Network


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

Hotspot


Dive into the research topics where Erica M. Weekman is active.

Publication


Featured researches published by Erica M. Weekman.


Biochimica et Biophysica Acta | 2016

Homocysteine, hyperhomocysteinemia and vascular contributions to cognitive impairment and dementia (VCID)

Atticus H. Hainsworth; Natalie E. Yeo; Erica M. Weekman; Donna M. Wilcock

Homocysteine is produced physiologically in all cells, and is present in plasma of healthy individuals (plasma [HCy]: 3-10μM). While rare genetic mutations (CBS, MTHFR) cause severe hyperhomocysteinemia ([HCy]: 100-200μM), mild-moderate hyperhomocysteinemia ([HCy]: 10-100μM) is common in older people, and is an independent risk factor for stroke and cognitive impairment. As B-vitamin supplementation (B6, B12 and folate) has well-validated homocysteine-lowering efficacy, this may be a readily-modifiable risk factor in vascular contributions to cognitive impairment and dementia (VCID). Here we review the biochemical and cellular actions of HCy related to VCID. Neuronal actions of HCy were at concentrations above the clinically-relevant range. Effects of HCy <100μM were primarily vascular, including myocyte proliferation, vessel wall fibrosis, impaired nitric oxide signalling, superoxide generation and pro-coagulant actions. HCy-lowering clinical trials relevant to VCID are discussed. Extensive clinical and preclinical data support HCy as a mediator for VCID. In our view further trials of combined B-vitamin supplementation are called for, incorporating lessons from previous trials and from recent experimental work. To maximise likelihood of treatment effect, a future trial should: supply a high-dose, combination supplement (B6, B12 and folate); target the at-risk age range; and target cohorts with low baseline B-vitamin status. This article is part of a Special Issue entitled: Vascular Contributions to Cognitive Impairment and Dementia edited by M. Paul Murphy, Roderick A. Corriveau and Donna M. Wilcock.


Journal of Neuroinflammation | 2015

Determining the role of IL-4 induced neuroinflammation in microglial activity and amyloid-β using BV2 microglial cells and APP/PS1 transgenic mice.

Clare H Latta; Tiffany L. Sudduth; Erica M. Weekman; Erin L. Abner; Gabriel Popa; Michael D. Mendenhall; Floracita Gonzalez-Oregon; Kaitlyn Braun; Donna M. Wilcock

BackgroundMicroglia are considered the resident immune cells of the central nervous system (CNS). In response to harmful stimuli, an inflammatory reaction ensues in which microglia are activated in a sequenced spectrum of pro- and antiinflammatory phenotypes that are akin to the well-characterized polarization states of peripheral macrophages. A “classically” activated M1 phenotype is known to eradicate toxicity. The transition to an “alternatively” activated M2 phenotype encompasses neuroprotection and repair. In recent years, inflammation has been considered an accompanying pathology in response to the accumulation of extracellular amyloid-β (Aβ) in Alzheimer’s disease (AD). This study aimed to drive an M2a-biased immune phenotype with IL-4 in vitro and in vivo and to determine the subsequent effects on microglial activation and Aβ pathology.MethodsIn vitro, exogenous IL-4 was applied to BV2 microglial cell cultures to evaluate the temporal progression of microglial responses. In vivo, intracranial injections of an adeno-associate-virus (AAV) viral vector were performed to assess long-term expression of IL-4 in the frontal cortex and hippocampus of Aβ-depositing, APP/PS1 transgenic mice. Quantitative real-time PCR was used to assess the fold change in expression of biomarkers representing each of the microglial phenotypes in both the animal tissue and the BV2 cells. ELISAs quantified IL-4 expression and Aβ levels. Histological staining permitted quantification of microglial and astrocytic activity.ResultsBoth in vitro and in vivo models showed an enhanced M2a phenotype, and the in vivo model revealed a trend toward a decreased trend in Aβ deposition.ConclusionsIn summary, this study offers insight into the therapeutic potential of microglial immune response in AD.


Journal of Alzheimer's Disease | 2015

Matrix Metalloproteinase in Blood-Brain Barrier Breakdown in Dementia

Erica M. Weekman; Donna M. Wilcock

The neurovascular unit, which consists of astrocytic end-feet, neurons, pericytes, and endothelial cells, plays a key role in maintaining brain homeostasis by forming the blood-brain barrier and carefully controlling local cerebral blood flow. When the blood-brain barrier is disrupted, blood components can leak into the brain, damage the surrounding tissue and lead to cognitive impairment. This disruption in the blood-brain barrier and subsequent impairment in cognition are common after stroke and during cerebral amyloid angiopathy and Alzheimers disease. Matrix metalloproteinases are proteases that degrade the extracellular matrix as well as tight junctions between endothelial cells and have been implicated in blood-brain barrier breakdown in neurodegenerative diseases. This review will focus on the roles of MMP2 and MMP9 in dementia, primarily post-stroke events that lead to dementia, cerebral amyloid angiopathy, and Alzheimers disease.


The Journal of Neuroscience | 2016

Reduced Efficacy of Anti-Aβ Immunotherapy in a Mouse Model of Amyloid Deposition and Vascular Cognitive Impairment Comorbidity

Erica M. Weekman; Tiffany L. Sudduth; Carly N. Caverly; Timothy J. Kopper; Oliver W. Phillips; David K. Powell; Donna M. Wilcock

Vascular cognitive impairment and dementia (VCID) is the second most common form of dementia behind Alzheimers disease (AD). It is estimated that 40% of AD patients also have some form of VCID. One promising therapeutic for AD is anti-Aβ immunotherapy, which uses antibodies against Aβ to clear it from the brain. While successful in clearing Aβ and improving cognition in mice, anti-Aβ immunotherapy failed to reach primary cognitive outcomes in several different clinical trials. We hypothesized that one potential reason the anti-Aβ immunotherapy clinical trials were unsuccessful was due to this high percentage of VCID comorbidity in the AD population. We used our unique model of VCID-amyloid comorbidity to test this hypothesis. We placed 9-month-old wild-type and APP/PS1 mice on either a control diet or a diet that induces hyperhomocysteinemia (HHcy). After being placed on the diet for 3 months, the mice then received intraperotineal injections of either IgG2a control or 3D6 for another 3 months. While we found that treatment of our comorbidity model with 3D6 resulted in decreased total Aβ levels, there was no cognitive benefit of the anti-Aβ immunotherapy in our AD/VCID mice. Further, microhemorrhages were increased by 3D6 in the APP/PS1/control but further increased in an additive fashion when 3D6 was administered to the APP/PS1/HHcy mice. This suggests that the use of anti-Aβ immunotherapy in patients with both AD and VCID would be ineffective on cognitive outcomes. SIGNIFICANCE STATEMENT Despite significant mouse model data demonstrating both pathological and cognitive efficacy of anti-Aβ immunotherapy for the treatment of Alzheimers disease, clinical trial outcomes have been underwhelming, failing to meet any primary endpoints. We show here that vascular cognitive impairment and dementia (VCID) comorbidity eliminates cognitive efficacy of anti-Aβ immunotherapy, despite amyloid clearance. Further, cerebrovascular adverse events of the anti-Aβ immunotherapy are significantly exacerbated by the VCID comorbidity. These data suggest that VCID comorbidity with Alzheimers disease may mute the response to anti-Aβ immunotherapy.


Neuroscience | 2017

Time-course of glial changes in the hyperhomocysteinemia model of vascular cognitive impairment and dementia (VCID)

Tiffany L. Sudduth; Erica M. Weekman; Brittani R. Price; Jennifer L. Gooch; Abigail Woolums; Christopher M. Norris; Donna M. Wilcock

Vascular cognitive impairment and dementia (VCID) is the second leading cause of dementia behind Alzheimers disease (AD) and is a frequent co-morbidity with AD. Despite its prevalence, little is known about the molecular mechanisms underlying the cognitive dysfunction resulting from cerebrovascular disease. Astrocytic end-feet almost completely surround intraparenchymal blood vessels in the brain and express a variety of channels and markers indicative of their specialized functions in the maintenance of ionic and osmotic homeostasis and gliovascular signaling. These functions are mediated by end-foot enrichment of the aquaporin 4 water channel (AQP4), the inward rectifying potassium channel Kir4.1 and the calcium-dependent potassium channel MaxiK. Using our hyperhomocysteinemia (HHcy) model of VCID we examined the time-course of astrocytic end-foot changes along with cognitive and neuroinflammatory outcomes. We found that there were significant astrocytic end-foot disruptions in the HHcy model. AQP4 becomes dislocalized from the end-feet, there is a loss of Kir4.1 and MaxiK protein expression, as well as a loss of the Dp71 protein known to anchor the Kir4.1, MaxiK and AQP4 channels to the end-foot membrane. Neuroinflammation occurs prior to the astrocytic changes, while cognitive impairment continues to decline with the exacerbation of the astrocytic changes. We have previously reported similar astrocytic changes in models of cerebral amyloid angiopathy (CAA) and therefore, we believe astrocytic end-foot disruption could represent a common cellular mechanism of VCID and may be a target for therapeutic development.


