Abstract

BackgroundMethamphetamine (METH)-associated alterations in the striatal dopamine (DA) system or dopamine transport (DAT) have been identified in clinical and preclinical studies with positron emission tomography (PET) imaging but have not been well correlated with in vivo serotonin transporter (SERT) availability due to the lack of appropriate imaging agents to assess SERTs. N,N-dimethyl-2-(2-amino-4-[18F]-fluorophenylthio) benzylamine (4-[18F]-ADAM) has been developed by our group and validated for its high affinity and selectivity for SERTs, allowing the in vivo examination of SERT density, location, and binding function. The aims of this study were to investigate the potential of SERT imaging using 4-[18F]-ADAM PET to estimate the long-lasting effects of METH-induced serotonergic neurotoxicity, and further determine whether a correlative relationship exists between SERT availability/activity and tyrosine hydroxylase (TH) activity in various brain regions due to the long-lasting consequences of METH treatment.ResultsMale rats received four administrations of METH (5 or 10 mg/kg, s.c.) or saline (1 ml/kg, s.c.) at 1-h intervals. At 30 days post-administration, in vivo SERT availability and activity were measured by 4-[18F]ADAM PET imaging. In contrast to the controls, the uptake of 4-[18F]ADAM in METH-treated mice was significantly reduced in a dose-dependent manner in the midbrain, followed by the hypothalamus, thalamus, striatum, hippocampus, and frontal cortex. The regional effects of METH on TH activity were assessed by quantitative immunohistochemistry and presented as integrated optical density (IOD). A significant decrease in TH immunostaining and IOD ratios was seen in the caudate, putamen, nucleus accumbens, substantia nigra pars compacta, and substantia nigra pars reticulata in the METH-treated rats compared to controls.ConclusionThe present results suggested that the long-lasting response to METH decreased the uptake of 4-[18F]-ADAM and varied regionally along with TH immunoreactivity. In addition, 4-[18F]ADAM PET could be used to detect serotonergic neuron loss and to evaluate the severity of serotonergic neurotoxicity of METH.

Highlights

  • Methamphetamine (METH)-associated alterations in the striatal dopamine (DA) system or dopamine transport (DAT) have been identified in clinical and preclinical studies with positron emission tomography (PET) imaging but have not been well correlated with in vivo serotonin transporter (SERT) availability due to the lack of appropriate imaging agents to assess serotonin transporters (SERTs)

  • The crucial role of the central serotoninergic system has been increasingly recognized in the field of addiction, as the reduction of serotonin transporters (SERTs) or upregulated serotonin levels contribute to the pathological mechanism and behavioral changes induced by drug abuse [1, 2]

  • The present PET imaging and IHC findings, together with previous studies [16], implied that real-time in vivo 4-[18F]ADAM PET imaging might provide a reliable biomarker to observe the effects of drugs such as METH on METH-induced changes in hydroxylase levels were associated with SERTs Current results demonstrated that tyrosine hydroxylase (TH) immunoreactivity in the caudate, putamen, nucleus accumbens, and substantia nigra was depleted after METH treatment

Read more

Summary

Introduction

Methamphetamine (METH)-associated alterations in the striatal dopamine (DA) system or dopamine transport (DAT) have been identified in clinical and preclinical studies with positron emission tomography (PET) imaging but have not been well correlated with in vivo serotonin transporter (SERT) availability due to the lack of appropriate imaging agents to assess SERTs. The crucial role of the central serotoninergic system has been increasingly recognized in the field of addiction, as the reduction of serotonin transporters (SERTs) or upregulated serotonin levels contribute to the pathological mechanism and behavioral changes induced by drug abuse [1, 2]. While SERTs are mainly located on serotonergic terminals and cell bodies in the brainstem nuclei [5], they are heterogeneously distributed in rat and human brains [6]. The binding of autoradiographic [3H]citalopram to its binding sites in SERTs in the human brain was highest in the limbic cortices, followed by the brainstem, striatum, pallidum, isocortex, and thalamus [8]

Objectives
Methods
Results
Discussion
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call