Network


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

Hotspot


Dive into the research topics where Shane Bemiller is active.

Publication


Featured researches published by Shane Bemiller.


Journal of Experimental Medicine | 2015

TREM2 deficiency eliminates TREM2+ inflammatory macrophages and ameliorates pathology in Alzheimer’s disease mouse models

Taylor R. Jay; Crystal M. Miller; Paul J. Cheng; Leah C. Graham; Shane Bemiller; Margaret L. Broihier; Guixiang Xu; Daniel Margevicius; J. Colleen Karlo; Gregory L. Sousa; Anne C. Cotleur; Oleg Butovsky; Lynn M. Bekris; Susan M. Staugaitis; James B. Leverenz; Sanjay W. Pimplikar; Gary E. Landreth; Gareth R. Howell; Richard M. Ransohoff; Bruce T. Lamb

Jay and colleagues show that TREM2 deficiency reduces the number of macrophages infiltrating the brain and is protective against disease pathogenesis in mouse models of Alzheimer’s disease.


Molecular Neurodegeneration | 2017

TREM2 deficiency exacerbates tau pathology through dysregulated kinase signaling in a mouse model of tauopathy

Shane Bemiller; Tyler J. McCray; Kevin C. Allan; Shane V. Formica; Guixiang Xu; Gina Wilson; Olga N. Kokiko-Cochran; Samuel D. Crish; Cristian A. Lasagna-Reeves; Richard M. Ransohoff; Gary E. Landreth; Bruce T. Lamb

BackgroundGenetic variants of the Triggering Receptor Expressed on Myeloid Cells-2 (TREM2) confer increased risk of developing late-onset Alzheimer’s Disease (LOAD) and other neurodegenerative disorders. Recent studies provided insight into the multifaceted roles of TREM2 in regulating extracellular β-amyloid (Aβ) pathology, myeloid cell accumulation, and inflammation observed in AD, yet little is known regarding the role of TREM2 in regulating intracellular microtubule associated protein tau (MAPT; tau) pathology in neurodegenerative diseases and in AD, in particular.ResultsHere we report that TREM2 deficiency leads to accelerated and exacerbated hyperphosphorylation and aggregation of tau in a humanized mouse model of tauopathy. TREM2 deficiency also results, indirectly, in dramatic widespread dysregulation of neuronal stress kinase pathways.ConclusionsOur results suggest that deficiency of microglial TREM2 leads to heightened tau pathology coupled with widespread increases in activated neuronal stress kinases. These findings offer new insight into the complex, multiple roles of TREM2 in regulating Aβ and tau pathologies.


Journal of Neurotrauma | 2016

Altered Neuroinflammation and Behavior after Traumatic Brain Injury in a Mouse Model of Alzheimer's Disease

Olga N. Kokiko-Cochran; Lena Ransohoff; Mike Veenstra; Sungho Lee; Maha Saber; Matt Sikora; Ryan Teknipp; Guixiang Xu; Shane Bemiller; Gina Wilson; Samuel D. Crish; Kiran Bhaskar; Yu Shang Lee; Richard M. Ransohoff; Bruce T. Lamb

Traumatic brain injury (TBI) has acute and chronic sequelae, including an increased risk for the development of Alzheimers disease (AD). TBI-associated neuroinflammation is characterized by activation of brain-resident microglia and infiltration of monocytes; however, recent studies have implicated beta-amyloid as a major manipulator of the inflammatory response. To examine neuroinflammation after TBI and development of AD-like features, these studies examined the effects of TBI in the presence and absence of beta-amyloid. The R1.40 mouse model of cerebral amyloidosis was used, with a focus on time points well before robust AD pathologies. Unexpectedly, in R1.40 mice, the acute neuroinflammatory response to TBI was strikingly muted, with reduced numbers of CNS myeloid cells acquiring a macrophage phenotype and decreased expression of inflammatory cytokines. At chronic time points, macrophage activation substantially declined in non-Tg TBI mice; however, it was relatively unchanged in R1.40 TBI mice. The persistent inflammatory response coincided with significant tissue loss between 3 and 120 days post-injury in R1.40 TBI mice, which was not observed in non-Tg TBI mice. Surprisingly, inflammatory cytokine expression was enhanced in R1.40 mice compared with non-Tg mice, regardless of injury group. Although R1.40 TBI mice demonstrated task-specific deficits in cognition, overall functional recovery was similar to non-Tg TBI mice. These findings suggest that accumulating beta-amyloid leads to an altered post-injury macrophage response at acute and chronic time points. Together, these studies emphasize the role of post-injury neuroinflammation in regulating long-term sequelae after TBI and also support recent studies implicating beta-amyloid as an immunomodulator.


Journal of Neurotrauma | 2018

Traumatic Brain Injury in hTau Model Mice: Enhanced Acute Macrophage Response and Altered Long-Term Recovery

Olga N. Kokiko-Cochran; Maha Saber; Shweta S. Puntambekar; Shane Bemiller; Atsuko Katsumoto; Yu Shang Lee; Kiran Bhaskar; Richard M. Ransohoff; Bruce T. Lamb

Traumatic brain injury (TBI) induces widespread neuroinflammation and accumulation of microtubule associated protein tau (MAPT): two key pathological features of tauopathies. This study sought to characterize the microglial/macrophage response to TBI in genomic-based MAPT transgenic mice in a Mapt knockout background (called hTau). Two-month-old hTau and age-matched control male and female mice received a single lateral fluid percussion TBI or sham injury. Separate groups of mice were aged to an acute (3 days post-injury [DPI]) or chronic (135 DPI) post-injury time point. As judged by tissue immunostaining for macrophage markers, microglial/macrophage response to TBI was enhanced at 3 DPI in hTau mice compared with control TBI and sham mice. However, MAPT phosphorylation increased in hTau mice regardless of injury group. Flow cytometric analysis revealed distinct populations of microglia and macrophages within all groups at 135 DPI. Unexpectedly, microglial reactivity was significantly reduced in hTau TBI mice compared with all other groups. Instead, hTau TBI mice showed a persistent macrophage response. In addition, TBI enhanced MAPT pathology in the temporal cortex and hippocampus of hTau TBI mice compared with controls 135 DPI. A battery of behavioral tests revealed that TBI in hTau mice resulted in compromised use of spatial search strategies to complete a water maze task, despite lack of motor or visual deficits. Collectively, these data indicate that the presence of wild-type human tau alters the microglial/macrophage response to a single TBI, induces delayed, region-specific MAPT pathology, and alters cognitive recovery; however, the causal relationship between these events remains unclear. These results highlight the potential significance of communication between MAPT and microglia/macrophages following TBI, and emphasize the role of neuroinflammation in post-injury recovery.


Nature Methods | 2018

Active PSF shaping and adaptive optics enable volumetric localization microscopy through brain sections

Michael J. Mlodzianoski; Paul J. Cheng-Hathaway; Shane Bemiller; Tyler J. McCray; Sheng Liu; David A. Miller; Bruce T. Lamb; Gary E. Landreth; Fang Huang

Application of single-molecule switching nanoscopy (SMSN) beyond the coverslip surface poses substantial challenges due to sample-induced aberrations that distort and blur single-molecule emission patterns. We combined active shaping of point spread functions and efficient adaptive optics to enable robust 3D-SMSN imaging within tissues. This development allowed us to image through 30-μm-thick brain sections to visualize and reconstruct the morphology and the nanoscale details of amyloid-β filaments in a mouse model of Alzheimer’s disease.Active PSF shaping and adaptive optics are combined to enable 3D localization microscopy throughout thick tissues. The method was used to study the nanoscale architecture of amyloid fibrils in a mouse model of Alzheimer’s disease.


Journal of Neuroinflammation | 2018

Genetically enhancing the expression of chemokine domain of CX 3 CL1 fails to prevent tau pathology in mouse models of tauopathy

Shane Bemiller; Nicole Maphis; Shane V. Formica; Gina N. Wilson; Crystal M. Miller; Guixiang Xu; Olga N. Kokiko-Cochran; Ki-Wook Kim; Steffen Jung; Samuel D. Crish; Astrid E. Cardona; Bruce T. Lamb; Kiran Bhaskar

BackgroundFractalkine (CX3CL1) and its receptor (CX3CR1) play an important role in regulating microglial function. We have previously shown that Cx3cr1 deficiency exacerbated tau pathology and led to cognitive impairment. However, it is still unclear if the chemokine domain of the ligand CX3CL1 is essential in regulating neuronal tau pathology.MethodsWe used transgenic mice lacking endogenous Cx3cl1 (Cx3cl1−/−) and expressing only obligatory soluble form (with only chemokine domain) and lacking the mucin stalk of CX3CL1 (referred to as Cx3cl1105Δ mice) to assess tau pathology and behavioral function in both lipopolysaccharide (LPS) and genetic (hTau) mouse models of tauopathy.ResultsFirst, increased basal tau levels accompanied microglial activation in Cx3cl1105Δ mice compared to control groups. Second, increased CD45+ and F4/80+ neuroinflammation and tau phosphorylation were observed in LPS, hTau/Cx3cl1−/−, and hTau/Cx3cl1105Δ mouse models of tau pathology, which correlated with impaired spatial learning. Finally, microglial cell surface expression of CX3CR1 was reduced in Cx3cl1105Δ mice, suggesting enhanced fractalkine receptor internalization (mimicking Cx3cr1 deletion), which likely contributes to the elevated tau pathology.ConclusionsCollectively, our data suggest that overexpression of only chemokine domain of CX3CL1 does not protect against tau pathology.


Alzheimer's & Dementia: Translational Research & Clinical Interventions | 2018

Inflammation as a central mechanism in Alzheimer's disease

Jefferson W. Kinney; Shane Bemiller; Andrew S. Murtishaw; Amanda M. Leisgang; Bruce T. Lamb

Alzheimers disease (AD) is a progressive neurodegenerative disorder that is characterized by cognitive decline and the presence of two core pathologies, amyloid β plaques and neurofibrillary tangles. Over the last decade, the presence of a sustained immune response in the brain has emerged as a third core pathology in AD. The sustained activation of the brains resident macrophages (microglia) and other immune cells has been demonstrated to exacerbate both amyloid and tau pathology and may serve as a link in the pathogenesis of the disorder. In the following review, we provide an overview of inflammation in AD and a detailed coverage of a number of microglia‐related signaling mechanisms that have been implicated in AD. Additional information on microglia signaling and a number of cytokines in AD are also reviewed. We also review the potential connection of risk factors for AD and how they may be related to inflammatory mechanisms.


Alzheimers & Dementia | 2016

THE R47H TREM2 VARIANT MODIFIES ALZHEIMER'S DISEASE PATHOLOGY AND NEUROINFLAMMATION IN A NOVEL KNOCK-IN MOUSE MODEL

Paul J. Cheng-Hathaway; Taylor R. Jay; Erin Reed; Shane Bemiller; Shweta S. Puntambekar; Guixiang Xu; Colleen Carlo; Richard M. Ransohoff; Gary E. Landreth; Bruce T. Lamb

Background:Microglia and associated neuroinflammation play a significant role in Alzheimer’s disease (AD). The voltage-gated potassium channel Kv1.3 (KCNA3) fine-tunes microglia activation by modulating Ca signaling. Because selective pharmacological targeting of Kv1.3 inmicroglia in the brain is feasible and safe, we tested the effects of a selective Kv1.3 blocker PAP-1 in models of AD.Methods: Human AD brains drawn from the brain repositories of the University of California Davis and the Emory University were used for immunohistochemical analyses ofKv1.3. 5xFADmice,which harbor three amyloid-b precursor protein (APP) and two presenilin-1 (PS1) mutations resulting in AD-like Ab amyloidosis and neuroinflammation, were used for behavioral, electrophysiological, biochemical and neuropathological analyses. Results: Kv1.3 immunoreactivities were localized exclusively to microglia in both human and mouse brains and were increased in human AD subjects and 5xFAD mice. Microglial Kv1.3 activity as measured by whole-cell patch-clamp was increased in young and middle-aged 5xFAD mice, but decreased in old (>13 months) 5xFAD mice compared to ageand gender-matched wildtype controls. Ab oligomers (AbO) were able to enhance microlgial Kv1.3 expression. PAP-1 treatment reduced AbO-induced microglial neurotoxicity in vitro and in vivo. A three-month oral regimen of PAP1 improved the hippocampus-dependent memory performance and rectified the hippocampal long-term potentiation of 5xFADmice.Conclusions:Microglial Kv1.3 is a potential therapeutic target and oral dosing of PAP-1 is a promising anti-inflammatory approach for AD.


Molecular Neurodegeneration | 2018

The Trem2 R47H variant confers loss-of-function-like phenotypes in Alzheimer's disease

Paul J. Cheng-Hathaway; Erin G. Reed-Geaghan; Taylor R. Jay; Brad T. Casali; Shane Bemiller; Shweta S. Puntambekar; Victoria E. von Saucken; Roxanne Williams; J. Colleen Karlo; Miguel Moutinho; Guixiang Xu; Richard M. Ransohoff; Bruce T. Lamb; Gary E. Landreth


Alzheimers & Dementia | 2014

THE ROLE OF TREM2 EXPRESSION ON MYELOID CELLS IN ALZHEIMER'S DISEASE

Bruce T. Lamb; Crystal M. Miller; Taylor R. Jay; Leah C. Graham; Shane Bemiller; Guixiang Xu; Daniel Margevicius; Colleen Karlo; Gregory L. Sousa; Bunny Cotleur; Lynn M. Bekris; Susan M. Staugaitis; James B. Leverenz; Sanjay W. Pimplikar; Gary E. Landreth; Gareth R. Howell; Richard M. Ransohoff

Collaboration


Dive into the Shane Bemiller's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Gary E. Landreth

Case Western Reserve University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Kiran Bhaskar

University of New Mexico

View shared research outputs
Top Co-Authors

Avatar

Taylor R. Jay

Case Western Reserve University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Samuel D. Crish

Northeast Ohio Medical University

View shared research outputs
Researchain Logo
Decentralizing Knowledge