Network


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

Hotspot


Dive into the research topics where James A. Clark is active.

Publication


Featured researches published by James A. Clark.


Endocrinology | 2000

Induction of Mammary Gland Development in Estrogen Receptor-α Knockout Mice

Wayne P. Bocchinfuso; Jonathan Lindzey; Sylvia C. Hewitt; James A. Clark; Page Myers; Ralph L. Cooper; Kenneth S. Korach

Mammary glands from the estrogen receptor-a knockout (alphaERKO) mouse do not undergo ductal morphogenesis or alveolar development. Disrupted ERalpha signaling may result in reduced estrogen-responsive gene products in the mammary gland or reduced mammotropic hormones that contribute to the alphaERKO mammary phenotype. We report that circulating PRL is reduced in the female alphaERKO mouse. Implantation of an age-matched, heterozygous ERalpha pituitary isograft under the renal capsule of 25-day-old or 12-week-old alphaERKO mice increased circulating PRL and progesterone levels, and induced mammary gland development. Grafted alphaERKO mice also possessed hypertrophied corpora lutea demonstrating that PRL is luteotropic in the alphaERKO ovary. By contrast, ovariectomy at the time of pituitary grafting prevented mammary gland development in alphaERKO mice despite elevated PRL levels. Hormone replacement using pellet implants demonstrated that pharmacological doses of estradiol induced limited mammary ductal elongation, and estradiol in combination with progesterone stimulated lobuloalveolar development. PRL alone or in combination with progesterone or estradiol did not induce alphaERKO mammary growth. Estradiol and progesterone are required for the structural development of the alphaERKO mammary gland, and PRL contributes to this development by inducing ovarian progesterone levels. Therefore, the manifestation of the alphaERKO mammary phenotype appears due to the lack of direct estrogen action at the mammary gland and an indirect contributory role of estrogen signaling at the hypothalamic/pituitary axis.


Endocrinology | 2002

Mammary Gland Development in Adult Mice Requires Epithelial and Stromal Estrogen Receptor α

Stefan O. Mueller; James A. Clark; Page Myers; Kenneth S. Korach

Complete mammary gland development takes place following puberty and depends on the estrogen receptor (ER)α and the progesterone receptor (PR) and is tightly regulated by the interaction of the mammary epithelium with the stromal compartment. Studies using mammary tissues of immature mice have indicated that stromal but not epithelial ERα is required for mammary gland growth. This study investigates whether these same tissue growth requirements of neonate tissue are necessary for mammary development and response in adult mice. Mammary epithelial cells were isolated from adult mice with a targeted disruption of the ERα gene (αERKO) or from wild-type counterparts and injected into epithelial-free mammary fat pads of 3-wk-old female αERKO or wild-type mice. Ten weeks after cell injection, analysis of mammary gland whole mounts showed that both stromal and epithelial ERα were required for complete mammary gland development in adult mice. However, when the mice were treated with high doses of estradiol (E2) an...


The FASEB Journal | 2010

Endothelial expression of human cytochrome P450 epoxygenases lowers blood pressure and attenuates hypertension-induced renal injury in mice

Craig R. Lee; John D. Imig; Matthew L. Edin; Julie F. Foley; Laura M. DeGraff; J. Alyce Bradbury; Joan P. Graves; Fred B. Lih; James A. Clark; Page Myers; A. Ligon Perrow; Adrienne Lepp; M. Alison Kannon; Oline K. Rønnekleiv; Nabil J. Alkayed; John R. Falck; Kenneth B. Tomer; Darryl C. Zeldin

Renal cytochrome P450 (CYP)‐derived epoxyeicosatrienoic acids (EETs) regulate sodium transport and blood pressure. Although endothelial CYP‐derived EETs are potent vasodilators, their contribution to the regulation of blood pressure remains unclear. Consequently, we developed transgenic mice with endothelial expression of the human CYP2J2 and CYP2C8 epoxygenases to increase endothelial EET biosynthesis. Compared to wild‐type littermate controls, an attenuated afferent arteriole constrictor response to endothelin‐1 and enhanced dilator response to acetylcholine was observed in CYP2J2 and CYP2C8 transgenic mice. CYP2J2 and CYP2C8 transgenic mice demonstrated modestly, but not significantly, lower mean arterial pressure under basal conditions compared to wild‐type controls. However, mean arterial pressure was significantly lower in both CYP2J2 and CYP2C8 transgenic mice during coadministration of N‐nitro‐l‐arginine methyl ester and indomethacin. In a separate experiment, a high‐salt diet and subcutaneous angiotensin II was administered over 4 wk. The angiotensin/high‐salt‐induced increase in systolic blood pressure, proteinuria, and glomerular injury was significantly attenuated in CYP2J2 and CYP2C8 transgenic mice compared to wild‐type controls. Collectively, these data demonstrate that increased endothelial CYP epoxygenase expression attenuates afferent arteriolar constrictor reactivity and hypertension‐induced increases in blood pressure and renal injury in mice. We conclude that endothelial CYP epoxygenase function contributes to the regulation of blood pressure.—Lee, C. R., Imig, J. D., Edin, M. E., Foley, J., DeGraff, L. M., Bradbury, J. A., Graves, J. P., Lih, F. B., Clark, J., Myers, P., Perrow, A. L., Lepp, A. N., Kannon, M. A., Ronnekleiv, O. K., Alkayed, N.J., Falck, J. R., Tomer, K B., Zeldin, D. C. Endothelial expression of human cytochrome P450 epoxygenases lowers blood pressure and attenuates hypertension‐induced renal injury in mice. FASEB J. 24, 3770–3781 (2010). www.fasebj.org


American Journal of Physiology-heart and Circulatory Physiology | 2008

The genetic contribution to heart rate and heart rate variability in quiescent mice

Reuben Howden; Eric Liu; Laura Miller-DeGraff; Heather L. Keener; Christopher Walker; James A. Clark; Page Myers; D. Clay Rouse; Tim Wiltshire; Steven R. Kleeberger

Recent studies have suggested a genetic component to heart rate (HR) and HR variability (HRV). However, a systematic examination of the genetic contribution to the variation in HR and HRV has not been performed. This study investigated the genetic contribution to HR and HRV using a wide range of inbred and recombinant inbred (RI) mouse strains. Electrocardiogram data were recorded from 30 strains of inbred mice and 29 RI strains. Significant differences in mean HR and total power (TP) HRV were identified between inbred strains and RI strains. Multiple significant differences within the strain sets in mean low-frequency (LF) and high-frequency (HF) power were also found. No statistically significant concordance was found between strain distribution patterns for HR and HRV phenotypes. Genomewide interval mapping identified a significant quantitative trait locus (QTL) for HR [LOD (likelihood of the odds) score = 3.763] on chromosome 6 [peak at 53.69 megabases (Mb); designated HR 1 (Hr1)]. Suggestive QTLs for TP were found on chromosomes 2, 4, 5, 6, and 14. A suggestive QTL for LF was found on chromosome 16; for HF, we found one significant QTL on chromosome 5 (LOD score = 3.107) [peak at 53.56 Mb; designated HRV-high-frequency 1 (Hrvhf1)] and three suggestive QTLs on chromosomes 2, 11 and 15. In conclusion, the results demonstrate a strong genetic component in the regulation of resting HR and HRV evidenced by the significant differences between strains. A lack of correlation between HR and HRV phenotypes in some inbred strains suggests that different sets of genes control the phenotypes. Furthermore, QTLs were found that will provide important insight to the genetic regulation of HR and HRV at rest.


Circulation Research | 2013

β-Arrestin-2 Deficiency Attenuates Abdominal Aortic Aneurysm Formation in Mice

Darshini B. Trivedi; Charles D. Loftin; James A. Clark; Page Myers; Laura M. DeGraff; Jennifer Cheng; Darryl C. Zeldin; Robert Langenbach

Rationale: Abdominal aortic aneurysms (AAAs) are a chronic inflammatory vascular disease for which pharmacological treatments are not available. A mouse model of AAA formation involves chronic infusion of angiotensin II (AngII), and previous studies indicated a primary role for the AngII type 1a receptor in AAA formation. &bgr;-arrestin (&bgr;arr)-2 is a multifunctional scaffolding protein that binds G-protein–coupled receptors such as AngII type 1a and regulates numerous signaling pathways and pathophysiological processes. However, a role for &bgr;arr2 in AngII-induced AAA formation is currently unknown. Objective: To determine whether &bgr;arr2 played a role in AngII-induced AAA formation in mice. Methods and Results: Treatment of &bgr;arr2+/+ and &bgr;arr2−/− mice on the hyperlipidemic apolipoprotein E–deficient (apoE−/−) background or on normolipidemic C57BL/6 background with AngII for 28 days indicated that &bgr;arr2 deficiency significantly attenuated AAA formation. &bgr;arr2 deficiency attenuated AngII-induced expression of cyclooxygenase-2, monocyte chemoattractant protein-1, macrophage inflammatory protein 1&agr;, and macrophage infiltration. AngII also increased the levels of phosphorylated extracellular signal-regulated kinase 1/2 in apoE−/−/&bgr;arr2+/+ aortas, whereas &bgr;arr2 deficiency diminished this increase. Furthermore, inhibition of extracellular signal-regulated kinase 1/2 activation with CI1040 (100 mg/kg per day) reduced the level of AngII-induced cyclooxygenase-2 expression in apoE−/−/&bgr;arr2+/+ mice to the level observed in apoE−/−/&bgr;arr2−/− mice. AngII treatment also increased matrix metalloproteinase expression and disruption of the elastic layer in apoE−/−/&bgr;arr2+/+ aortas, and &bgr;arr2 deficiency reduced these effects. Conclusions: &bgr;arr2 contributes to AngII-induced AAA formation in mice by phosphorylated extracellular signal-regulated kinase 1/2–mediated cyclooxygenase-2 induction and increased inflammation. These studies suggest that for the AngII type 1a receptor, G-protein–independent, &bgr;arr2-dependent signaling plays a major role in AngII-induced AAA formation.


American Journal of Respiratory Cell and Molecular Biology | 2012

Cardiac Physiologic and Genetic Predictors of Hyperoxia-Induced Acute Lung Injury in Mice

Reuben Howden; Hye-Youn Cho; Laura Miller-DeGraff; Christopher Walker; James A. Clark; Page Myers; D. Clay Rouse; Steven R. Kleeberger

Exposure of mice to hyperoxia produces pulmonary toxicity similar to acute lung injury/acute respiratory distress syndrome, but little is known about the interactions within the cardiopulmonary system. This study was designed to characterize the cardiopulmonary response to hyperoxia, and to identify candidate susceptibility genes in mice. Electrocardiogram and ventilatory data were recorded continuously from 4 inbred and 29 recombinant inbred strains during 96 hours of hyperoxia (100% oxygen). Genome-wide linkage analysis was performed in 27 recombinant inbred strains against response time indices (TIs) calculated from each cardiac phenotype. Reductions in minute ventilation, heart rate (HR), low-frequency (LF) HR variability (HRV), high-frequency HRV, and total power HRV were found in all mice during hyperoxia exposure, but the lag time before these changes began was strain dependent. Significant (chromosome 9) or suggestive (chromosomes 3 and 5) quantitative trait loci were identified for the HRTI and LFTI. Functional polymorphisms in several candidate susceptibility genes were identified within the quantitative trait loci and were associated with hyperoxia susceptibility. This is the first study to report highly significant interstrain variation in hyperoxia-induced changes in minute ventilation, HR, and HRV, and to identify polymorphisms in candidate susceptibility genes that associate with cardiac responses. Results indicate that changes in HR and LF HRV could be important predictors of subsequent adverse outcome during hyperoxia exposure, specifically the pathogenesis of acute lung injury. Understanding the genetic mechanisms of these responses may have significant diagnostic clinical value.


Clinical Cancer Research | 2011

Impact of ABCB1 allelic variants on QTc interval prolongation

Tristan M. Sissung; Erin R. Gardner; Richard Piekarz; Reuben Howden; Xiaohong Chen; Sukyung Woo; Ryan M. Franke; James A. Clark; Laura Miller-DeGraff; Seth M. Steinberg; David Venzon; David J. Liewehr; Steven R. Kleeberger; Susan E. Bates; Douglas K. Price; Douglas R. Rosing; Christopher H. Cabell; Alex Sparreboom; William D. Figg

Purpose: Although the ABCB1 (P-glycoprotein) drug transporter is a constituent of several blood–tissue barriers (i.e., blood–brain and blood–nerve), its participation in a putative blood–heart barrier has been poorly explored. ABCB1 could decrease the intracardiac concentrations of drugs that cause QT prolongation and cardiotoxicity. Experimental Design: ABCB1-related romidepsin transport kinetics were explored in LLC-PK1 cells transfected with different ABCB1 genetic variants. ABCB1 plasma and intracardiac concentrations were determined in Abcb1a/1b (−/−) mice and wild-type FVB controls. These same mice were used to evaluate romidepsin-induced heart rate-corrected QT interval (QTc) prolongation over time. Finally, a cohort of 83 individuals with available QTcB and ABCB1 genotyping data were used to compare allelic variation in ABCB1 versus QTc-prolongation phenotype. Results: Here, we show that mice lacking the ABCB1-type P-glycoprotein have higher intracardiac concentrations of a model ABCB1 substrate, romidepsin, that correspond to changes in QT prolongation from baseline (ΔQTc) over time. Consistent with this observation, we also show that patients carrying genetic variants that could raise ABCB1 expression in the cardiac endothelium have lower ΔQTc following a single dose of romidepsin. Conclusions: To our knowledge, this is the first evidence that Abcb1-type P-glycoprotein can limit intracardiac exposure to a drug that mediates QT prolongation and suggests that certain commonly inherited polymorphisms in ABCB1 may serve as markers for QT prolongation following the administration of ABCB1-substrate drugs. Clin Cancer Res; 17(4); 937–46. ©2010 AACR.


Xenobiotica | 1994

Toxicokinetics of pentachlorophenol in the F344 rat. Gavage and dosed feed studies

Jinhua Yuan; Thomas J. Goehl; Evelyn Murrill; R. Moore; James A. Clark; H. L. Hong; R. Irwin

1. The toxicokinetics of pentachlorophenol (PCP) were studied in the Fischer 344 rat using i.v. and oral (gavage, dosed feed) routes of exposure. 2. Only minor sex differences were observed in the elimination kinetics of PCP after i.v. administration at 5 mg/kg. 3. Absorption of PCP from the gastrointestinal tract after gavage doses of 9.5 and 38 mg/kg in aqueous methylcellulose vehicles was first order with an absorption half-life of about 1.3 h. 4. The absorption rate constant of PCP from doses feed was comparable with that obtained from aqueous methylcellulose gavage formulations. 5. Bioavailability of PCP administered in dosed feed was significantly lower than the bioavailability of PCP administered by gavage. 6. Dose proportionality was established to a dosage of at least 38 mg/kg. 7. Daily fluctuation of PCP plasma concentrations was observed during the dosed feed study with peak and trough concentrations occurring in early morning and late afternoon, respectively. 8. The time course of PCP plasma concentrations during the dosed feed study were simulated using a computer model based on linear theory. The simulations were comparable with the experimentally determined concentrations.


Journal of Toxicology and Environmental Health | 1997

Biliary elimination of oral 2,4-dichlorophenoxyacetic acid and its metabolites in male and female Sprague-Dawley rats, B6C3F1 mice, and Syrian hamsters

Robert J. Griffin; Jean Salemme; James A. Clark; Page Myers; Leo T. Burka

The role of biliary elimination in the metabolic disposition of 2,4-D was evaluated in male and female Sprague-Dawley rats, B6C3F1 mice, and Syrian hamsters. Following cannulation of the bile duct, an intragastric (ig) dose of 2,4-D (200 mg/kg) was administered and bile was collected at 30- or 60-min intervals for up to 6 h. Bile flow rates were constant in rats, increased in mice, and decreased in hamsters throughout the collection periods. Total recovery of radioactivity was greatest in male mice (about 7% of administered dose over 4 h). Female mice and rats of both sexes excreted about 3% over the same interval and male and female hamsters about 1%. About 71-88% of the activity in bile was parent compound. The glycine conjugate of 2,4-D was found in bile from mice, rats, and hamsters and the taurine conjugate in bile from mice. The only sex-dependent difference in the metabolite profile was in mice. Male mice excreted twice as much glycine conjugate as female mice. An additional minor metabolite (4-7%) was present in rat and mouse bile. This was tentatively identified as 2,4-D-glucuronide based on its hydrolysis by beta-glucuronidase. One more very minor metabolite (3%) was detected in rat bile but was not characterized due to its lability. The results of this study indicate that there are species-dependent differences in the biliary elimination of 2,4-D but not sex-dependent differences.


Xenobiotica | 1993

Toxicokinetics of pentachloroanisole in F344 rats and B6C3F1 mice

Jinhua Yuan; Thomas J. Goehl; Evelyn Murrill; R. Moore; James A. Clark; Lily H. Hong; R. Irwin

1. Toxicokinetics of pentachloroanisole (PCA) were studied in F344 rat and B6C3F1 mouse of both sexes by gavage at doses of 10, 20 and 40 mg/kg and by i.v. at 10 mg/kg. 2. PCA was rapidly demethylated to pentachlorophenol (PCP) in both rat and mouse and the resulting PCP plasma concentrations were much higher than that of parent PCA due to the much smaller apparent volume of distribution of PCP. 3. Peak plasma concentrations of PCA and PCP increased with dose in both rat and mouse. 4. Bioavailability of PCA was low in both rat and mouse and was sex independent. 5. The high plasma concentrations and relatively long biological half-life of PCP in both species after both i.v. and oral dosing with PCA indicate possible bioaccumulation of PCP upon multiple oral administrations of PCA.

Collaboration


Dive into the James A. Clark's collaboration.

Top Co-Authors

Avatar

Page Myers

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Reuben Howden

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Christopher Walker

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

D. Clay Rouse

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Kenneth S. Korach

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Laura Miller-DeGraff

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Steven R. Kleeberger

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Darryl C. Zeldin

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Darshini B. Trivedi

National Institutes of Health

View shared research outputs
Researchain Logo
Decentralizing Knowledge