Network


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

Hotspot


Dive into the research topics where Dhananjay Namjoshi is active.

Publication


Featured researches published by Dhananjay Namjoshi.


Journal of Biological Chemistry | 2010

ATP-binding Cassette Transporter A1 Mediates the Beneficial Effects of the Liver X Receptor Agonist GW3965 on Object Recognition Memory and Amyloid Burden in Amyloid Precursor Protein/Presenilin 1 Mice

James Donkin; Sophie Stukas; Veronica Hirsch-Reinshagen; Dhananjay Namjoshi; Anna Wilkinson; Sharon May; Jeniffer Chan; Jianjia Fan; Jon L. Collins; Cheryl L. Wellington

The cholesterol transpoter ATP-binding cassette transporter A1 (ABCA1) moves lipids onto apolipoproteins including apolipoprotein E (apoE), which is the major cholesterol carrier in the brain and an established genetic risk factor for late-onset Alzheimer disease (AD). In amyloid mouse models of AD, ABCA1 deficiency exacerbates amyloidogenesis, whereas ABCA1 overexpression ameliorates amyloid load, suggesting a role for ABCA1 in Aβ metabolism. Agonists of liver X receptors (LXR), including GW3965, induce transcription of several genes including ABCA1 and apoE, and reduce Aβ levels and improve cognition in AD mice. However, the specific role of ABCA1 in mediating beneficial responses to LXR agonists in AD mice is unknown. We evaluated behavioral and neuropathogical outcomes in GW3965-treated female APP/PS1 mice with and without ABCA1. Treatment of APP/PS1 mice with GW3965 increased ABCA1 and apoE protein levels. ABCA1 was required to observe significantly elevated apoE levels in brain tissue and cerebrospinal fluid upon therapeutic (33 mg/kg/day) GW3965 treatment. At 33 mg/kg/day, GW3965 was also associated with a trend toward redistribution of Aβ to the carbonate-soluble pool independent of ABCA1. APP/PS1 mice treated with either 2.5 or 33 mg/kg/day of GW3965 showed a clear trend toward reduced amyloid burden in hippocampus and whole brain, whereas APP/PS1-treated mice lacking ABCA1 failed to display reduced amyloid load in the whole brain and showed trends toward increased hippocampal amyloid. Treatment of APP/PS1 mice with either dose of GW3965 completely restored novel object recognition memory to wild-type levels, which required ABCA1. These results suggest that ABCA1 contributes to several beneficial effects of the LXR agonist GW3965 in APP/PS1 mice.


Disease Models & Mechanisms | 2013

Towards clinical management of traumatic brain injury: a review of models and mechanisms from a biomechanical perspective

Dhananjay Namjoshi; Craig A. Good; Wai Hang Cheng; William J. Panenka; Darrin Richards; Peter A. Cripton; Cheryl L. Wellington

Traumatic brain injury (TBI) is a major worldwide healthcare problem. Despite promising outcomes from many preclinical studies, the failure of several clinical studies to identify effective therapeutic and pharmacological approaches for TBI suggests that methods to improve the translational potential of preclinical studies are highly desirable. Rodent models of TBI are increasingly in demand for preclinical research, particularly for closed head injury (CHI), which mimics the most common type of TBI observed clinically. Although seemingly simple to establish, CHI models are particularly prone to experimental variability. Promisingly, bioengineering-oriented research has advanced our understanding of the nature of the mechanical forces and resulting head and brain motion during TBI. However, many neuroscience-oriented laboratories lack guidance with respect to fundamental biomechanical principles of TBI. Here, we review key historical and current literature that is relevant to the investigation of TBI from clinical, physiological and biomechanical perspectives, and comment on how the current challenges associated with rodent TBI models, particularly those involving CHI, could be improved.


Molecular Neurodegeneration | 2014

Merging pathology with biomechanics using CHIMERA (Closed-Head Impact Model of Engineered Rotational Acceleration): a novel, surgery-free model of traumatic brain injury

Dhananjay Namjoshi; Wai Hang Cheng; Kurt A. McInnes; Kris M. Martens; Michael Carr; Anna Wilkinson; Jianjia Fan; Jérôme Robert; Arooj Hayat; Peter A. Cripton; Cheryl L. Wellington

BackgroundTraumatic brain injury (TBI) is a major health care concern that currently lacks any effective treatment. Despite promising outcomes from many preclinical studies, clinical evaluations have failed to identify effective pharmacological therapies, suggesting that the translational potential of preclinical models may require improvement. Rodents continue to be the most widely used species for preclinical TBI research. As most human TBIs result from impact to an intact skull, closed head injury (CHI) models are highly relevant, however, traditional CHI models suffer from extensive experimental variability that may be due to poor control over biomechanical inputs. Here we describe a novel CHI model called CHIMERA (Closed-Head Impact Model of Engineered Rotational Acceleration) that fully integrates biomechanical, behavioral, and neuropathological analyses. CHIMERA is distinct from existing neurotrauma model systems in that it uses a completely non-surgical procedure to precisely deliver impacts of prescribed dynamic characteristics to a closed skull while enabling kinematic analysis of unconstrained head movement. In this study, we characterized head kinematics as well as functional, neuropathological, and biochemical outcomes up to 14d following repeated TBI (rTBI) in adult C57BL/6 mice using CHIMERA.ResultsHead kinematic analysis showed excellent repeatability over two closed head impacts separated at 24h. Injured mice showed significantly prolonged loss of righting reflex and displayed neurological, motor, and cognitive deficits along with anxiety-like behavior. Repeated TBI led to diffuse axonal injury with extensive microgliosis in white matter from 2-14d post-rTBI. Injured mouse brains also showed significantly increased levels of TNF-α and IL-1β and increased endogenous tau phosphorylation.ConclusionsRepeated TBI using CHIMERA mimics many of the functional and pathological characteristics of human TBI with a reliable biomechanical response of the head. This makes CHIMERA well suited to investigate the pathophysiology of TBI and for drug development programs.


PLOS ONE | 2013

The Liver X Receptor Agonist GW3965 Improves Recovery from Mild Repetitive Traumatic Brain Injury in Mice Partly through Apolipoprotein E

Dhananjay Namjoshi; Georgina Martin; James Donkin; Anna Wilkinson; Sophie Stukas; Jianjia Fan; Michael Carr; Sepideh Tabarestani; Kelli Wuerth; Robert E. W. Hancock; Cheryl L. Wellington

Traumatic brain injury (TBI) increases Alzheimer’s disease (AD) risk and leads to the deposition of neurofibrillary tangles and amyloid deposits similar to those found in AD. Agonists of Liver X receptors (LXRs), which regulate the expression of many genes involved in lipid homeostasis and inflammation, improve cognition and reduce neuropathology in AD mice. One pathway by which LXR agonists exert their beneficial effects is through ATP-binding cassette transporter A1 (ABCA1)-mediated lipid transport onto apolipoprotein E (apoE). To test the therapeutic utility of this pathway for TBI, we subjected male wild-type (WT) and apoE−/− mice to mild repetitive traumatic brain injury (mrTBI) followed by treatment with vehicle or the LXR agonist GW3965 at 15 mg/kg/day. GW3965 treatment restored impaired novel object recognition memory in WT but not apoE−/− mice. GW3965 did not significantly enhance the spontaneous recovery of motor deficits observed in all groups. Total soluble Aβ40 and Aβ42 levels were significantly elevated in WT and apoE−/− mice after injury, a response that was suppressed by GW3965 in both genotypes. WT mice showed mild but significant axonal damage at 2 d post-mrTBI, which was suppressed by GW3965. In contrast, apoE−/− mice showed severe axonal damage from 2 to 14 d after mrTBI that was unresponsive to GW3965. Because our mrTBI model does not produce significant inflammation, the beneficial effects of GW3965 we observed are unlikely to be related to reduced inflammation. Rather, our results suggest that both apoE-dependent and apoE-independent pathways contribute to the ability of GW3965 to promote recovery from mrTBI.


