Network


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

Hotspot


Dive into the research topics where Erica K.M. Reeves is active.

Publication


Featured researches published by Erica K.M. Reeves.


Embo Molecular Medicine | 2013

VBP15, a novel anti-inflammatory and membrane-stabilizer, improves muscular dystrophy without side effects.

Christopher R. Heier; Jesse M. Damsker; Qing Yu; Blythe C. Dillingham; Tony Huynh; Jack H. Van der Meulen; Arpana Sali; Brittany K. Miller; Aditi Phadke; Luana Scheffer; James Quinn; Kathleen Tatem; Sarah Jordan; Sherry Dadgar; Olga Rodriguez; Chris Albanese; Michael E. Calhoun; Heather Gordish-Dressman; Jyoti K. Jaiswal; Edward M. Connor; John M. McCall; Eric P. Hoffman; Erica K.M. Reeves; Kanneboyina Nagaraju

Absence of dystrophin makes skeletal muscle more susceptible to injury, resulting in breaches of the plasma membrane and chronic inflammation in Duchenne muscular dystrophy (DMD). Current management by glucocorticoids has unclear molecular benefits and harsh side effects. It is uncertain whether therapies that avoid hormonal stunting of growth and development, and/or immunosuppression, would be more or less beneficial. Here, we discover an oral drug with mechanisms that provide efficacy through anti‐inflammatory signaling and membrane‐stabilizing pathways, independent of hormonal or immunosuppressive effects. We find VBP15 protects and promotes efficient repair of skeletal muscle cells upon laser injury, in opposition to prednisolone. Potent inhibition of NF‐κB is mediated through protein interactions of the glucocorticoid receptor, however VBP15 shows significantly reduced hormonal receptor transcriptional activity. The translation of these drug mechanisms into DMD model mice improves muscle strength, live‐imaging and pathology through both preventive and post‐onset intervention regimens. These data demonstrate successful improvement of dystrophy independent of hormonal, growth, or immunosuppressive effects, indicating VBP15 merits clinical investigation for DMD and would benefit other chronic inflammatory diseases.


BMC Medical Genetics | 2007

PPARα L162V underlies variation in serum triglycerides and subcutaneous fat volume in young males

Julieta Uthurralt; Heather Gordish-Dressman; Meg Bradbury; Carolina Tesi-Rocha; Joseph M. Devaney; Brennan Harmon; Erica K.M. Reeves; Cinzia Brandoli; Barbara C. Hansen; Richard L. Seip; Paul D. Thompson; Thomas B. Price; Theodore J. Angelopoulos; Priscilla M. Clarkson; Niall M. Moyna; Linda S. Pescatello; Paul S. Visich; Robert F. Zoeller; Paul M. Gordon; Eric P. Hoffman

BackgroundOf the five sub-phenotypes defining metabolic syndrome, all are known to have strong genetic components (typically 50–80% of population variation). Studies defining genetic predispositions have typically focused on older populations with metabolic syndrome and/or type 2 diabetes. We hypothesized that the study of younger populations would mitigate many confounding variables, and allow us to better define genetic predisposition loci for metabolic syndrome.MethodsWe studied 610 young adult volunteers (average age 24 yrs) for metabolic syndrome markers, and volumetric MRI of upper arm muscle, bone, and fat pre- and post-unilateral resistance training.ResultsWe found the PPARα L162V polymorphism to be a strong determinant of serum triglyceride levels in young White males, where carriers of the V allele showed 78% increase in triglycerides relative to L homozygotes (LL = 116 ± 11 mg/dL, LV = 208 ± 30 mg/dL; p = 0.004). Men with the V allele showed lower HDL (LL = 42 ± 1 mg/dL, LV = 34 ± 2 mg/dL; p = 0.001), but women did not. Subcutaneous fat volume was higher in males carrying the V allele, however, exercise training increased fat volume of the untrained arm in V carriers, while LL genotypes significantly decreased in fat volume (LL = -1,707 ± 21 mm3, LV = 17,617 ± 58 mm3 ; p = 0.002), indicating a systemic effect of the V allele on adiposity after unilateral training. Our study suggests that the primary effect of PPARα L162V is on serum triglycerides, with downstream effects on adiposity and response to training.ConclusionOur results on association of PPARα and triglycerides in males showed a much larger effect of the V allele than previously reported in older and less healthy populations. Specifically, we showed the V allele to increase triglycerides by 78% (p = 0.004), and this single polymorphism accounted for 3.8% of all variation in serum triglycerides in males (p = 0.0037).


Bioorganic & Medicinal Chemistry | 2013

VBP15: Preclinical characterization of a novel anti-inflammatory delta 9,11 steroid

Erica K.M. Reeves; Eric P. Hoffman; Kanneboyina Nagaraju; Jesse M. Damsker; John M. McCall

Δ9,11 modifications of glucocorticoids (21-aminosteroids) have been developed as drugs for protection against cell damage (lipid peroxidation; lazaroids) and inhibition of neovascularization (anecortave). Part of the rationale for developing these compounds has been the loss of glucocorticoid receptor binding due to the Δ9,11 modification, thus avoiding many immunosuppressive activities and deleterious side effect profiles associated with binding to glucocorticoid and mineralocorticoid receptors. We recently demonstrated that anecortave acetate and its 21-hydroxy analog (VBP1) do, in fact, show glucocorticoid and mineralocorticoid receptor binding activities, with potent translocation of the glucocorticoid receptor to the cell nucleus. We concluded that Δ9,11 steroids showed novel anti-inflammatory properties, retaining NF-κB inhibition, but losing deleterious glucocorticoid side effect profiles. Evidence for this was developed in pre-clinical trials of chronic muscle inflammation. Here, we describe a drug development program aimed at optimizing the Δ9,11 chemistry. Twenty Δ9,11 derivatives were tested in in vitro screens for NF-κB inhibition and GR translocation to the nucleus, and low cell toxicity. VBP15 was selected as the lead compound due to potent NF-κB inhibition and GR translocation similar to prednisone and dexamethasone, lack of transactivation properties, and good bioavailability. Phamacokinetics were similar to traditional glucocorticoid drugs with terminal half-life of 0.35 h (mice), 0.58 h (rats), 5.42 h (dogs), and bioavailability of 74.5% (mice), and 53.2% (dogs). Metabolic stability showed ≥80% remaining at 1 h of VBP6 and VBP15 in human, dog, and monkey liver microsomes. Solubility, permeability and plasma protein binding were within acceptable limits. VBP15 moderately induced CYP3A4 across the three human hepatocyte donors (24-42%), similar to other steroids. VBP15 is currently under development for treatment of Duchenne muscular dystrophy.


Journal of Pharmacology and Experimental Therapeutics | 2012

Δ-9,11 Modification of Glucocorticoids Dissociates Nuclear Factor-κB Inhibitory Efficacy from Glucocorticoid Response Element-Associated Side Effects

Andreas R. Baudy; Erica K.M. Reeves; Jesse M. Damsker; Christopher R. Heier; Lindsay M. Garvin; Blythe C. Dillingham; John M. McCall; Sree Rayavarapu; Zuyi Wang; Jack Vandermeulen; Arpana Sali; Vanessa Jahnke; Stephanie Duguez; Debra C. DuBois; Mary C. Rose; Kanneboyina Nagaraju; Eric P. Hoffman

Glucocorticoids are standard of care for many inflammatory conditions, but chronic use is associated with a broad array of side effects. This has led to a search for dissociative glucocorticoids—drugs able to retain or improve efficacy associated with transrepression [nuclear factor-κB (NF-κB) inhibition] but with the loss of side effects associated with transactivation (receptor-mediated transcriptional activation through glucocorticoid response element gene promoter elements). We investigated a glucocorticoid derivative with a Δ-9,11 modification as a dissociative steroid. The Δ-9,11 analog showed potent inhibition of tumor necrosis factor-α-induced NF-κB signaling in cell reporter assays, and this transrepression activity was blocked by 17β-hydroxy-11β-[4-dimethylamino phenyl]-17α-[1-propynyl]estra-4,9-dien-3-one (RU-486), showing the requirement for the glucocorticoid receptor (GR). The Δ-9,11 analog induced the nuclear translocation of GR but showed the loss of transactivation as assayed by GR-luciferase constructs as well as mRNA profiles of treated cells. The Δ-9,11 analog was tested for efficacy and side effects in two mouse models of muscular dystrophy: mdx (dystrophin deficiency), and SJL (dysferlin deficiency). Daily oral delivery of the Δ-9,11 analog showed a reduction of muscle inflammation and improvements in multiple muscle function assays yet no reductions in body weight or spleen size, suggesting the loss of key side effects. Our data demonstrate that a Δ-9,11 analog dissociates the GR-mediated transcriptional activities from anti-inflammatory activities. Accordingly, Δ-9,11 analogs may hold promise as a source of safer therapeutic agents for chronic inflammatory disorders.


Cellular and Molecular Neurobiology | 2015

VBP15, a Novel Anti-Inflammatory, is Effective at Reducing the Severity of Murine Experimental Autoimmune Encephalomyelitis

Blythe C. Dillingham; Susan M. Knoblach; Gina M. Many; Brennan Harmon; Amanda M. Mullen; Christopher R. Heier; Luca Bello; John M. McCall; Eric P. Hoffman; Edward M. Connor; Kanneboyina Nagaraju; Erica K.M. Reeves; Jesse M. Damsker

Multiple sclerosis is a chronic disease of the central nervous system characterized by an autoimmune inflammatory reaction that leads to axonal demyelination and tissue damage. Glucocorticoids, such as prednisolone, are effective in the treatment of multiple sclerosis in large part due to their ability to inhibit pro-inflammatory pathways (e.g., NFκB). However, despite their effectiveness, long-term treatment is limited by adverse side effects. VBP15 is a recently described compound synthesized based on the lazeroid steroidal backbone that shows activity in acute and chronic inflammatory conditions, yet displays a much-reduced side effect profile compared to traditional glucocorticoids. The purpose of this study was to determine the effectiveness of VBP15 in inhibiting inflammation and disease progression in experimental autoimmune encephalomyelitis (EAE), a widely used mouse model of multiple sclerosis. Our data show that VBP15 is effective at reducing both disease onset and severity. In parallel studies, we observed that VBP15 was able to inhibit the production of NFκB-regulated pro-inflammatory transcripts in human macrophages. Furthermore, treatment with prednisolone—but not VBP15—increased expression of genes associated with bone loss and muscle atrophy, suggesting lack of side effects of VBP15. These findings suggest that VBP15 may represent a potentially safer alternative to traditional glucocorticoids in the treatment of multiple sclerosis and other inflammatory diseases.


PLOS ONE | 2013

VBP15, a glucocorticoid analogue, is effective at reducing allergic lung inflammation in mice.

Jesse M. Damsker; Blythe C. Dillingham; Mary C. Rose; Molly A. Balsley; Christopher R. Heier; Alan M. Watson; Erik J. Stemmy; Rosalyn A. Jurjus; Tony Huynh; Kathleen Tatem; Kitipong Uaesoontrachoon; Dana M. Berry; Angela S. Benton; Robert J. Freishtat; Eric P. Hoffman; John M. McCall; Heather Gordish-Dressman; Stephanie L. Constant; Erica K.M. Reeves; Kanneboyina Nagaraju

Asthma is a chronic inflammatory condition of the lower respiratory tract associated with airway hyperreactivity and mucus obstruction in which a majority of cases are due to an allergic response to environmental allergens. Glucocorticoids such as prednisone have been standard treatment for many inflammatory diseases for the past 60 years. However, despite their effectiveness, long-term treatment is often limited by adverse side effects believed to be caused by glucocorticoid receptor-mediated gene transcription. This has led to the pursuit of compounds that retain the anti-inflammatory properties yet lack the adverse side effects associated with traditional glucocorticoids. We have developed a novel series of steroidal analogues (VBP compounds) that have been previously shown to maintain anti-inflammatory properties such as NFκB-inhibition without inducing glucocorticoid receptor-mediated gene transcription. This study was undertaken to determine the effectiveness of the lead compound, VBP15, in a mouse model of allergic lung inflammation. We show that VBP15 is as effective as the traditional glucocorticoid, prednisolone, at reducing three major hallmarks of lung inflammation—NFκB activity, leukocyte degranulation, and pro-inflammatory cytokine release from human bronchial epithelial cells obtained from patients with asthma. Moreover, we found that VBP15 is capable of reducing inflammation of the lung in vivo to an extent similar to that of prednisone. We found that prednisolone–but not VBP15 shortens the tibia in mice upon a 5 week treatment regimen suggesting effective dissociation of side effects from efficacy. These findings suggest that VBP15 may represent a potent and safer alternative to traditional glucocorticoids in the treatment of asthma and other inflammatory diseases.


Physical Medicine and Rehabilitation Clinics of North America | 2012

Novel Approaches to Corticosteroid Treatment in Duchenne Muscular Dystrophy

Eric P. Hoffman; Erica K.M. Reeves; Jesse M. Damsker; Kanneboyina Nagaraju; John M. McCall; Edward M. Connor; Kate Bushby


Endocrine‚ Metabolic & Immune Disorders-Drug Targets | 2012

Glucocorticoid Analogues: Potential Therapeutic Alternatives for Treating Inflammatory Muscle Diseases

Erica K.M. Reeves; Sree Rayavarapu; Jesse M. Damsker; Kanneboyina Nagaraju


Human Genetics | 2010

Functional characterization of a haplotype in the AKT1 gene associated with glucose homeostasis and metabolic syndrome.

Brennan Harmon; Stephanie A. Devaney; Heather Gordish-Dressman; Erica K.M. Reeves; Po Zhao; Joseph M. Devaney; Eric P. Hoffman


Proteome Science | 2009

Proteomic profiling of glucocorticoid-exposed myogenic cells: Time series assessment of protein translocation and transcription of inactive mRNAs

Erica K.M. Reeves; Heather Gordish-Dressman; Eric P. Hoffman; Yetrib Hathout

Collaboration


Dive into the Erica K.M. Reeves's collaboration.

Top Co-Authors

Avatar

Eric P. Hoffman

Children's National Medical Center

View shared research outputs
Top Co-Authors

Avatar

Jesse M. Damsker

Children's National Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Heather Gordish-Dressman

Children's National Medical Center

View shared research outputs
Top Co-Authors

Avatar

Blythe C. Dillingham

Children's National Medical Center

View shared research outputs
Top Co-Authors

Avatar

Brennan Harmon

Children's National Medical Center

View shared research outputs
Top Co-Authors

Avatar

Christopher R. Heier

Children's National Medical Center

View shared research outputs
Top Co-Authors

Avatar

Mary C. Rose

Children's National Medical Center

View shared research outputs
Top Co-Authors

Avatar

Alan M. Watson

University of Pittsburgh

View shared research outputs
Top Co-Authors

Avatar

Angela S. Benton

Children's National Medical Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge