Network


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

Hotspot


Dive into the research topics where Ansel T. Hillmer is active.

Publication


Featured researches published by Ansel T. Hillmer.


Neurobiology of Aging | 2014

Amyloid burden and neural function in people at risk for Alzheimer's Disease

Sterling C. Johnson; Bradley T. Christian; Ozioma C. Okonkwo; Jennifer M. Oh; Sandra Harding; Guofan Xu; Ansel T. Hillmer; Dustin Wooten; Dhanabalan Murali; Todd E. Barnhart; Lance Hall; Annie M. Racine; William E. Klunk; Chester A. Mathis; Barbara B. Bendlin; Catherine L. Gallagher; Cynthia M. Carlsson; Howard A. Rowley; Bruce P. Hermann; N. Maritza Dowling; Sanjay Asthana; Mark A. Sager

To determine the relationship between amyloid burden and neural function in healthy adults at risk for Alzheimers Disease (AD), we used multimodal imaging with [C-11]Pittsburgh compound B positron emission tomography, [F-18]fluorodeoxyglucose, positron emission tomography , and magnetic resonance imaging, together with cognitive measurement in 201 subjects (mean age, 60.1 years; range, 46-73 years) from the Wisconsin Registry for Alzheimers Prevention. Using a qualitative rating, 18% of the samples were strongly positive Beta-amyloid (Aβ+), 41% indeterminate (Aβi), and 41% negative (Aβ-). Aβ+ was associated with older age, female sex, and showed trends for maternal family history of AD and APOE4. Relative to the Aβ- group, Aβ+ and Aβi participants had increased glucose metabolism in the bilateral thalamus; Aβ+ participants also had increased metabolism in the bilateral superior temporal gyrus. Aβ+ participants exhibited increased gray matter in the lateral parietal lobe bilaterally relative to the Aβ- group, and no areas of significant atrophy. Cognitive performance and self report cognitive and affective symptoms did not differ between groups. Amyloid burden can be identified in adults at a mean age of 60 years and is accompanied by glucometabolic increases in specific areas, but not atrophy or cognitive loss. This asymptomatic stage may be an opportune window for intervention to prevent progression to symptomatic AD.


NeuroImage: Clinical | 2014

Associations between white matter microstructure and amyloid burden in preclinical Alzheimer's disease: A multimodal imaging investigation.

Annie M. Racine; Nagesh Adluru; Andrew L. Alexander; Bradley T. Christian; Ozioma C. Okonkwo; Jennifer M. Oh; Caitlin A. Cleary; Alex C. Birdsill; Ansel T. Hillmer; Dhanabalan Murali; Todd E. Barnhart; Catherine L. Gallagher; Cynthia M. Carlsson; Howard A. Rowley; N. Maritza Dowling; Sanjay Asthana; Mark A. Sager; Barbara B. Bendlin; Sterling C. Johnson

Some cognitively healthy individuals develop brain amyloid accumulation, suggestive of incipient Alzheimers disease (AD), but the effect of amyloid on other potentially informative imaging modalities, such as Diffusion Tensor Imaging (DTI), in characterizing brain changes in preclinical AD requires further exploration. In this study, a sample (N = 139, mean age 60.6, range 46 to 71) from the Wisconsin Registry for Alzheimers Prevention (WRAP), a cohort enriched for AD risk factors, was recruited for a multimodal imaging investigation that included DTI and [C-11]Pittsburgh Compound B (PiB) positron emission tomography (PET). Participants were grouped as amyloid positive (Aβ+), amyloid indeterminate (Aβi), or amyloid negative (Aβ−) based on the amount and pattern of amyloid deposition. Regional voxel-wise analyses of four DTI metrics, fractional anisotropy (FA), mean diffusivity (MD), axial diffusivity (Da), and radial diffusivity (Dr), were performed based on amyloid grouping. Three regions of interest (ROIs), the cingulum adjacent to the corpus callosum, hippocampal cingulum, and lateral fornix, were selected based on their involvement in the early stages of AD. Voxel-wise analysis revealed higher FA among Aβ+ compared to Aβ− in all three ROIs and in Aβi compared to Aβ− in the cingulum adjacent to the corpus callosum. Follow-up exploratory whole-brain analyses were consistent with the ROI findings, revealing multiple regions where higher FA was associated with greater amyloid. Lower fronto-lateral gray matter MD was associated with higher amyloid burden. Further investigation showed a negative correlation between MD and PiB signal, suggesting that Aβ accumulation impairs diffusion. Interestingly, these findings in a largely presymptomatic sample are in contradistinction to relationships reported in the literature in symptomatic disease stages of Mild Cognitive Impairment and AD, which usually show higher MD and lower FA. Together with analyses showing that cognitive function in these participants is not associated with any of the four DTI metrics, the present results suggest an early relationship between PiB and DTI, which may be a meaningful indicator of the initiating or compensatory mechanisms of AD prior to cognitive decline.


Synapse | 2011

In Vivo Kinetics of (F-18)MEFWAY: A Comparison With (C-11)WAY100635 and (F-18)MPPF in the Nonhuman Primate

Dustin Wooten; J.D. Moraino; Ansel T. Hillmer; Jonathan W. Engle; O.J. DeJesus; Dhanabalan Murali; Todd E. Barnhart; Robert J. Nickles; Richard J. Davidson; Mary L. Schneider; Jogeshwar Mukherjee; Brad T. Christian

[F‐18]Mefway was developed to provide an F‐18 labeled positron emission tomography (PET) neuroligand with high affinity for the serotonin 5‐HT1A receptor to improve the in vivo assessment of the 5‐HT1A system. The goal of this work was to compare the in vivo kinetics of [F‐18]mefway, [F‐18]MPPF, and [C‐11]WAY100635 in the rhesus monkey. Methods: Each of four monkeys were given bolus injections of [F‐18]mefway, [C‐11]WAY100635, and [F‐18]MPPF and scans were acquired with a microPET P4 scanner. Arterial blood was sampled to assay parent compound throughout the time course of the PET experiment. Time activity curves were extracted in the high 5‐HT1A binding areas of the anterior cingulate cortex (ACG), mesial temporal cortex, raphe nuclei, and insula cortex. Time activity curves were also extracted in the cerebellum, which was used as a reference region. The in vivo kinetics of the radiotracers were compared based on the nondisplaceable distribution volume (VND) and binding potential (BPND). Results: At 30 min, the fraction of radioactivity in the plasma due to parent compound was 19%, 28%, and 29% and cleared from the arterial plasma at rates of 0.0031, 0.0078, and 0.0069 (min−1) ([F‐18]mefway, [F‐18]MPPF, [C‐11]WAY100635). The BPND in the brain regions were mesial temporal cortex: 7.4 ± 0.6, 3.1 ± 0.4, 7.0 ± 1.2, ACG: 7.2 ± 1.2, 2.1 ± 0.2, 7.9 ± 1.2; raphe nuclei: 3.7 ± 0.6, 1.3 ± 0.3, 3.3 ± 0.7; and insula cortex: 4.2 ± 0.6, 1.2 ± 0.1, 4.7 ± 1.0 for [F‐18]mefway, [F‐18]MPPF, and [C‐11]WAY100635 respectively. Conclusions: In the rhesus monkey, [F‐18]mefway has similar in vivo kinetics to [C‐11]WAY100635 and yields greater than 2‐fold higher BPND than [F‐18]MPPF. These properties make [F‐18]mefway a promising radiotracer for 5‐HT1A assay, providing higher counting statistics and a greater dynamic range in BPND. Synapse, 2011.


Alzheimers & Dementia | 2016

The effects of normal aging on amyloid-β deposition in nondemented adults with Down syndrome as imaged by carbon 11-labeled Pittsburgh compound B.

Patrick J. Lao; Tobey J. Betthauser; Ansel T. Hillmer; Julie C. Price; William E. Klunk; Iulia Mihaila; Andrew T. Higgins; Peter D. Bulova; Sigan L. Hartley; Regina M. Hardison; Rameshwari V. Tumuluru; Dhanabalan Murali; Chester A. Mathis; Annie D. Cohen; Todd E. Barnhart; Darlynne A. Devenny; Marsha R. Mailick; Sterling C. Johnson; Benjamin L. Handen; Bradley T. Christian

In Down syndrome (DS), the overproduction of amyloid precursor protein is hypothesized to predispose young adults to early expression of Alzheimer‐like neuropathology.


Synapse | 2011

Specific α4β2 Nicotinic Acetylcholine Receptor Binding of [F-18]Nifene in the Rhesus Monkey

Ansel T. Hillmer; Dustin Wooten; Jeffrey Moirano; Maxim Slesarev; Todd E. Barnhart; Jonathan W. Engle; Robert J. Nickles; Dhanabalan Murali; Mary L. Schneider; Jogeshwar Mukherjee; Bradley T. Christian

[F‐18]Nifene is a PET radioligand developed to image α4β2* nicotinic acetylcholine receptors (nAChR) in the brain. This work assesses the in vivo binding and imaging characteristics of [F‐18]nifene in rhesus monkeys for the development of PET experiments examining nAChR binding.


The Journal of Nuclear Medicine | 2012

PET Imaging of α4β2* Nicotinic Acetylcholine Receptors: Quantitative Analysis of 18F-Nifene Kinetics in the Nonhuman Primate

Ansel T. Hillmer; Dustin Wooten; Maxim Slesarev; Elizabeth O. Ahlers; Todd E. Barnhart; Dhanabalan Murali; Mary L. Schneider; Jogeshwar Mukherjee; Bradley T. Christian

The PET radioligand 2-fluoro-3-[2-((S)-3-pyrrolinyl)methoxy]pyridine (18F-nifene) is an α4β2* nicotinic acetylcholine receptor (nAChR) agonist developed to provide accelerated in vivo equilibrium compared with existing α4β2* radioligands. The goal of this work was to analyze the in vivo kinetic properties of 18F-nifene with both kinetic modeling and graphical analysis techniques. Methods: Dynamic PET experiments were performed on 4 rhesus monkeys (female; age range, 9–13 y) using a small-animal PET scanner. Studies began with a high-specific-activity 18F-nifene injection, followed by a coinjection of 18F-nifene and unlabeled nifene at 60 min. Sampling of arterial blood with metabolite analysis was performed throughout the experiment to provide a parent radioligand input function. In vivo kinetics were characterized with both a 1-tissue-compartment model (1TCM) and a 2-tissue-compartment model, Logan graphical methods (both with and without blood sampling), and the multilinear reference tissue model. Total distribution volumes and nondisplaceable binding potentials (BPND) were used to compare regional binding of 18F-nifene. Regions examined include the anteroventral thalamus, lateral geniculate body, frontal cortex, subiculum, and cerebellum. Results: The rapid uptake and binding of 18F-nifene in nAChR-rich regions of the brain was appropriately modeled using the 1TCM. No evidence for specific binding of 18F-nifene in the cerebellum was detected on the basis of the coinjection studies, suggesting the suitability of the cerebellum as a reference region. Total distribution volumes in the cerebellum were 6.91 ± 0.61 mL/cm3. BPND values calculated with the 1TCM were 1.60 ± 0.17, 1.35 ± 0.16, 0.26 ± 0.08, and 0.30 ± 0.07 in the anteroventral thalamus, lateral geniculate body, frontal cortex, and subiculum, respectively. For all brain regions, there was a less than 0.04 absolute difference in the average BPND values calculated with each of the 1TCM, multilinear reference tissue model, and Logan methods. Conclusion: The fast kinetic properties and specific regional binding of 18F-nifene promote extension of the radioligand into preclinical animal models and human subjects.


The Journal of Neuroscience | 2014

Deficient Import of Acetyl-CoA into the ER Lumen Causes Neurodegeneration and Propensity to Infections, Inflammation, and Cancer

Yajing Peng; Mi Li; Ben Clarkson; Mariana Pehar; Patrick J. Lao; Ansel T. Hillmer; Todd E. Barnhart; Bradley T. Christian; Heather A. Mitchell; Barbara B. Bendlin; Matyas Sandor; Luigi Puglielli

The import of acetyl-CoA into the ER lumen by AT-1/SLC33A1 is essential for the Nε-lysine acetylation of ER-resident and ER-transiting proteins. A point-mutation (S113R) in AT-1 has been associated with a familial form of spastic paraplegia. Here, we report that AT-1S113R is unable to form homodimers in the ER membrane and is devoid of acetyl-CoA transport activity. The reduced influx of acetyl-CoA into the ER lumen results in reduced acetylation of ER proteins and an aberrant form of autophagy. Mice homozygous for the mutation display early developmental arrest. In contrast, heterozygous animals develop to full term, but display neurodegeneration and propensity to infections, inflammation, and cancer. The immune and cancer phenotypes are contingent on the presence of pathogens in the colony, whereas the nervous system phenotype is not. In conclusion, our results reveal a previously unknown aspect of acetyl-CoA metabolism that affects the immune and nervous systems and the risk for malignancies.


The Journal of Neuroscience | 2013

Serotonin Transporter Genotype Affects Serotonin 5-HT1A Binding in Primates

Bradley T. Christian; Dustin Wooten; Ansel T. Hillmer; Dana L. Tudorascu; Alexander K. Converse; Colleen F. Moore; Elizabeth O. Ahlers; Todd E. Barnhart; Ned H. Kalin; Christina S. Barr; Mary L. Schneider

Disruption of the serotonin system has been implicated in anxiety and depression and a related genetic variation has been identified that may predispose individuals for these illnesses. The relationship of a functional variation of the serotonin transporter promoter gene (5-HTTLPR) on serotonin transporter binding using in vivo imaging techniques have yielded inconsistent findings when comparing variants for short (s) and long (l) alleles. However, a significant 5-HTTLPR effect on receptor binding at the 5-HT1A receptor site has been reported in humans, suggesting the 5-HTTLPR polymorphism may play a role in serotonin (5-HT) function. Rhesus monkeys possess a 5-HTTLPR length polymorphism similar to humans and serve as an excellent model for studying the effects of this orthologous genetic variation on behaviors and neurochemical functions related to the 5-HT system. In this study, PET imaging of [18F]mefway was performed on 58 rhesus monkeys (33 l/l, 25 s-carriers) to examine the relation between 5-HT1A receptor-specific binding and 5-HTTLPR genotypes. Significantly lower 5-HT1A binding was found in s-carrier subjects throughout both cortical brain regions and the raphe nuclei. These results demonstrate that the underlying 5-HT neurochemical system is influenced by this functional polymorphism and illustrate the strong potential for extending the nonhuman primate model into investigating the role of this genetic variant on behavior and gene–environment interactions.


Nuclear Medicine and Biology | 2011

An In Vivo Comparison of Cis- and Trans- [18F]Mefway in the Nonhuman Primate

Dustin Wooten; Ansel T. Hillmer; Dhanabalan Murali; Todd E. Barnhart; Mary L. Schneider; Jogeshwar Mukherjee; Bradley T. Christian

INTRODUCTION [(18)F]Mefway is a serotonin 5-HT(1A) PET radiotracer with high specificity and favorable in vivo imaging properties. The chemical structure of [(18)F]mefway permits (18)F labeling in either the cis or trans positions at the 4-cyclohexyl site. We have previously reported on the in vivo kinetics of trans-[(18)F]mefway in the nonhuman primate. In this work, we compare the in vivo binding of cis-[(18)F]mefway and trans-[(18)F]mefway to evaluate the properties of cis-[(18)F]mefway for 5-HT(1A) PET imaging. METHODS The cis- and trans-[(18)F]mefway tracers were synthesized via nucleophilic substitution with their respective tosyl precursors. Two monkeys (one male, one female) were given bolus injections of both cis- and trans-labeled [(18)F]mefway in separate experiments. Dynamic scans were acquired for 90 min with a microPET P4 scanner. Time-activity curves were extracted in the areas of the mesial temporal cortex (MTC), anterior cingulate gyrus (aCG), insular cortex (IC), raphe nuclei (RN) and cerebellum (CB). The in vivo behavior of the radiotracers was compared based upon the nondisplaceable binding potential (BP(ND)) using the CB as a reference region. RESULTS Averaged over the two subjects, BP(ND) values were as follows: MTC: 7.7, 0.58; aCG: 4.95, 0.32; IC: 3.27, 0.2; and RN: 3.05, 0.13, for trans-[(18)F]mefway and cis-[(18)F]mefway, respectively. CONCLUSION The cis-labeled [(18)F]mefway tracer has low specific binding throughout the 5-HT(1A) regions of the brain compared to trans-[(18)F]mefway, suggesting that the target-to-background binding of cis-[(18)F]mefway may limit its use for in vivo assessment of 5-HT(1A) binding.


Journal of Cerebral Blood Flow and Metabolism | 2013

Measuring α4β2∗ Nicotinic Acetylcholine Receptor Density in Vivo with [18F]nifene PET in the Nonhuman Primate

Ansel T. Hillmer; Dustin Wooten; Maxim Slesarev; Elizabeth O. Ahlers; Todd E. Barnhart; Mary L. Schneider; Jogeshwar Mukherjee; Bradley T. Christian

[18F]Nifene is an agonist PET radioligand developed to image α4β2∗ nicotinic acetylcholine receptors (nAChRs). This work aims to quantify the receptor density (Bmax) of α4β2∗ nAChRs and the in vivo (apparent) dissociation constant (KDapp) of [18F]nifene. Multiple-injection [18F]nifene experiments with varying cold nifene masses were conducted on four rhesus monkeys with a microPET P4 scanner. Compartment modeling techniques were used to estimate regional Bmax values and a global value of KDapp. The fast kinetic properties of [18F]nifene also permitted alternative estimates of Bmax and KDapp at transient equilibrium with the same experimental data using Scatchard-like methodologies. Averaged across subjects, the compartment modeling analysis yielded Bmax values of 4.8 ± 1.4, 4.3 ±1.0, 1.2 ± 0.4, and 1.2 ± 0.3 pmol/mL in the regions of antereoventral thalamus, lateral geniculate, frontal cortex, and subiculum, respectively. The KDapp of nifene was 2.4 ± 0.3 pmol/mL. The Scatchard analysis based on graphical evaluation of the data after transient equilibrium yielded Bmax estimations comparable to the modeling results with a positive bias of 28%. These findings show the utility of [18F]nifene for measuring α4β2∗ nAChR Bmax in vivo in the rhesus monkey with a single PET experiment.

Collaboration


Dive into the Ansel T. Hillmer's collaboration.

Top Co-Authors

Avatar

Todd E. Barnhart

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Bradley T. Christian

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Mary L. Schneider

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Dhanabalan Murali

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Patrick J. Lao

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Bradley Christian

University of Texas Health Science Center at San Antonio

View shared research outputs
Top Co-Authors

Avatar

Elizabeth O. Ahlers

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Tobey J. Betthauser

University of Wisconsin-Madison

View shared research outputs
Researchain Logo
Decentralizing Knowledge