Asn Neuro | 2017

Hyperhomocysteinemia-Induced Gene Expression Changes in the Cell Types of the Brain:

Erica M. Weekman; Abigail Woolums; Tiffany L. Sudduth; Donna M. Wilcock

High plasma levels of homocysteine, termed hyperhomocysteinemia, are a risk factor for vascular cognitive impairment and dementia, which is the second leading cause of dementia. While hyperhomocysteinemia induces microhemorrhages and cognitive decline in mice, the specific effect of hyperhomocysteinemia on each cell type remains unknown. We took separate cultures of astrocytes, microglia, endothelial cells, and neuronal cells and treated each with moderate levels of homocysteine for 24, 48, 72, and 96 hr. We then determined the gene expression changes for cell-specific markers and neuroinflammatory markers including the matrix metalloproteinase 9 system. Astrocytes had decreased levels of several astrocytic end feet genes, such as aquaporin 4 and an adenosine triphosphate (ATP)-sensitive inward rectifier potassium channel at 72 hr, as well as an increase in matrix metalloproteinase 9 at 48 hr. Gene changes in microglia indicated a peak in proinflammatory markers at 48 hr followed by a peak in the anti-inflammatory marker, interleukin 1 receptor antagonist, at 72 hr. Endothelial cells had reduced occludin expression at 72 hr, while kinases and phosphatases known to alter tau phosphorylation states were increased in neuronal cells. This suggests that hyperhomocysteinemia induces early proinflammatory changes in microglia and astrocytic changes relevant to their interaction with the vasculature. Overall, the data show how hyperhomocysteinemia could impact Alzheimer’s disease and vascular cognitive impairment and dementia.


Alzheimers & Dementia | 2015

Reduced efficacy of anti-Aβ immunotherapy in a mouse model of amyloid deposition and vascular cognitive impairment co-morbidity

Erica M. Weekman; Timothy J. Kopper; Tiffany L. Sudduth; Donna M. Wilcock

mice. cSNK vaccination was assessed in AD mouse models treated at 3 months of age with monthly vaccination and then evaluated at 13 months of age. Results:A monoclonal antibody raised against this novel epitope (5E3) binds to synthetic Ab oligomers (AbO), but is unreactive to monomeric and fibrillar Ab, as well as to amyloid plaques. 5E3 is effective at blocking AbO behavioural toxicity in vivo in wild type mice. Moreover acute treatment with 5E3 reduced AbO concentration in CSF and brains of aged APP/PS1 and Tg2576 mice, with no apparent impact on pre-existing plaques. Furthermore, cSNK-vaccinated mice produced an AbO specific immune response, without cross-reactivity to Ab monomers. Conclusions: These experiments demonstrate three important findings: 1) murine 5E3 can neutralize the toxicity of synthetic AbOs in the molecular weight range of AbOs previously implicated in synaptic dysfunction; 2) systemically administered 5E3 demonstrates target engagement for AbOs in two mouse models of Alzheimer disease in vivo; and 3) active vaccination with the cSNK epitope generates AbO reactivity in mice without recognition of other Ab molecular species. Together, our data demonstrates that cSNK is a well-defined AbO-specific epitope with therapeutic potential.


Alzheimer's Research & Therapy | 2014

β-amyloid deposition is shifted to the vasculature and memory impairment is exacerbated when hyperhomocysteinemia is induced in APP/PS1 transgenic mice

Tiffany L. Sudduth; Erica M. Weekman; Kaitlyn Braun; Donna M. Wilcock


Journal of Neuroinflammation | 2014

Transition from an M1 to a mixed neuroinflammatory phenotype increases amyloid deposition in APP/PS1 transgenic mice

Erica M. Weekman; Tiffany L. Sudduth; Erin L. Abner; Gabriel Popa; Michael D. Mendenhall; Kaitlyn Braun; Abigail Greenstein; Donna M. Wilcock


Alzheimers & Dementia | 2017

ASTROCYTE RESPONSE TO HYPERHOMOCYSTEINEMIA (HHCY)

Brittani R. Price; Tiffany L. Sudduth; Erica M. Weekman; Donna M. Wilcock; Abigail Woolums

Collaboration


Dive into the Erica M. Weekman's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge