Network


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

Hotspot


Dive into the research topics where Robert H. Scannevin is active.

Publication


Featured researches published by Robert H. Scannevin.


Nature | 2016

The antibody aducanumab reduces Aβ plaques in Alzheimer's disease.

Jeff Sevigny; Ping Chiao; Thierry Bussiere; Paul H. Weinreb; Leslie Williams; Marcel Maier; Robert Dunstan; Stephen Salloway; Tianle Chen; Yan Ling; O'Gorman J; Fang Qian; Mahin Arastu; Mingwei Li; Sowmya Chollate; Melanie Brennan; Omar Quintero-Monzon; Robert H. Scannevin; Arnold Hm; Thomas Engber; Kenneth J. Rhodes; James Ferrero; Hang Y; Alvydas Mikulskis; Jan Grimm; Christoph Hock; Roger M. Nitsch; Alfred Sandrock

Alzheimer’s disease (AD) is characterized by deposition of amyloid-β (Aβ) plaques and neurofibrillary tangles in the brain, accompanied by synaptic dysfunction and neurodegeneration. Antibody-based immunotherapy against Aβ to trigger its clearance or mitigate its neurotoxicity has so far been unsuccessful. Here we report the generation of aducanumab, a human monoclonal antibody that selectively targets aggregated Aβ. In a transgenic mouse model of AD, aducanumab is shown to enter the brain, bind parenchymal Aβ, and reduce soluble and insoluble Aβ in a dose-dependent manner. In patients with prodromal or mild AD, one year of monthly intravenous infusions of aducanumab reduces brain Aβ in a dose- and time-dependent manner. This is accompanied by a slowing of clinical decline measured by Clinical Dementia Rating—Sum of Boxes and Mini Mental State Examination scores. The main safety and tolerability findings are amyloid-related imaging abnormalities. These results justify further development of aducanumab for the treatment of AD. Should the slowing of clinical decline be confirmed in ongoing phase 3 clinical trials, it would provide compelling support for the amyloid hypothesis.


Journal of Pharmacology and Experimental Therapeutics | 2012

Fumarates Promote Cytoprotection of Central Nervous System Cells against Oxidative Stress via the Nuclear Factor (Erythroid-Derived 2)-Like 2 Pathway

Robert H. Scannevin; Sowmya Chollate; Mi-young Jung; Melanie Shackett; Hiral Patel; Pradeep Bista; Weike Zeng; Sarah Ryan; Masayuki Yamamoto; Matvey E. Lukashev; Kenneth J. Rhodes

Oxidative stress is central to the pathology of several neurodegenerative diseases, including multiple sclerosis, and therapeutics designed to enhance antioxidant potential could have clinical value. The objective of this study was to characterize the potential direct neuroprotective effects of dimethyl fumarate (DMF) and its primary metabolite monomethyl fumarate (MMF) on cellular resistance to oxidative damage in primary cultures of central nervous system (CNS) cells and further explore the dependence and function of the nuclear factor (erythroid-derived 2)-like 2 (Nrf2) pathway in this process. Treatment of animals or primary cultures of CNS cells with DMF or MMF resulted in increased nuclear levels of active Nrf2, with subsequent up-regulation of canonical antioxidant target genes. DMF-dependent up-regulation of antioxidant genes in vivo was lost in mice lacking Nrf2 [Nrf2(−/−)]. DMF or MMF treatment increased cellular redox potential, glutathione, ATP levels, and mitochondrial membrane potential in a concentration-dependent manner. Treating astrocytes or neurons with DMF or MMF also significantly improved cell viability after toxic oxidative challenge in a concentration-dependent manner. This effect on viability was lost in cells that had eliminated or reduced Nrf2. These data suggest that DMF and MMF are cytoprotective for neurons and astrocytes against oxidative stress-induced cellular injury and loss, potentially via up-regulation of an Nrf2-dependent antioxidant response. These data also suggest DMF and MMF may function through improving mitochondrial function. The clinical utility of DMF in multiple sclerosis is being explored through phase III trials with BG-12, which is an oral therapeutic containing DMF as the active ingredient.


Stroke | 2008

Extension of the Thrombolytic Time Window With Minocycline in Experimental Stroke

Yoshihiro Murata; Anna Rosell; Robert H. Scannevin; Kenneth J. Rhodes; Xiaoying Wang; Eng H. Lo

Background and Purpose— Thrombolysis with tPA is the only FDA-approved therapy for acute ischemic stroke. But its widespread application remains limited by narrow treatment time windows and the related risks of cerebral hemorrhage. In this study, we ask whether minocycline can prevent tPA-associated cerebral hemorrhage and extend the reperfusion window in an experimental stroke model in rats. Methods— Spontaneously hypertensive rats were subjected to embolic focal ischemia using homologous clots and treated with: saline at 1 hour; early tPA at 1 hour, delayed tPA at 6 hours; minocycline at 4 hours; combined minocycline at 4 hours plus tPA at 6 hours. Infarct volumes and hemorrhagic transformation were quantified at 24 hours. Gelatin zymography was used to measure blood levels of circulating matrix metalloproteinase-9 (MMP-9). Results— Early 1-hour thrombolysis restored perfusion and reduced infarction. Late 6-hour tPA did not decrease infarction but instead worsened hemorrhagic conversion. Combining minocycline with delayed 6-hour tPA decreased plasma MMP-9 levels, reduced infarction, and ameliorated brain hemorrhage. Blood levels of MMP-9 were also significantly correlated with volumes of infarction and hemorrhage. Conclusion— Combination therapy with minocycline may extend tPA treatment time windows in ischemic stroke.


Proceedings of the National Academy of Sciences of the United States of America | 2001

Identification of a trafficking determinant localized to the Kv1 potassium channel pore

Louis N. Manganas; Qiang Wang; Robert H. Scannevin; Dana E. Antonucci; Kenneth J. Rhodes; James S. Trimmer

The repertoire of Kv1 potassium channels expressed in presynaptic terminals of mammalian central neurons is shaped by intrinsic trafficking signals that determine surface-expression efficiencies of homomeric and heteromeric Kv1 channel complexes. Here, we show that a determinant controlling surface expression of Kv1 channels is localized to the highly conserved pore region. Point-mutation analysis revealed two residues as critical for channel trafficking, one in the extracellular “turret” domain and one in the region distal to the selectivity filter. Interestingly, these same residues also form the binding sites for polypeptide neurotoxins. Our findings demonstrate a previously uncharacterized function for the channel-pore domain as a regulator of channel trafficking.


Neuroscience Letters | 2007

Induction of matrix metalloproteinase, cytokines and chemokines in rat cortical astrocytes exposed to plasminogen activators

Sun-Ryung Lee; Shuzhen Guo; Robert H. Scannevin; Brian C. Magliaro; Kenneth J. Rhodes; Xiaoying Wang; Eng H. Lo

Plasminogen activators are used in thrombolytic stroke therapy. However, it is increasingly recognized that they have other actions besides fibrinolysis. In this study, we assess potential pro-inflammatory effects of tissue-type plasminogen activator (tPA) and urokinase-type plasminogen activator (uPA) in rat cortical astrocytes. Both uPA and tPA induced rapid dose-dependent upregulation in MMP-2 and MMP-9, as demonstrated by zymography of conditioned media. In addition, a multiplex ELISA array demonstrated that patterned responses in chemokines and cytokines were also evoked. Exposure to tPA induced elevations in secreted MIP-2, MCP-1 and GRO/KC. Exposure to uPA induced elevations in secreted IFN-gamma, TNF-alpha, GMCSF, MIP-1alpha, MIP-2, MIP-3alpha, MCP-1, RANTES and fractalkine. These data suggest that plasminogen activators may trigger selected pro-inflammatory responses at the neurovascular interface. Whether these effects influence thrombolytic stroke therapy warrants further investigation.


Journal of Medicinal Chemistry | 2012

Design and Characterization of Optimized Adenosine A2A/A1 Receptor Antagonists for the Treatment of Parkinson's Disease

Brian C. Shook; Stefanie Rassnick; Nathaniel H. Wallace; Jeffrey Crooke; Mark Ault; Devraj Chakravarty; J. Kent Barbay; Aihua Wang; Mark T. Powell; Kristi Leonard; Vernon Alford; Robert H. Scannevin; Karen I. Carroll; Lisa Lampron; Lori Westover; Heng-Keang Lim; Ronald K. Russell; Shawn Branum; Kenneth M. Wells; Sandra Damon; Scott Youells; Xun Li; Derek A. Beauchamp; Kenneth J. Rhodes; Paul F. Jackson

The design and characterization of two, dual adenosine A(2A)/A(1) receptor antagonists in several animal models of Parkinsons disease is described. Compound 1 was previously reported as a potential treatment for Parkinsons disease. Further characterization of 1 revealed that it was metabolized to reactive intermediates that caused the genotoxicity of 1 in the Ames and mouse lymphoma L51784 assays. The identification of the metabolites enabled the preparation of two optimized compounds 13 and 14 that were devoid of the metabolic liabilities associated with 1. Compounds 13 and 14 are potent dual A(2A)/A(1) receptor antagonists that have excellent activity, after oral administration, across a number of animal models of Parkinsons disease including mouse and rat models of haloperidol-induced catalepsy, mouse and rat models of reserpine-induced akinesia, and the rat 6-hydroxydopamine (6-OHDA) lesion model of drug-induced rotation.