Journal of the American Heart Association | 2014

Intravenously Injected Human Apolipoprotein A‐I Rapidly Enters the Central Nervous System via the Choroid Plexus

Sophie Stukas; Jérôme Robert; Michael Lee; Iva Kulic; Michael Carr; Katherine Tourigny; Jianjia Fan; Dhananjay Namjoshi; Kalistyne Lemke; Nicole DeValle; Jeniffer Chan; Tammy Wilson; Anna Wilkinson; Rafi Chapanian; Jayachandran N. Kizhakkedathu; John R. Cirrito; Michael N. Oda; Cheryl L. Wellington

Background Brain lipoprotein metabolism is dependent on lipoprotein particles that resemble plasma high‐density lipoproteins but that contain apolipoprotein (apo) E rather than apoA‐I as their primary protein component. Astrocytes and microglia secrete apoE but not apoA‐I; however, apoA‐I is detectable in both cerebrospinal fluid and brain tissue lysates. The route by which plasma apoA‐I enters the central nervous system is unknown. Methods and Results Steady‐state levels of murine apoA‐I in cerebrospinal fluid and interstitial fluid are 0.664 and 0.120 μg/mL, respectively, whereas brain tissue apoA‐I is ≈10% to 15% of its levels in liver. Recombinant, fluorescently tagged human apoA‐I injected intravenously into mice localizes to the choroid plexus within 30 minutes and accumulates in a saturable, dose‐dependent manner in the brain. Recombinant, fluorescently tagged human apoA‐I accumulates in the brain for 2 hours, after which it is eliminated with a half‐life of 10.3 hours. In vitro, human apoA‐I is specifically bound, internalized, and transported across confluent monolayers of primary human choroid plexus epithelial cells and brain microvascular endothelial cells. Conclusions Following intravenous injection, recombinant human apoA‐I rapidly localizes predominantly to the choroid plexus. Because apoA‐I mRNA is undetectable in murine brain, our results suggest that plasma apoA‐I, which is secreted from the liver and intestine, gains access to the central nervous system primarily by crossing the blood–cerebrospinal fluid barrier via specific cellular mediated transport, although transport across the blood–brain barrier may also contribute to a lesser extent.


PLOS ONE | 2016

Chronic Exposure to Androgenic-Anabolic Steroids Exacerbates Axonal Injury and Microgliosis in the CHIMERA Mouse Model of Repetitive Concussion.

Dhananjay Namjoshi; Wai Hang Cheng; Michael Carr; Kris M. Martens; Shahab Zareyan; Anna Wilkinson; Kurt A. McInnes; Peter A. Cripton; Cheryl L. Wellington

Concussion is a serious health concern. Concussion in athletes is of particular interest with respect to the relationship of concussion exposure to risk of chronic traumatic encephalopathy (CTE), a neurodegenerative condition associated with altered cognitive and psychiatric functions and profound tauopathy. However, much remains to be learned about factors other than cumulative exposure that could influence concussion pathogenesis. Approximately 20% of CTE cases report a history of substance use including androgenic-anabolic steroids (AAS). How acute, chronic, or historical AAS use may affect the vulnerability of the brain to concussion is unknown. We therefore tested whether antecedent AAS exposure in young, male C57Bl/6 mice affects acute behavioral and neuropathological responses to mild traumatic brain injury (TBI) induced with the CHIMERA (Closed Head Impact Model of Engineered Rotational Acceleration) platform. Male C57Bl/6 mice received either vehicle or a cocktail of three AAS (testosterone, nandrolone and 17α-methyltestosterone) from 8–16 weeks of age. At the end of the 7th week of treatment, mice underwent two closed-head TBI or sham procedures spaced 24 h apart using CHIMERA. Post-repetitive TBI (rTBI) behavior was assessed for 7 d followed by tissue collection. AAS treatment induced the expected physiological changes including increased body weight, testicular atrophy, aggression and downregulation of brain 5-HT1B receptor expression. rTBI induced behavioral deficits, widespread axonal injury and white matter microgliosis. While AAS treatment did not worsen post-rTBI behavioral changes, AAS-treated mice exhibited significantly exacerbated axonal injury and microgliosis, indicating that AAS exposure can alter neuronal and innate immune responses to concussive TBI.


Experimental Neurology | 2017

Defining the biomechanical and biological threshold of murine mild traumatic brain injury using CHIMERA (Closed Head Impact Model of Engineered Rotational Acceleration)

Dhananjay Namjoshi; Wai Hang Cheng; Asma Bashir; Anna Wilkinson; Sophie Stukas; Kris M. Martens; Tom Whyte; Zelalem A. Abebe; Kurt A. McInnes; Peter A. Cripton; Cheryl L. Wellington

ABSTRACT CHIMERA (Closed Head Impact Model of Engineered Rotational Acceleration) is a recently described animal model of traumatic brain injury (TBI) that primarily produces diffuse axonal injury (DAI) characterized by white matter inflammation and axonal damage. CHIMERA was specifically designed to reliably generate a variety of TBI severities using precise and quantifiable biomechanical inputs in a nonsurgical user‐friendly platform. The objective of this study was to define the lower limit of single impact mild TBI (mTBI) using CHIMERA by characterizing the dose‐response relationship between biomechanical input and neurological, behavioral, neuropathological and biochemical outcomes. Wild‐type male mice were subjected to a single CHIMERA TBI using six impact energies ranging from 0.1 to 0.7 J, and post‐TBI outcomes were assessed over an acute period of 14 days. Here we report that single TBI using CHIMERA induces injury dose‐ and time‐dependent changes in behavioral and neurological deficits, axonal damage, white matter tract microgliosis and astrogliosis. Impact energies of 0.4 J or below produced no significant phenotype (subthreshold), 0.5 J led to significant changes for one or more phenotypes (threshold), and 0.6 and 0.7 J resulted in significant changes in all outcomes assessed (mTBI). We further show that linear head kinematics are the most robust predictors of duration of unconsciousness, severity of neurological deficits, white matter injury, and microgliosis following single TBI. Our data extend the validation of CHIMERA as a biofidelic animal model of DAI and establish working parameters to guide future investigations of the mechanisms underlying axonal pathology and inflammation induced by mechanical trauma. HIGHLIGHTSBiomechanical input energy predicts biological responses in mouse CHIMERA TBI.Impact energies of 0.4 J and below are subthreshold and produce no injury phenotype.Injury threshold is 0.5 J, where at least one biological outcome is altered.Mild TBI phenotypes are observed at 0.6 J and above.


Experimental Neurology | 2018

Age at injury and genotype modify acute inflammatory and neurofilament-light responses to mild CHIMERA traumatic brain injury in wild-type and APP/PS1 mice

Wai Hang Cheng; Sophie Stukas; Kris M. Martens; Dhananjay Namjoshi; Emily B. Button; Anna Wilkinson; Asma Bashir; Jérôme Robert; Peter A. Cripton; Cheryl L. Wellington

ABSTRACT Peak incidence of traumatic brain injury (TBI) occurs in both young and old individuals, and older age at injury is associated with worse outcome and poorer recovery. Moderate‐severe TBI is a reported risk factor for dementia, including Alzheimers disease (AD), but whether mild TBI (mTBI) alters AD pathogenesis is not clear. To delineate how age at injury and predisposition to amyloid formation affect the acute response to mTBI, we used the Closed Head Impact Model of Engineered Rotational Acceleration (CHIMERA) model of TBI to induce two mild injuries in wild‐type (WT) and APP/PS1 mice at either 6 or 13 months of age and assessed behavioural, histological and biochemical changes up to 14 days post‐injury. Age at injury did not alter acute behavioural responses to mTBI, including measures of neurological status, motor performance, spatial memory, fear, or anxiety, in either strain. Young APP/PS1 mice showed a subtle and transient increase in diffuse A&bgr; deposits after injury, whereas old APP/PS1 mice showed decreased amyloid deposits, without significant alterations in total soluble or insoluble A&bgr; levels at either age. Age at injury and genotype showed complex responses with respect to microglial and cytokine outcomes, where post‐injury neuroinflammation is increased in old WT mice but attenuated in old APP/PS1 mice. Intriguingly, silver staining confirmed axonal damage in both strains and ages, yet only young WT and APP/PS1 mice showed neurofilament‐positive axonal swellings after mTBI, as this response was almost entirely attenuated in old mice. Plasma neurofilament‐light levels were significantly elevated after injury only in young APP/PS1 mice. This study suggests that mild TBI has minimal effects on A&bgr; metabolism, but that age and genotype can each modify acute outcomes related to white matter injury. Graphical abstract Figure. No Caption available. HighlightsA&bgr; metabolism is not robustly affected by two mild concussive TBIs in APP/PS1 mice.Acute white matter inflammation after TBI is modified both by age and genotype.Age at injury markedly affects the neurofilament response to injury.


eNeuro | 2017

Defining an Analytic Framework to Evaluate Quantitative MRI Markers of Traumatic Axonal Injury: Preliminary Results in a Mouse Closed Head Injury Model.

M. Haber; E. B. Hutchinson; N. Sadeghi; Wai Hang Cheng; Dhananjay Namjoshi; Peter A. Cripton; M. O. Irfanoglu; Cheryl L. Wellington; R. Diaz-Arrastia; C. Pierpaoli

Visual Abstract Diffuse axonal injury (DAI) is a hallmark of traumatic brain injury (TBI) pathology. Recently, the Closed Head Injury Model of Engineered Rotational Acceleration (CHIMERA) was developed to generate an experimental model of DAI in a mouse. The characterization of DAI using diffusion tensor magnetic resonance imaging (MRI; diffusion tensor imaging, DTI) may provide a useful set of outcome measures for preclinical and clinical studies. The objective of this study was to identify the complex neurobiological underpinnings of DTI features following DAI using a comprehensive and quantitative evaluation of DTI and histopathology in the CHIMERA mouse model. A consistent neuroanatomical pattern of pathology in specific white matter tracts was identified across ex vivo DTI maps and photomicrographs of histology. These observations were confirmed by voxelwise and regional analysis of DTI maps, demonstrating reduced fractional anisotropy (FA) in distinct regions such as the optic tract. Similar regions were identified by quantitative histology and exhibited axonal damage as well as robust gliosis. Additional analysis using a machine-learning algorithm was performed to identify regions and metrics important for injury classification in a manner free from potential user bias. This analysis found that diffusion metrics were able to identify injured brains almost with the same degree of accuracy as the histology metrics. Good agreement between regions detected as abnormal by histology and MRI was also found. The findings of this work elucidate the complexity of cellular changes that give rise to imaging abnormalities and provide a comprehensive and quantitative evaluation of the relative importance of DTI and histological measures to detect brain injury.


Alzheimers & Dementia | 2014

INTRAVENOUSLY INJECTED HUMAN APOLIPOPROTEIN A-I RAPIDLY ENTERS THE CENTRAL NERVOUS SYSTEM VIA THE CHOROID PLEXUS IN MICE

Sophie Stukas; Jérôme Robert; Michael Lee; Iva Kulic; Nicole DeValle; Michael Carr; Jianjia Fan; Dhananjay Namjoshi; Kalistyne Lemke; Michael N. Oda; Cheryl L. Wellington

Background: Lipoprotein metabolism in the brain is based on particles that resemble high-density lipoproteins (HDL) that use apolipoprotein (apo) E as opposed to apoA-I as their primary protein component. Although apoA-I is not synthesized by astrocytes or microglia, which secrete apoE, it is abundant in cerebrospinal fluid (CSF) and is readily detectable in brain tissue lysates.However, the mechanisms by which plasma apoA-I enters and is metabolized within the central nervous system (CNS) are unknown. Methods and Results: Western blot analysis shows that steady state levels of endogenous apoA-I in CSF and brain are approximately 0.01% and 10-15% of its levels in plasma and liver, respectively. Recombinant, fluorescently tagged, lipid-free human (h) apoA-I injected into the tail vein of wild-type mice localizes to the choroid plexus within 0.5h and accumulates in a saturable, dose-dependent manner in brain. hApoA-I accumulates in the brain for up to 2h, after which it is turned over with a half life of ~133 minutes, 3 times longer than the relatively quick turnover 40-45 minutes found in plasma, liver, and kidney. In vitro, hApoA-I is taken up and actively transported across confluent monolayers of primary human choroid epithelial cells. Conclusions: Following intravenous injection, hApoA-I rapidly and strongly localizes to the choroid plexus, suggesting it gains access to the CNS primarily via the blood CSF barrier. Further, apoA-I found in the CNS of mice is exclusively derived from the circulation as apoA-I mRNA is not detectable in murine brain. These results suggest that apoA-I based HDL may primarily play a role in CSF lipoprotein metabolism in addition to potentially impacting cerebrovasculature health and function from the lumen of the vessel.

Collaboration


Dive into the Dhananjay Namjoshi's collaboration.

Top Co-Authors

Avatar

Cheryl L. Wellington

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Sophie Stukas

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Anna Wilkinson

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Jianjia Fan

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Michael Carr

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Peter A. Cripton

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Wai Hang Cheng

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

James Donkin

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jeniffer Chan

University of British Columbia

View shared research outputs
Researchain Logo
Decentralizing Knowledge