Network


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

Hotspot


Dive into the research topics where J. Fred Hess is active.

Publication


Featured researches published by J. Fred Hess.


Biochemical and Biophysical Research Communications | 1992

Cloning and pharmacological characterization of a human bradykinin (BK-2) receptor

J. Fred Hess; Joseph A. Borkowski; Gretchen S. Young; Catherine D. Strader; Richard W. Ransom

A human BK-2 bradykinin receptor was cloned from the lung fibroblast cell line CCD-16Lu. The cDNA clone encodes a 364 amino acid protein that has the characteristics of a seven transmembrane domain G-protein coupled receptor. The predicted amino acid sequence of the human BK-2 receptor is 81% identical to the smooth muscle rat BK-2 receptor (1). Transfection of the human BK-2 receptor cDNA into COS-7 cells results in the expression of high levels of specific BK binding sites. Saturation binding analysis indicates that the human BK-2 receptor expressed in COS-7 cells binds BK with a KD of 0.13 nM. Pharmacological characterization of the expressed BK receptor is consistent with the cDNA encoding a receptor of the BK-2 subtype. The BK-2 receptor antagonist Hoe 140 (2), D-Arg0[Hyp3, Thi5, D-Tic7, Oic8]BK has a high affinity (IC50 = 65 pM) for the cloned human receptor. The tissue distribution of the human BK-2 receptor was analyzed by competitive PCR with human tissue cDNA and is similar to that determined for the BK-2 receptor in the rat.


Pain | 1997

Effects of the bradykinin B1 receptor antagonist des-Arg9[Leu8]bradykinin and genetic disruption of the B2 receptor on nociception in rats and mice

N.M.J. Rupniak; Susan Boyce; J.K. Webb; Angela R. Williams; Emma J. Carlson; R.G. Hill; Joseph A. Borkowski; J. Fred Hess

Abstract The contributions of B1 and B2 bradykinin receptors to acute and chronic inflammatory hyperalgesia were examined using the peptide B1 receptor antagonist des‐Arg9[Leu8]bradykinin and transgenic Bk2r‐/‐ mice. In normal rats and mice, des‐Arg9[Leu8]bradykinin (30 nmol/kg i.v. or s.c.) inhibited carrageenan‐induced hyperalgesia and the late phase nociceptive response to formalin. The active dose range was narrow, suggesting partial agonist activity of this peptide. In rats with monoarthritis, des‐Arg9[Leu8]bradykinin (up to 30 nmol/kg i.v.) failed to reduce the number of vocalisations elicited by gentle flexion and extension of the inflamed limb; however, hyperalgesia was exacerbated by administration of the B1 receptor agonist des‐[Arg9]bradykinin (100 nmol/kg i.v.), consistent with other evidence for local induction of B1 receptors during adjuvant‐induced arthritis. The nociceptive response to intraplantar injection of bradykinin (10 nmol) and hyperalgesia induced by carrageenan (0.6 mg) were absent in Bk2r‐/‐ mice, indicating that stimulation of B2 receptors is an essential step in the initiation of some nociceptive and inflammatory reactions. However, the nociceptive response to formalin (2.5% intraplantar), including inhibition of the late phase by des‐Arg9[Leu8]bradykinin (0.3 nmol), and induction of thermal hyperalgesia by Freunds adjuvant (0.1%) appeared intact in Bk2r‐/‐ mice. These findings support other evidence for an involvement of B1 receptors in inflammatory hyperalgesia and suggest that B1 receptor antagonists may be clinically useful as anti‐inflammatory and analgesic drugs.


Biochimica et Biophysica Acta | 1995

Cloning and pharmacological characterization of a rabbit bradykinin B1 receptor

Tanya MacNeil; Kathleen K. Bierilo; John G. Menke; J. Fred Hess

A rabbit B1 bradykinin receptor cDNA was isolated from a rabbit aorta smooth muscle cell library. The 1223 bp cDNA clone encodes a protein of 352 amino acids which is 78% identical to the human bradykinin B1(3) receptor protein. Heterologous expression of the rabbit B1 receptor cDNA in COS-7 cells imparts a high affinity specific binding for 3H-labeled [des-Arg10,Leu9]kallidin. Scatchard analysis indicates that the receptor binds the radiolabeled ligand with a Kd of 0.5 nM. The ability of kallidin (Lys-bradykinin) and bradykinin analogues to compete with binding of 3H-labeled [des-Arg10,Leu9]kallidin was determined and defined a rank order of potency: [des-Arg10,Leu9]kallidin = [des-Arg10]kallidin > [des- Arg9]bradykinin = kallidin >> bradykinin. This receptor exhibits the classical B1 pharmacological property of preferentially binding to kinin analogues which lack the C-terminal arginine. In addition, the affinities for [des-Arg10]kallidin and [des-Arg10,Leu9]kallidin are 100-fold higher than those for the corresponding bradykinin analogues [des-Arg9]bradykinin and [des-Arg9,Leu8]bradykinin which lack the N-terminal lysine. This pharmacological profile is characteristic of the B1 receptor subtype.


Immunopharmacology | 1996

The agonist selectivity of a mouse B1 bradykinin receptor differs from human and rabbit B1 receptors

J. Fred Hess; Amanda W. Derrick; Tanya MacNeil; Joseph A. Borkowski

A genomic clone encoding the mouse B1 receptor was isolated by homology to the human B1 receptor cDNA. The deduced amino acid sequence of the mouse B1 receptor is 72% identical to the human B1 receptor and 73% identical to the rabbit B1 receptor. Ligand binding studies of the mouse B1 receptor expressed in COS cells indicate that it has the pharmacological properties associated with the B1 receptor subtype. However the pharmacology of the mouse receptor is unique in that it possesses a 2-3-fold selectivity for the classical B1 agonist des-Arg9BK over the agonist des-Arg10 kallidin. In contrast, the human and rabbit B1 receptors exhibit an approx. 2000- and 150-fold selectivity, respectively, for des-Arg10kallidin over des-Arg9BK. Thus relative to the human and rabbit B1 receptors the mouse B1 receptor has the opposite selectivity for kinin agonists. The DNA sequence of the region encoding bradykinin was determined for two different mouse kininogen cDNA clones, both encode the sequence Arg-BK. Antipeptide antibodies directed against a C-terminal peptide of the human B1 receptor were produced. Initial characterization of this antibody indicates that it detects specific bands by Western blot analyses that are present in membranes prepared from COS cells transfected with the human B1 receptor cDNA but not from mock transfected COS cells.


Journal of Neurochemistry | 2011

Attenuation of scratch-induced reactive astrogliosis by novel EphA4 kinase inhibitors.

Sophie Parmentier-Batteur; Eleftheria N. Finger; Raghu Krishnan; Hemaka A. Rajapakse; John M. Sanders; Geeta Kandpal; Hong Zhu; Keith P. Moore; Christopher P. Regan; Sujata Sharma; J. Fred Hess; Theresa M. Williams; Ian J. Reynolds; Joseph P. Vacca; Robert J. Mark; Philippe G. Nantermet

J. Neurochem. (2011) 118, 1016–1031.


Molecular Neurodegeneration | 2015

Analysis of tau post-translational modifications in rTg4510 mice, a model of tau pathology.

Lixin Song; Sherry X. Lu; Xuesong Ouyang; Jerry Melchor; Julie Lee; Giuseppe Terracina; Xiaohai Wang; Lynn Hyde; J. Fred Hess; Eric M. Parker; Lili Zhang

BackgroundMicrotubule associated protein tau is the major component of the neurofibrillary tangles (NFTs) found in the brains of patients with Alzheimer’s disease and several other neurodegenerative diseases. Tau mutations are associated with frontotemperal dementia with parkinsonism on chromosome 17 (FTDP-17). rTg4510 mice overexpress human tau carrying the P301L FTDP-17 mutation and develop robust NFT-like pathology at 4–5 months of age. The current study is aimed at characterizing the rTg4510 mice to better understand the genesis of tau pathology and to better enable the use of this model in drug discovery efforts targeting tau pathology.ResultsUsing a panel of immunoassays, we analyzed the age-dependent formation of pathological tau in rTg4510 mice and our data revealed a steady age-dependent accumulation of pathological tau in the insoluble fraction of brain homogenates. The pathological tau was associated with multiple post-translational modifications including aggregation, phosphorylation at a wide variety of sites, acetylation, ubiquitination and nitration. The change of most tau species reached statistical significance at the age of 16 weeks. There was a strong correlation between the different post-translationally modified tau species in this heterogeneous pool of pathological tau. Total tau in the cerebrospinal fluid (CSF) displayed a multiphasic temporal profile distinct from the steady accumulation of pathological tau in the brain. Female rTg4510 mice displayed significantly more aggressive accumulation of pathological tau in the brain and elevation of total tau in CSF than their male littermates.ConclusionThe immunoassays described here were used to generate the most comprehensive description of the changes in various tau species across the lifespan of the rTg4510 mouse model. The data indicate that development of tauopathy in rTg4510 mice involves the accumulation of a pool of pathological tau that carries multiple post-translational modifications, a process that can be detected well before the histological detection of NFTs. Therapeutic treatment targeting tau should therefore aim to reduce all tau species associated with the pathological tau pool rather than reduce specific post-translational modifications. There is still much to learn about CSF tau in physiological and pathological processes in order to use it as a translational biomarker in drug discovery.


Neurochemical Research | 2010

Genome-Wide Microarray Analysis of the Differential Neuroprotective Effects of Antioxidants in Neuroblastoma Cells Overexpressing the Familial Parkinson’s Disease α-Synuclein A53T Mutation

Lei Ma; Tracy T. Cao; Geeta Kandpal; Lee Warren; J. Fred Hess; Guy R. Seabrook; William J. Ray

In Parkinson’s disease substantia nigra neurons degenerate likely due to oxidative damage interacting with genetic risk factors. Here, SH-SY5Y cells expressing wild-type or A53T α-synuclein had increased sensitivity to methyl-4-phenylpyridinium iodide (MPP+), which induces mitochondrial dysfunction, and 6-hydroxydopamine (6-OHDA), which causes oxidative stress. Edaravone protected only against MPP+, and EGCG ((−)-epigallocatechin-3-O-gallate) protected only against 6-OHDA. Thus genomic responses to MPP+ and 6-OHDA in the presence of these antioxidants were analyzed using microarrays. Pathway analysis indicated that MPP+ activated p53 (Pxa0<xa00.001) while 6-OHDA induced the Nrf2 antioxidative stress response (Pxa0<xa00.0001). EGCG was more effective at blocking 6-OHDA-mediated genomic responses, while edaravone was more effective against MPP+. We identified 32 genes that responded to both toxins except in the presence of an effective anti-oxidant; eight are transcription factors and potentially constitute a stress-response transcriptional network. These data provide insights into the mechanisms of neurotoxicity and identifies genes that might mediate antioxidant efficacy.


Molecular Neurodegeneration | 2017

Inhibition of O-GlcNAcase leads to elevation of O-GlcNAc tau and reduction of tauopathy and cerebrospinal fluid tau in rTg4510 mice

Nicholas B. Hastings; Xiaohai Wang; Lixin Song; Brent D. Butts; Diane Grotz; Richard Hargreaves; J. Fred Hess; Kwok-Lam Karen Hong; Cathy Ruey-Ruey Huang; Lynn A. Hyde; Maureen Laverty; Julie Lee; Diane Levitan; Sherry X. Lu; Maureen Maguire; Veeravan Mahadomrongkul; Ernest McEachern; Xuesong Ouyang; Thomas W. Rosahl; Harold G. Selnick; Michaela Stanton; Giuseppe Terracina; David J. Vocadlo; Ganfeng Wang; Joseph L. Duffy; Eric M. Parker; Lili Zhang

BackgroundHyperphosphorylation of microtubule-associated protein tau is a distinct feature of neurofibrillary tangles (NFTs) that are the hallmark of neurodegenerative tauopathies. O-GlcNAcylation is a lesser known post-translational modification of tau that involves the addition of N-acetylglucosamine onto serine and threonine residues. Inhibition of O-GlcNAcase (OGA), the enzyme responsible for the removal of O-GlcNAc modification, has been shown to reduce tau pathology in several transgenic models. Clarifying the underlying mechanism by which OGA inhibition leads to the reduction of pathological tau and identifying translatable measures to guide human dosing and efficacy determination would significantly facilitate the clinical development of OGA inhibitors for the treatment of tauopathies.MethodsGenetic and pharmacological approaches are used to evaluate the pharmacodynamic response of OGA inhibition. A panel of quantitative biochemical assays is established to assess the effect of OGA inhibition on pathological tau reduction. A “click” chemistry labeling method is developed for the detection of O-GlcNAcylated tau.ResultsSubstantial (>80%) OGA inhibition is required to observe a measurable increase in O-GlcNAcylated proteins in the brain. Sustained and substantial OGA inhibition via chronic treatment with Thiamet G leads to a significant reduction of aggregated tau and several phosphorylated tau species in the insoluble fraction of rTg4510 mouse brain and total tau in cerebrospinal fluid (CSF). O-GlcNAcylated tau is elevated by Thiamet G treatment and is found primarily in the soluble 55 kD tau species, but not in the insoluble 64 kD tau species thought as the pathological entity.ConclusionThe present study demonstrates that chronic inhibition of OGA reduces pathological tau in the brain and total tau in the CSF of rTg4510 mice, most likely by directly increasing O-GlcNAcylation of tau and thereby maintaining tau in the soluble, non-toxic form by reducing tau aggregation and the accompanying panoply of deleterious post-translational modifications. These results clarify some conflicting observations regarding the effects and mechanism of OGA inhibition on tau pathology, provide pharmacodynamic tools to guide human dosing and identify CSF total tau as a potential translational biomarker. Therefore, this study provides additional support to develop OGA inhibitors as a treatment for Alzheimer’s disease and other neurodegenerative tauopathies.


PLOS ONE | 2018

Early intervention of tau pathology prevents behavioral changes in the rTg4510 mouse model of tauopathy

Xiaohai Wang; Karen Smith; Michelle Pearson; Anna E. Hughes; Mali Cosden; Jacob N. Marcus; J. Fred Hess; Mary J. Savage; Thomas W. Rosahl; Sean M. Smith; Joel B. Schachter; Jason M. Uslaner

Although tau pathology, behavioral deficits, and neuronal loss are observed in patients with tauopathies, the relationship between these endpoints has not been clearly established. Here we found that rTg4510 mice, which overexpress human mutant tau in the forebrain, develop progressive age-dependent increases in locomotor activity (LMA), which correlates with neurofibrillary tangle (NFT) pathology, hyperphosphorylated tau levels, and brain atrophy. To further clarify the relationship between these endpoints, we treated the rTg4510 mice with either doxycycline to reduce mutant tau expression or an O-GlcNAcase inhibitor Thiamet G, which has been shown to ameliorate tau pathology in animal models. We found that both doxycycline and Thiamet G treatments starting at 2 months of age prevented the progression of hyperactivity, slowed brain atrophy, and reduced brain hyperphosphorylated tau. In contrast, initiating doxycycline treatment at 4 months reduced neither brain hyperphosphorylated tau nor hyperactivity, further confirming the relationship between these measures. Collectively, our results demonstrate a unique behavioral phenotype in the rTg4510 mouse model of tauopathy that strongly correlates with disease progression, and that early interventions which reduce tau pathology ameliorate the progression of the locomotor dysfunction. These findings suggest that better understanding the relationship between locomotor deficits and tau pathology in the rTg4510 model may improve our understanding of the mechanisms underlying behavioral disturbances in patients with tauopathies.


Nature Genetics | 1996

LEPTIN RECEPTOR MISSENSE MUTATION IN THE FATTY ZUCKER RAT

Michael Sean Phillips; Qingyun Liu; Holly A. Hammond; Valarie Dugan; Patricia J. Hey; C. Thomas Caskey; J. Fred Hess

Collaboration


Dive into the J. Fred Hess's collaboration.

Top Co-Authors

Avatar

Xiaohai Wang

United States Military Academy

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge