The peripheral benzodiazepine receptor ligand PK11195 binds with high affinity to the acute phase reactant α1-acid glycoprotein: implications for the use of the ligand as a CNS inflammatory marker1

https://doi.org/10.1016/S0969-8051(02)00410-9Get rights and content

Abstract

The peripheral benzodiazepine receptor ligand PK11195 has been used as an in vivo marker of neuroinflammation in positron emission tomography studies in man. One of the methodological issues surrounding the use of the ligand in these studies is the highly variable kinetic behavior of [11C]PK11195 in plasma. We therefore undertook a study to measure the binding of [3H]PK11195 to whole human blood and found a low level of binding to blood cells but extensive binding to plasma proteins. Binding assays using [3H]PK11195 and purified human plasma proteins demonstrated a strong binding to α1-acid glycoprotein (AGP) and a much weaker interaction with albumin. Immunodepletion of AGP from plasma resulted in the loss of plasma [3H]PK11195 binding demonstrating: (i) the specificity of the interaction and (ii) that AGP is the major plasma protein to which PK11195 binds with high affinity. PK11195 was able to displace fluorescein-dexamethasone from AGP with IC50 of <1.2 μM, consistent with a high affinity interaction. These findings are important for understanding the behavior of the ligand in positron emission tomography studies for three reasons. Firstly, AGP is an acute phase protein and its levels will vary during infection and pathological inflammatory diseases such as multiple sclerosis. This could significantly alter the free plasma concentrations of the ligand and contribute to its variable kinetic behavior. Secondly, AGP and AGP-bound ligand may contribute to the access of [11C]PK11195 to the brain parenchyma in diseases with blood brain barrier breakdown. Finally, local synthesis of AGP at the site of brain injury may contribute the pattern of [11C]PK11195 binding observed in neuroinflammatory diseases.

Introduction

PK11195 (1-[2-chlorophenyl]-N-methyl-N-[1-methyl-propyl]-3-isoquinoline carboxamide) is a high affinity ligand of the peripheral benzodiazepine receptor (PBR). Labeled with carbon-11, PK11195 has been used in positron emission tomography studies as an in vivo marker of neuroinflammation and injury in a range of diseases including multiple sclerosis (MS) [1], Alzheimer’s disease (AD) [2], Rasmussen’s encephalitis [3], ischemic stroke [4] and herpes simplex encephalitis [5]. It is believed that the increased binding of [11C]PK11195 seen in these studies is primarily due to the presence of activated microglia, the brain’s resident phagocytic cell, and that as such it may provide an in vivo marker of disease activity in the brain [1].

PBRs are present at low levels in normal central nervous tissue, located primarily on astrocytes and microglia [6]. A number of studies have found that in animal models of brain injury there is time-dependent increase in the binding of PK11195 at the site of damage [7], [8], [9], [10], [11], [12]. The increase in ligand binding is believed to be due to an upregulation in the expression of the PBR, although in a number of studies this has not been substantiated by immunohistochemical data [7], [10]. The question of which glial cell type in CNS disease is primarily responsible for the increased ligand binding is not fully resolved and the answer is most likely model dependent. In a rat experimental allergic encephalitis model of multiple sclerosis it has been demonstrated that activated microglia and macrophages are responsible for a significant part of the observed increase in PK11195 binding [1], [11]. However Kuhlmann and Guilarte [9] demonstrated that both activated astrocytes and microglia were capable of expressing high levels of PBR following neurotoxic injury induced by trimethyltin.

Despite the increasing use of [11C]PK11195 as a PET ligand in studies of human brain disease, there remain a number of methodological issues surrounding the interpretation of results from these studies [1], [2]. Firstly, the very low signal from [11C]PK11195 in the normal brain results in a poorly defined pattern of binding. This in turn makes it difficult to define normal reference tissue in diseases that are disseminated throughout the brain, such as AD and MS. Secondly, standard methods of extracting reference data from the plasma compartment are not possible due to the highly variable kinetic behavior of [11C]PK11195 in plasma. This unstable kinetic behavior has been attributed to variable plasma protein binding [1].

We therefore undertook a study to investigate the distribution of the ligand in blood and to identify its binding to plasma proteins. Our results demonstrate that PK11195 binds with high affinity to the acute phase reactant α1-acid glycoprotein and the relevance of this interaction with regards to the use of the ligand in PET studies is discussed.

Section snippets

Collection of blood and the preparation of plasma

Whole blood was collected from healthy volunteers into EDTA coated tubes and either used immediately or centrifuged at 10000g for 2 min at 4°C to prepare EDTA-plasma. EDTA-plasma was aliquoted on ice and frozen at –80°C. Aliquots of plasma were thawed only once and unused material discarded.

Measurement of the binding of [3H]PK11195 in whole blood

100 μl aliquots of freshly collected EDTA treated human blood were incubated with 10 nM [3H]PK11195 (specific activity 85.5 Ci/mol, PerkinElmer Life Sciences) and incubated at 20°C for the times indicated.

Results

Experiments studying [3H]PK11195 binding to whole blood indicated that (i) equilibrium was reached within five minutes and (ii) ∼15% of the ligand was distributed in pelleted material following low speed centrifugation (Figure 1). The pellet contained the cellular elements of blood and a small amount of trapped plasma. This indicated that the binding of PK11195 to blood cells from healthy volunteers is low and that the majority of the compound was distributed in the plasma.

To determine whether

Discussion

A number of studies examining the binding of [3H]PK11195 have reported the presence of the PBR in a wide range of blood cells [14], [15], [16]. Canat et al. [17] reported the highest levels of [3H]PK11195 binding to monocytes, polymorphonuclear cells and lymphocytes, and much lower levels of binding to platelets and erythrocytes (see Table 1). They demonstrated that granulocytes account for ∼70% of the total number of cellular PK11195 binding sites in whole blood, with the remaining sites being

Acknowledgements

We are grateful to Dr Murray Brown and Dr Andrew Pope for assistance with the fluorescent displacement assay and to Dr. Ron Leslie for helpful discussions.

References (30)

  • S. Pappata et al.

    Thalamic microglial activation in ischemic stroke detected in vivo by PET and [11C]PK11195

    Neurology

    (2000)
  • A. Cagnin et al.

    In vivo visualization of activated glia by [11C] (R)-PK11195-PET following herpes encephalitis reveals projected neuronal damage beyond the primary focal lesion

    Brain

    (2001)
  • M. Gavish et al.

    Enigma of the peripheral benzodiazepine receptor

    Pharmacol Rev

    (1999)
  • A.C. Kuhlmann et al.

    Regional and temporal expression of the peripheral benzodiazepine receptor in MPTP neurotoxicity

    Toxicol Sci

    (1999)
  • A.C. Kuhlmann et al.

    Cellular and subcellular localization of peripheral benzodiazepine receptors after trimethyltin neurotoxicity

    J Neurochem

    (2000)
  • Cited by (0)

    1

    List of abbreviations: AD, Alzheimer’s disease; AGP, α1-acid glycoprotein; APO, apolipoprotein A1; BBB, blood brain barrier; HSA, human serum albumin; MS, multiple sclerosis; PBR, peripheral benzodiazepine receptor; PBS, phosphate buffered saline; PET, positron emission tomography; PK11195, 1-[2-chlorophenyl]-N-methyl-N-[1-methyl-propyl]-3-isoquinoline carboxamide

    View full text