Elsevier

Schizophrenia Research

Volume 195, May 2018, Pages 206-214
Schizophrenia Research

Brain TSPO imaging and gray matter volume in schizophrenia patients and in people at ultra high risk of psychosis: An [11C]PBR28 study

https://doi.org/10.1016/j.schres.2017.08.063Get rights and content

Abstract

Patients with schizophrenia show whole brain and cortical gray matter (GM) volume reductions which are progressive early in their illness. Microglia, the resident immune cells in the CNS, phagocytose neurons and synapses. Some post mortem and in vivo studies in schizophrenia show evidence for elevated microglial activation compared to matched controls. However, it is currently unclear how these results relate to changes in cortical structure.

Methods

Fourteen patients with schizophrenia and 14 ultra high risk for psychosis (UHR) subjects alongside two groups of age and genotype matched healthy controls received [11C]PBR28 PET scans to index TSPO expression, a marker of microglial activation and a 3 T MRI scan. We investigated the relationship between the volume changes of cortical regions and microglial activation in cortical GM (as indexed by [11C]PBR28 distribution volume ratio (DVR).

Results

The total cortical GM volume was significantly lower in SCZ than the controls [mean (SD)/cm3: SCZ = 448.83 (39.2) and controls = 499.6 (59.2) (p = 0.02) but not in UHR (mean (SD) = 503.06 (57.9) and controls = 524.46 (45.3) p = 0.3). Regression model fitted the total cortical GM DVR values with the cortical regional volumes in SCZ (r = 0.81; p < 0.001) and in UHR (r = 0.63; p = 0.02). We found a significant negative correlation between the TSPO signal and total cortical GM volume in SCZ with the highest absolute correlation coefficient in the right superior-parietal cortex (r =  0.72; p = 0.006).

Conclusions

These findings suggest that microglial activity is related to the altered cortical volume seen in schizophrenia. Longitudinal investigations are required to determine whether microglial activation leads to cortical gray matter loss.

Introduction

Patients with schizophrenia have been consistently shown to have reduced whole brain and cortical gray matter volume relative to matched controls (Ellison-Wright et al., 2008, Haijma et al., 2013). Moreover, longitudinal brain morphometric studies show there are progressive reductions in gray and white matter volume in schizophrenia (Andreasen et al., 2011, Kahn and Sommer, 2015, van Haren et al., 2008). Similarly people at ultra high risk for psychosis (UHR), have reduced cortical and gray matter volumes (Dazzan et al., 2012), and demonstrate evidence for progressive loss during the transition to psychosis (Cannon et al., 2015, Pantelis et al., 2003, Sun et al., 2009, Ziermans et al., 2012). In contrast, elevations in white matter volume have been observed in schizophrenia, although less consistently (Walterfang et al., 2008, Witthaus et al., 2008). It is interesting to note that the change over time in cortical volume has been found to be related to change in symptoms and a transition from at risk symptoms to first episode psychosis (Borgwardt et al., 2007b). Baseline volumes have also been shown to act as a predictive marker for transition (Borgwardt et al., 2007a).

Though the exact pathophysiology of the brain morphometric changes in schizophrenia is not clear, immune dysregulation has been suggested as an underlying mechanism (Bergink et al., 2014, Fillman et al., 2016, Kirkpatrick and Miller, 2013, Laskaris et al., 2016). Supporting this, experimental animal studies of maternal immune activation show altered fetal brain development and behavioral and brain histopathological changes in offspring (Smith et al., 2007). Elevated levels of pro-inflammatory cytokines have been associated with the reductions in gray matter volume in patients with schizophrenia (Meisenzahl et al., 2001), and loss of gray matter in frontal cortex was associated with greater levels of pro-inflammatory cytokines in people at risk of psychosis who transitioned to psychosis (Cannon et al., 2015). Microglia, the brain's resident immune cells, have a major role in synaptic pruning and phagocytosis within the central nervous system (Kreutzberg, 1996, Paolicelli et al., 2011). They are both activated by pro-inflammatory cytokines and release them (Hanisch, 2002, Kreutzberg, 1996). A recent rodent model based on immune loci identified in the largest genome-wide association study in schizophrenia to date showed that these genetic alterations lead to altered microglial pruning of synapses (Sekar et al., 2016). Moreover, post mortem studies show elevated markers of inflammation (Fillman et al., 2013, Foster et al., 2006, van Kesteren et al., 2017, Volk et al., 2015) and elevated microglial cell density (with a hypertrophic morphology) in the brains of schizophrenia patients compared with healthy controls (Bayer et al., 1999), particularly in the frontal and temporal lobes (Radewicz et al., 2000, van Kesteren et al., 2017). A recent systematic review and meta-analysis of post mortem brain studies in schizophrenia found significant increase in the density of microglia in patients compared to healthy controls (van Kesteren et al., 2017) but another review did not find any consistent difference in microglia markers in schizophrenia (Trépanier et al., 2016). Microglial activation might thus disrupt synaptic pruning to result in the gray matter volume loss seen in schizophrenia (Laskaris et al., 2016). Supporting this, a recent post-mortem study has shown that greater inflammatory brain markers are associated with smaller brain volumes in schizophrenia (Zhang et al., 2016).

Microglia express the 18 kDa translocator specific protein (TSPO), a mitochondrial membrane protein, when they become activated, and can be indexed by PET tracers such as [11C]PBR28 that selectively bind to it (Brown et al., 2007, Owen et al., 2014, Owen et al., 2011). Thus [11C]PBR28 is an in vivo biomarker of microglial activation (Abourbeh et al., 2012, Dickens et al., 2014, Karlstetter et al., 2014, Lartey et al., 2014, Mattner et al., 2013, Rupprecht et al., 2010). We have recently reported increased [11C]PBR28 distribution volume ratio (DVR) in cortical gray matter in medicated patients with schizophrenia as well as UHR subjects when compared to healthy controls (Bloomfield et al., 2016b) using positron emission tomography (PET) imaging. This finding is consistent with some (Doorduin et al., 2009b, van Berckel et al., 2008), but not all PET studies, which either have reported no change (Collste et al., 2017, Coughlin et al., 2016, Hafizi et al., 2017, Holmes et al., 2016, Kenk et al., 2015, van der Doef et al., 2016) or a decrease in volume of distribution (VT) (Collste et al., 2017) in patients with psychosis or schizophrenia compared to healthy controls, suggesting there may be clinical or methodological differences between studies (Turkheimer et al., 2015b).

Microglial activation has been long associated with diaschisis: e.g. the phenomenon whereby local brain changes lead to functional alterations in areas distant from but connected to the initial area of damage (von Monakow in 1906) (Carrera and Tononi, 2014). As a result of diaschisis, secondary changes in the thalamus, substantia nigra pars reticulata, hippocampus and spinal cord have been well reported after focal ischemic or excitotoxic lesions of the cortex and/or striatum, and these are associated with inflammatory changes characterized by activation of microglia and astrocytes (Block et al., 2005). Notably, the facial nerve lesion model that Georg Kreutzberg introduced as the stereotypical preparation to study microglial response to injury in tissue with an intact blood-brain barrier, demonstrates microglial activation as a result of secondary injury (Blinzinger and Kreutzberg, 1968). PET imaging using [11C]-(R)-PK11195 has reported secondary thalamic and hippocampal increases in [11C]-(R)-PK11195 after stroke, traumatic brain injury, neurodegeneration and aging both in animal models and humans (Arlicot et al., 2010, Cagnin et al., 2001, Holmberg et al., 2009, Myers et al., 1991, Ouchi et al., 2005, Pappata et al., 2000, Ramlackhansingh et al., 2011, Schuitemaker et al., 2012a). The specificity of these findings has been elegantly demonstrated by Radlinska et al. (Radlinska et al., 2009) who showed [11C]-(R)-PK11195 increases in the spinal tract of patients with anterograde infarct as determined by diffusion tensor imaging. Notably, secondary microglia activation does not seem limited to anterograde and retrograde projections. Banati et al. (Banati et al., 2001) reported a trans‑synaptic increase in [11C]-(R)-PK11195 binding in the thalamus of a male patient with limb denervation. Subsequently, Gerhard et al.(Gerhard et al., 2005) reported [11C]-(R)-PK11195 increases in the ipsilateral thalamic nuclei of six stroke patients. A meta-analysis of post-mortem studies of microglia in schizophrenia indicate that alterations are seen across a number of brain regions (van Kesteren et al., 2017), and there is no clear evidence for regional specificity of microglial or TSPO changes, albeit relatively few regions have been investigated and studies are probably underpowered to detect regional differences (Bloomfield et al., 2016b, Collste et al., 2017, Coughlin et al., 2016, Doorduin et al., 2009b, Hafizi et al., 2017, Holmes et al., 2016, Kenk et al., 2015, van Berckel et al., 2008, van der Doef et al., 2016, van Kesteren et al., 2017). Our prior [11C]PBR28 TSPO study found an alteration in cortical gray matter throughout the brains of patients with schizophrenia (Bloomfield et al., 2016b). Given the above, we predicted that alterations in microglial in schizophrenia would be linked to the generalized structural cortical reductions seen in schizophrenia (Ellison-Wright and Bullmore, 2010). In view of this, we modeled [11C]PBR28 TSPO alterations in schizophrenia with a multivariate statistical model to capture the cross-correlation between alterations in the TSPO measure and structural changes across the brain. As this involved a large number of regions, we used an elastic net approach (Zou and Hastie, 2005) because this controls for the multiple potential cross-correlations. To our knowledge, to-date, no studies that have investigated the link between microglial activation with brain volumetric changes in vivo in schizophrenia and subjects at risk of psychosis. Our a priori hypothesis was that microglial activity would inversely correlate with the total gray matter volume in schizophrenia patients and UHR subjects (increased [11C]PBR28 in patients with more gray matter volume deficits).

Section snippets

Subjects

Fourteen subjects meeting UHR criteria, as assessed on the comprehensive assessment of the at risk mental state (CAARMS) (Yung et al., 2005), were recruited from a London mental health clinic (Mean age ± SD: 24.3 ± 5.40; (M:F = 7:7)). Fourteen subjects with schizophrenia (Mean age ± SD: 47.0 ± 9.31; (M:F = 12:2)) were recruited from London mental health centres (South London and Maudsley NHS Foundation Trust). A pool of 22 healthy control subjects recruited through newspaper and poster adverts were used to

Cortical gray matter volume analysis (SCZ vs controls)

The total cortical gray matter volume was significantly lower in SCZ than the healthy controls (p = 0.02) (Table 1). Temporal cortical gray matter volume was also lower in Scz than controls but not frontal cortical gray matter volume (Table 1). Whole brain analysis with false discovery rate set at 0.05, revealed significantly lower gray matter volumes in schizophrenia patients compared to healthy control subjects in the following three regions; left cuneus, right superior parietal cortex and

Discussion

In this experiment, we investigated the relationship between cortical volumes and TSPO signal from [11C]PBR28 PET scans in patients with schizophrenia, UHR subjects and respective matched control groups. We demonstrated that patients with schizophrenia exhibit a negative relationship between total cortical gray matter [11C]PBR-28 DVR and cortical gray matter volumes, and this was also seen for temporal cortex DVR and temporal cortical volumes. The lack of relationship in the healthy control

Limitations

Except one patient with schizophrenia, all were taking antipsychotic medications (Supplementary Table 2) which may be a potential confounder moderating the relationship between microglial activity and brain GM volume. Antipsychotic medications are associated with brain volume reductions in schizophrenia (Ho et al., 2011) but volume change is related to illness progression after adjusting for antipsychotic effects (Van Haren et al., 2013). Some preclinical evidence suggests that antipsychotic

Future directions

The causal relationship between neuroinflammation and brain volume changes will likely require longitudinal clinical studies that specifically look into whether TSPO changes predict the onset or worsening of brain volume reduction in individuals prior to the onset of psychosis and are antipsychotic medication naïve.

Conclusions

In schizophrenia, cortical volume reduction is associated with elevated microglial activation using a multimodal neuroimaging approach. This relationship was also observed at a marginal level in UHR subjects suggesting the relationship may become stronger with the development of the disorder.

The following are the supplementary data related to this article.

.

Funding

This study was funded by Medical Research Council-UK (no. MC-A656-5QD30), Maudsley Charity (no. 666), Brain and Behavior Research Foundation, and Wellcome Trust (no. 094849/Z/10/Z) grants to Dr. Howes and the National Institute for Health Research (NIHR) Biomedical Research Centre at South London and Maudsley NHS Foundation Trust and King's College London.

The views expressed are those of the author(s) and not necessarily those of the NHS, the NIHR or the Department of Health.

The authors would

Conflicts of interest

Dr. Howes has received investigator-initiated research funding from and/or participated in advisory/speaker meetings organised by Astra-Zeneca, Autifony, BMS, Eli Lilly, Heptares, Jansenn, Lundbeck, Lyden-Delta, Otsuka, Servier, Sunovion, Rand and Roche. Neither Dr. Howes or his family have been employed by or have holdings/a financial stake in any biomedical company. Sudhakar Selvaraj, Peter S Bloomfield, Cao Bo, Mattia Veronese and Federico Turkheimer have no conflicts of interest for this

Contributors

SS, PSB and ODH designed the study and wrote the protocol. SS and PSB recruited the subjects and administered clinical assessment. CB, MV and FT worked on image processing and analysis. PSB, CB and MV did statistical analyses and wrote the first draft of the manuscript, along with other authors. All authors contributed to the interpretation of the data, revised the manuscript and agreed with the final content of the manuscript.

References (107)

  • A.M. Dale et al.

    Cortical surface-based analysis. I. Segmentation and surface reconstruction

    NeuroImage

    (1999)
  • R.S. Desikan et al.

    An automated labeling system for subdividing the human cerebral cortex on MRI scans into gyral based regions of interest

    NeuroImage

    (2006)
  • I. Ellison-Wright et al.

    Anatomy of bipolar disorder and schizophrenia: a meta-analysis

    Schizophr. Res.

    (2010)
  • A. Gerhard et al.

    Evolution of microglial activation in patients after ischemic stroke: a [11C](R)-PK11195 PET study

    NeuroImage

    (2005)
  • N.E. van Haren et al.

    Progressive brain volume loss in schizophrenia over the course of the illness: evidence of maturational abnormalities in early adulthood

    Biol. Psychiatry

    (2008)
  • M. Imaizumi et al.

    PET imaging with [11C]PBR28 can localize and quantify upregulated peripheral benzodiazepine receptors associated with cerebral ischemia in rat

    Neurosci. Lett.

    (2007)
  • G.W. Kreutzberg

    Microglia: a sensor for pathological events in the CNS

    Trends Neurosci.

    (1996)
  • A. Lockhart et al.

    The peripheral benzodiazepine receptor ligand PK11195 binds with high affinity to the acute phase reactant alpha1-acid glycoprotein: implications for the use of the ligand as a CNS inflammatory marker

    Nucl. Med. Biol.

    (2003)
  • R. Myers et al.

    [3H]PK 11195 and the localisation of secondary thalamic lesions following focal ischaemia in rat motor cortex

    Neurosci. Lett.

    (1991)
  • C. Pantelis et al.

    Neuroanatomical abnormalities before and after onset of psychosis: a cross-sectional and longitudinal MRI comparison

    Lancet

    (2003)
  • V.L.R. Rao et al.

    Characterization of binding sites for the ω3 receptor ligands [3H]PK11195 and [3H]RO5-4864 in human brain

    Eur. J. Pharmacol.

    (1997)
  • A. Schuitemaker et al.

    Microglial activation in healthy aging

    Neurobiol. Aging

    (2012)
  • A. Schuitemaker et al.

    Microglial activation in healthy aging

    Neurobiol. Aging

    (2012)
  • D. Sun et al.

    Progressive brain structural changes mapped as psychosis develops in 'at risk' individuals

    Schizophr. Res.

    (2009)
  • A.C. Tziortzi et al.

    Imaging dopamine receptors in humans with [11C]-(+)-PHNO: dissection of D3 signal and anatomy

    NeuroImage

    (2011)
  • N.E. Van Haren et al.

    Confounders of excessive brain volume loss in schizophrenia

    Neurosci. Biobehav. Rev.

    (2013)
  • G. Abourbeh et al.

    Imaging microglial/macrophage activation in spinal cords of experimental autoimmune encephalomyelitis rats by positron emission tomography using the mitochondrial 18 kDa translocator protein radioligand [18F]DPA-714

    J. Neurosci.

    (2012)
  • D.S. Albrecht et al.

    In vivo imaging of human neuroinflammation

    ACS Chem. Neurosci.

    (2016)
  • N. Arlicot et al.

    Detection and quantification of remote microglial activation in rodent models of focal ischaemia using the TSPO radioligand CLINDE

    Eur. J. Nucl. Med. Mol. Imaging

    (2010)
  • R.B. Banati et al.

    Long-term trans‑synaptic glial responses in the human thalamus after peripheral nerve injury

    Neuroreport

    (2001)
  • Y. Benjamini et al.

    Controlling the false discovery rate: a practical and powerful approach to multiple testing

    J. R. Stat. Soc. Ser. B Methodol.

    (1995)
  • K. Blinzinger et al.

    Displacement of synaptic terminals from regenerating motoneurons by microglial cells

    Z. Zellforsch. Mikrosk. Anat.

    (1968)
  • P.S. Bloomfield et al.

    Response to Narendran and Frankle: the interpretation of PET microglial imaging in schizophrenia

    Am. J. Psychiatry

    (2016)
  • P.S. Bloomfield et al.

    Microglial activity in people at ultra high risk of psychosis and in schizophrenia: an [(11)C]PBR28 PET brain imaging study

    Am. J. Psychiatry

    (2016)
  • S.J. Borgwardt et al.

    Structural brain abnormalities in individuals with an at-risk mental state who later develop psychosis

    Br. J. Psychiatry Suppl.

    (2007)
  • A.K. Brown et al.

    Radiation dosimetry and biodistribution in monkey and man of 11C-PBR28: a PET radioligand to image inflammation

    J. Nucl. Med.

    (2007)
  • 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)
  • E. Carrera et al.

    Diaschisis: past, present, future

    Brain

    (2014)
  • K. Collste et al.

    Lower Levels of the Glial Cell Marker TSPO in Drug-Naive First-Episode Psychosis Patients as Measured Using PET and [(11)C]PBR28

    (2017)
  • A.K. Converse et al.

    11C-(R)-PK11195 PET imaging of microglial activation and response to minocycline in zymosan-treated rats

    J. Nucl. Med.

    (2011)
  • J.M. Coughlin et al.

    In vivo markers of inflammatory response in recent-onset schizophrenia: a combined study using [(11)C]DPA-713 PET and analysis of CSF and plasma

    Transl. Psychiatry

    (2016)
  • P. Dazzan et al.

    Volumetric abnormalities predating the onset of schizophrenia and affective psychoses: an MRI study in subjects at ultrahigh risk of psychosis

    Schizophr. Bull.

    (2012)
  • A.M. Dickens et al.

    Detection of microglial activation in an acute model of neuroinflammation using PET and radiotracers 11C-(R)-PK11195 and 18F-GE-180

    J. Nucl. Med.

    (2014)
  • R. Dimber et al.

    Evidence of brain inflammation in patients with human T-Lymphotropic virus type 1-associated myelopathy (HAM): a pilot, multimodal imaging study using 11C-PBR28 PET, MR T1-weighted, and diffusion-weighted imaging

    J. Nucl. Med.

    (2016)
  • T.F. van der Doef et al.

    In vivo (R)-[(11)C]PK11195 PET imaging of 18 kDa translocator protein in recent onset psychosis

    NPJ Schizophr.

    (2016)
  • J. Doorduin et al.

    Neuroinflammation in schizophrenia-related psychosis: a PET study

    J. Nucl. Med.

    (2009)
  • J. Doorduin et al.

    Neuroinflammation in schizophrenia-related psychosis: a PET study

    J. Nucl. Med.

    (2009)
  • I. Ellison-Wright et al.

    The anatomy of first-episode and chronic schizophrenia: an anatomical likelihood estimation meta-analysis

    Am. J. Psychiatry

    (2008)
  • S.G. Fillman et al.

    Increased inflammatory markers identified in the dorsolateral prefrontal cortex of individuals with schizophrenia

    Mol. Psychiatry

    (2013)
  • S.G. Fillman et al.

    Elevated peripheral cytokines characterize a subgroup of people with schizophrenia displaying poor verbal fluency and reduced Broca's area volume

    Mol. Psychiatry

    (2016)
  • Cited by (41)

    • Novel pharmacological strategies to treat cognitive dysfunction in chronic obstructive pulmonary disease

      2022, Pharmacology and Therapeutics
      Citation Excerpt :

      Importantly, they assessed blood pressures and oxygen/carbon dioxide concentrations and found a positive correlation between the PaO2 in the arterioles and reduction in grey matter density in people with COPD compared to people without COPD (Zhang et al., 2013). Other neurological diseases such as multiple sclerosis, bipolar disease and Alzheimer's disease have also shown correlations between reduction in grey matter volume and/or white matter hyperintensities (representing small white matter lesions observed in T2-weighted MRIs) and neuroinflammation (Datta et al., 2017; Selvaraj et al., 2018; Singhal et al., 2019; Tsai et al., 2019; Zhang et al., 2016). Currently there are limited studies linking neuroinflammation with grey and white matter alterations in COPD patients, however, we could speculate that the spillage of pulmonary inflammation and oxidative stress into the CNS induces alterations in the structure of grey and white matter in the brain thus contributing to cognitive dysfunction in people with COPD.

    • Changes in glial gene expression in the prefrontal cortex in relation to major depressive disorder, suicide and psychotic features

      2021, Journal of Affective Disorders
      Citation Excerpt :

      In addition, a distinction was noticed related to the psychotic symptoms in MDD. In vivo, a psychotic features-dependent reduction in frontal grey matter volume was noticed in TSPO imaging, which indicates that MDD pathophysiology may at least in part be driven by microglial deficits (Collste et al., 2017; Selvaraj et al., 2018). In our data, the DLPFC did not show psychotic features-related alterations in TSPO expression, which is consistent with an imaging study on patients with a high risk for psychotic features (Hafizi et al., 2018a, 2018b).

    • Brain morphology is differentially impacted by peripheral cytokines in schizophrenia-spectrum disorder

      2021, Brain, Behavior, and Immunity
      Citation Excerpt :

      The ‘neuroinflammatory’ hypothesis states that the cytotoxic effects of persistent activation of the brain’s immune cells, namely microglia and astrocytes, in addition to dysregulation of the peripheral inflammatory system, might cause neuronal damage and synaptic dysfunction (Banati and Hickie, 2009; Howes and McCutcheon, 2016; Kirkpatrick and Miller, 2013; Laskaris et al., 2016; Monji et al., 2009), thus potentially contributing to the thickness and volume loss seen in imaging studies of schizophrenia (Borgwardt et al., 2007; Olabi et al., 2011; Pantelis et al., 2005). Some studies have shown increased microglial activation at various stages of SSD (Bloomfield et al., 2016; Doorduin et al., 2009; Selvaraj et al., 2018; van Berckel et al., 2008), while several studies (Goldsmith et al., 2016; Miller et al., 2011a; Rodrigues-Amorim et al., 2018) have concluded that there is dysregulation of peripheral inflammatory molecules across all stages of the illness. Although the brain was once considered to be immune privileged, over the last decade, this has been reconceptualised.

    • Predictive validity of conversion from the clinical high risk syndrome to frank psychosis

      2020, Schizophrenia Research
      Citation Excerpt :

      A recent meta-analysis concluded that 22% of at-risk patients convert to psychosis within one year, 29% within two years, and 36% within three years (Fusar-Poli et al., 2012). Structured interviews to diagnose CHR (Fusar-Poli et al., 2015; McGlashan et al., 2010; Yung et al., 2004a) have been widely employed, as both research and clinical tools, (Allen et al., 2018; Bossong et al., 2018; Cannon et al., 2015; Cao et al., 2018; Corcoran et al., 2018; Davies et al., 2018a; Davies et al., 2018b; Dutt et al., 2015; Egerton et al., 2014; Fusar-Poli et al., 2013b; Ho et al., 2017; Howes et al., 2011; Jagannath et al., 2017; Jeffries et al., 2016; Jeffries et al., 2018; Koutsouleris et al., 2018; Lepock et al., 2018; McGorry et al., 2008; Miller et al., 2002; Modinos et al., 2018; NHS England, 2016; Schmidt et al., 2017; Schneider et al., 2016; Seidman et al., 2016; Selvaraj et al., 2018; US Substance Abuse and Mental Health Services Administration, 2018; Yung, 2017; Yung et al., 2005). The reliability and validity of one such tool, the Structured Interview for Psychosis-risk Syndromes (SIPS) (McGlashan et al., 2010), have each been reported in >20 samples (Addington et al., 2011; Carrión et al., 2018; Koike et al., 2013; Liu et al., 2011; McFarlane et al., 2015; Miller et al., 2002; Raballo et al., 2018; Simon et al., 2012; Tso et al., 2017; Waltz et al., 2015), and the large majority of reported data strongly support the accuracy of the instrument (Powers III et al., in press; Woods et al., 2019).

    View all citing articles on Scopus
    1

    Authors contributed equally to this manuscript.

    View full text