ACS Medicinal Chemistry Letters | 2011

Discovery of BIIB042, a Potent, Selective, and Orally Bioavailable γ-Secretase Modulator.

Hairuo Peng; Tina Talreja; Zhili Xin; J. Hernan Cuervo; Gnanasambandam Kumaravel; Michael J. Humora; Lin Xu; Ellen Rohde; Lawrence Gan; Mi-young Jung; Melanie Shackett; Sowmya Chollate; Anthone W. Dunah; Pamela A. Snodgrass-Belt; H. Moore Arnold; Arthur G. Taveras; Kenneth J. Rhodes; Robert H. Scannevin

We have investigated a novel series of acid-derived γ-secretase modulators as a potential treatment of Alzheimers disease. Optimization based on cellular potency and brain pharmacodynamics after oral dosing led to the discovery of 10a (BIIB042). Compound 10a is a potent γ-secretase modulator, which lowered Aβ42, increased Aβ38, but had little to no effect on Aβ40 levels both in vitro and in vivo. In addition, compound 10a did not affect Notch signaling in our in vitro assessment. Compound 10a demonstrated excellent pharmacokinetic parameters in multiple species. Oral administration of 10a significantly reduced brain Aβ42 levels in CF-1 mice and Fischer rats, as well as plasma Aβ42 levels in cynomolgus monkeys. Compound 10a was selected as a candidate for preclinical safety evaluation.


MedChemComm | 2011

Aminomethyl substituted thieno[2,3-d]pyrimidines as adenosine A2A receptor antagonists

Brian C. Shook; Devraj Charavarty; J. Kent Barbay; Aihua Wang; Kristi Leonard; Vernon Alford; Mark P. Powell; Derek A. Beauchamp; Stefanie Rassnick; Robert H. Scannevin; Karen Carroll; Nathaniel Wallace; Jeffrey Crooke; Mark Ault; Lisa Lampron; Lori Westover; Kenneth J. Rhodes; Paul F. Jackson

A novel series of aminomethyl substituted thieno[2,3-d]pyrimidines have been identified as adenosine A2A receptor antagonists. Analogues show excellent in vitro activities and have excellent activity in vivo in mouse models of Parkinsons disease.


Bioorganic & Medicinal Chemistry Letters | 2010

Stereochemistry activity relationship of orally active tetralin S1P agonist prodrugs

Bin Ma; Kevin Guckian; Edward Lin; Wen-Cherng Lee; Daniel J. Scott; Gnanasambandam Kumaravel; Timothy L. Macdonald; Kevin R. Lynch; Cheryl Black; Sowmya Chollate; Kyungmin Hahm; Gregg Hetu; Ping Jin; Yi Luo; Ellen Rohde; Anthony Rossomando; Robert H. Scannevin; Joy Wang; Chunhua Yang

Modifying FTY720, an immunosuppressant modulator, led to a new series of well phosphorylated tetralin analogs as potent S1P1 receptor agonists. The stereochemistry effect of tetralin ring was probed, and (-)-(R)-2-amino-2-((S)-6-octyl-1,2,3,4-tetrahydronaphthalen-2-yl)propan-1-ol was identified as a good SphK2 substrate and potent S1P1 agonist with good oral bioavailability.


Scientific Reports | 2017

The NRF2 transcriptional target, OSGIN1 , contributes to monomethyl fumarate-mediated cytoprotection in human astrocytes

Melanie Brennan; Maria Matos; Karl E. Richter; Bing Li; Robert H. Scannevin

Dimethyl fumarate (DMF) is indicated for the treatment of relapsing multiple sclerosis and may exert therapeutic effects via activation of the nuclear factor (erythroid-derived 2)-like 2 (NRF2) pathway. Following oral DMF administration, central nervous system (CNS) tissue is predominantly exposed to monomethyl fumarate (MMF), the bioactive metabolite of DMF, which can stabilize NRF2 and induce antioxidant gene expression; however, the detailed NRF2-dependent mechanisms modulated by MMF that lead to cytoprotection are unknown. Our data identify a mechanism for MMF-mediated cytoprotection in human astrocytes that functions in an OSGIN1-dependent manner, specifically via upregulation of the OSGIN1-61 kDa isoform. NRF2-dependent OSGIN1 expression induced P53 nuclear translocation following MMF administration, leading to cell-cycle inhibition and cell protection against oxidative challenge. This study provides mechanistic insight into MMF-mediated cytoprotection via NRF2, OSGIN1, and P53 in human CNS-derived cells and contributes to our understanding of how DMF may act clinically to ameliorate pathological processes in neurodegenerative disease.

Collaboration


Dive into the Robert H. Scannevin's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Matthew J. Todd

Michigan State University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Aihua Wang

University of Science and Technology

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge