in the Ventral Pallidum

Yonatan M. Kupchik; Michael D. Scofield; Kenner C. Rice; Kejun Cheng; Bernard P. Roques; Peter W. Kalivas

The ventral pallidum (VP) is a target of dense nucleus accumbens projections. Many of these projections coexpress GABA and the neuropeptide enkephalin, a δ and μ opioid receptor (MOR) ligand. Of these two, the MOR in the VP is known to be involved in reward-related behaviors, such as hedonic responses to palatable food, alcohol intake, and reinstatement of cocaine seeking. Stimulating MORs in the VP decreases extracellular GABA, indicating that the effects of MORs in the VP on cocaine seeking are via modulating GABA neurotransmission. Here, we use whole-cell patch-clamp on a rat model of withdrawal from cocaine self-administration to test the hypothesis that MORs presynaptically regulate GABA transmission in the VP and that cocaine withdrawal changes the interaction between MORs and GABA. We found that in cocaine-extinguished rats pharmacological activation of MORs no longer presynaptically inhibited GABA release, whereas blocking the MORs disinhibited GABA release. Moreover, MOR-dependent long-term depression of GABA neurotransmission in the VP was lost in cocaine-extinguished rats. Last, GABA neurotransmission was found to be tonically suppressed in cocaine-extinguished rats. These substantial synaptic changes indicated that cocaine was increasing tone on MOR receptors. Accordingly, increasing endogenous tone by blocking the enzymatic degradation of enkephalin inhibited GABA neurotransmission in yoked saline rats but not in cocaine-extinguished rats. In conclusion, our results indicate that following withdrawal from cocaine self-administration enkephalin levels in the VP are elevated and the opioid modulation of GABA neurotransmission is impaired. This may contribute to the difficulties withdrawn addicts experience when trying to resist relapse.

Collaboration


Dive into the Michael D. Scofield's collaboration.

Top Co-Authors

Avatar

Peter W. Kalivas

Medical University of South Carolina

View shared research outputs
Top Co-Authors

Avatar

Cassandra D. Gipson

Medical University of South Carolina

View shared research outputs
Top Co-Authors

Avatar

Heather A. Boger

Medical University of South Carolina

View shared research outputs
Top Co-Authors

Avatar

Alexander C.W. Smith

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Constanza Garcia-Keller

Medical University of South Carolina

View shared research outputs
Top Co-Authors

Avatar

Jasper A. Heinsbroek

Medical University of South Carolina

View shared research outputs
Top Co-Authors

Avatar

Sade Spencer

Medical University of South Carolina

View shared research outputs
Top Co-Authors

Avatar

Yonatan M. Kupchik

Medical University of South Carolina

View shared research outputs
Top Co-Authors

Avatar

Carmela M. Reichel

Medical University of South Carolina

View shared research outputs
Top Co-Authors

Avatar

Daniela Neuhofer

Medical University of South Carolina

View shared research outputs
Researchain Logo
Decentralizing Knowledge