Network


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

Hotspot


Dive into the research topics where Paul C. Simpson is active.

Publication


Featured researches published by Paul C. Simpson.


Journal of Clinical Investigation | 1983

Norepinephrine-stimulated hypertrophy of cultured rat myocardial cells is an alpha 1 adrenergic response.

Paul C. Simpson

We have shown recently that norepinephrine stimulates muscle cell hypertrophy in primary cultures from the neonatal rat ventricle and that this stimulation is not blocked by the beta adrenergic antagonist propranolol. The present study was done to define the adrenergic specificity of the myocyte hypertrophic response to norepinephrine. 90% pure, single-cell cultures of nongrowing myocytes were maintained in serum-free medium 199 with transferin and insulin. Myocyte size was quantitated 48 h after addition of adrenergic agents, by measuring cell volume, cell surface area, and cell protein. L-norepinephrine increased myocyte size to a maximum 150% of control; half-maximum effect was obtained at a concentration of 0.2 microM. This increase in cell size was inhibited by the nonselective alpha adrenergic antagonist phentolamine and by the alpha 1 adrenergic antagonists prazosin and terazosin; it was not inhibited by propranolol or by the alpha 2 adrenergic antagonist yohimbine. The beta adrenergic agonist isoproterenol did not increase cell size. Thus, norepinephrine-stimulated hypertrophy of cultured rat myocardial cells is an alpha 1 adrenergic response.


Methods of Molecular Biology | 2007

Isolation and Culture of Adult Mouse Cardiac Myocytes

Timothy D. O'Connell; Manoj C. Rodrigo; Paul C. Simpson

Cardiac myocytes are activated by hormonal and mechanical signals and respond in a variety of ways, from altering contractile function to inducing cardio-protection and growth responses. The use of genetic mouse models allows one to examine the role of cardiac-specific and other genes in cardiac function, hypertrophy, cardio-protection, and diseases such as ischemia and heart failure. However, studies at the cellular level have been hampered by a lack of suitable techniques for isolating and culturing calcium-tolerant, adult mouse cardiac myocytes. We have developed a straightforward, reproducible protocol for isolating and culturing large numbers of adult mouse cardiac myocytes. This protocol is based on the traditional approach of retrograde perfusion of collagenase through the coronary arteries to digest the extracellular matrix of the heart and release rod-shaped myocytes. However, we have made modifications that are essential for isolating calcium-tolerant, rod-shaped adult mouse cardiac myocytes and maintaining them in culture. This protocol yields freshly isolated adult mouse myocytes that are suitable for biochemical assays and for measuring contractile function and calcium transients, and cultured myocytes that are suitable for most biochemical and signaling assays, as well as gene transduction using adenovirus.


Journal of Biological Chemistry | 1995

M-CAT, CArG, and Sp1 Elements Are Required for -Adrenergic Induction of the Skeletal -Actin Promoter during Cardiac Myocyte Hypertrophy TRANSCRIPTIONAL ENHANCER FACTOR-1 AND PROTEIN KINASE C AS CONSERVED TRANSDUCERS OF THE FETAL PROGRAM IN CARDIAC GROWTH

Larry R. Karns; Ken-ichi Kariya; Paul C. Simpson

Induction of the fetal isogenes skeletal α-actin (skACT) and β-myosin heavy chain (β-MHC) is characteristic of cardiac growth in many models, suggesting a conserved signaling pathway. However, divergent regulation has also been observed. β-Protein kinase C (PKC) and transcriptional enhancer factor-1 (TEF-1) are involved in induction of β-MHC in α1-adrenergic-stimulated hypertrophy of cultured cardiac myocytes (Kariya, K., Farrance, I. K. G., and Simpson, P. C.(1993) J. Biol. Chem. 268, 26658-26662; Kariya, K., Karns, L. R., and Simpson, P. C.(1994) J. Biol. Chem. 269, 3775-3782). In the present study, we asked whether the skACT promoter used the same mechanism. A mouse skACT promoter fragment (−113/−46) was induced by both α1-adrenergic stimulation and co-transfection of activated β-PKC, and contained three required DNA sequence elements: M-CAT, CArG, and Sp1. The skACT M-CAT element bound TEF-1 in cardiac myocytes. Thus the skACT and β-MHC promoters both require a TEF-1 binding site for activation by α1-adrenergic stimulation, but differ in that skACT also requires a CArG box. These results provide a potential molecular basis for divergent regulation of the fetal program, and also imply that PKC and TEF-1 are conserved transducers for this program during cardiac growth.


Proceedings of the National Academy of Sciences of the United States of America | 2002

Knockout of the alpha 1A/C-adrenergic receptor subtype: the alpha 1A/C is expressed in resistance arteries and is required to maintain arterial blood pressure.

D. Gregg Rokosh; Paul C. Simpson

α1-adrenergic receptors (ARs) play a major role in blood pressure regulation. The three α1-AR subtypes (A/C, B, and D) stimulate contraction of isolated arteries, but it is uncertain how different subtypes contribute to blood pressure regulation in the intact animal. We studied the role of the α1A/C subtype by using gene knockout. α1A/C knockout (KO) mice were viable and overtly normal. The LacZ reporter gene replaced α1A/C coding sequence in the KO, and β-galactosidase staining was present in resistance arteries and arterioles, but not in the thoracic aorta or its main branches. By tail cuff manometer and arterial catheter in conscious mice, α1A/C KO mice were hypotensive at rest, with an 8–12% reduction of blood pressure dependent on α1A/C gene copy number. A61603, an α1A/C-selective agonist, caused a pressor response that was lost in the KO and reduced but significant in heterozygous mice with a single copy of the α1A/C. A subtype-nonselective agonist [phenylephrine (PE)] caused a pressor response in KO mice, but the final arterial pressure was only 85% of wild type. The baroreflex was reset in the KO, and heart rate variability was decreased. After baroreflex blockade with atropine, PE increased blood pressure but did not change heart rate. Cardiac and vascular responses to the β-AR agonist isoproterenol were unchanged, and the arterial lumen area was not altered. We conclude that the α1A/C-AR subtype is a vasopressor expressed in resistance arteries and is required for normal arterial blood pressure regulation. α1A/C-selective antagonists might be desirable antihypertensive agents.


Nature Genetics | 2012

Large-scale discovery of enhancers from human heart tissue

Dalit May; Matthew J. Blow; Tommy Kaplan; David J. McCulley; Brian C. Jensen; Jennifer A. Akiyama; Amy Holt; Ingrid Plajzer-Frick; Malak Shoukry; Crystal Wright; Veena Afzal; Paul C. Simpson; Edward M. Rubin; Brian L. Black; James Bristow; Len A. Pennacchio; Axel Visel

Development and function of the human heart depend on the dynamic control of tissue-specific gene expression by distant-acting transcriptional enhancers. To generate an accurate genome-wide map of human heart enhancers, we used an epigenomic enhancer discovery approach and identified ∼6,200 candidate enhancer sequences directly from fetal and adult human heart tissue. Consistent with their predicted function, these elements were markedly enriched near genes implicated in heart development, function and disease. To further validate their in vivo enhancer activity, we tested 65 of these human sequences in a transgenic mouse enhancer assay and observed that 43 (66%) drove reproducible reporter gene expression in the heart. These results support the discovery of a genome-wide set of noncoding sequences highly enriched in human heart enhancers that is likely to facilitate downstream studies of the role of enhancers in development and pathological conditions of the heart.


Journal of Clinical Investigation | 2010

Limited forward trafficking of connexin 43 reduces cell-cell coupling in stressed human and mouse myocardium

James W. Smyth; TingTing Hong; Danchen Gao; Jacob M. Vogan; Brian C. Jensen; Tina S. Fong; Paul C. Simpson; Didier Y. R. Stainier; Neil C. Chi; Robin M. Shaw

Gap junctions form electrical conduits between adjacent myocardial cells, permitting rapid spatial passage of the excitation current essential to each heartbeat. Arrhythmogenic decreases in gap junction coupling are a characteristic of stressed, failing, and aging myocardium, but the mechanisms of decreased coupling are poorly understood. We previously found that microtubules bearing gap junction hemichannels (connexons) can deliver their cargo directly to adherens junctions. The specificity of this delivery requires the microtubule plus-end tracking protein EB1. We performed this study to investigate the hypothesis that the oxidative stress that accompanies acute and chronic ischemic disease perturbs connexon forward trafficking. We found that EB1 was displaced in ischemic human hearts, stressed mouse hearts, and isolated cells subjected to oxidative stress. As a result, we observed limited microtubule interaction with adherens junctions at intercalated discs and reduced connexon delivery and gap junction coupling. A point mutation within the tubulin-binding domain of EB1 reproduced EB1 displacement and diminished connexon delivery, confirming that EB1 displacement can limit gap junction coupling. In zebrafish hearts, oxidative stress also reduced the membrane localization of connexin and slowed the spatial spread of excitation. We anticipate that protecting the microtubule-based forward delivery apparatus of connexons could improve cell-cell coupling and reduce ischemia-related cardiac arrhythmias.


Journal of Clinical Investigation | 1990

The cardiac beta-myosin heavy chain isogene is induced selectively in alpha 1-adrenergic receptor-stimulated hypertrophy of cultured rat heart myocytes.

L E Waspe; Charles P. Ordahl; Paul C. Simpson

Cardiac hypertrophy produced in vivo by pressure overload is characterized by selective up-regulation of the fetal/neonatal beta-cardiac myosin heavy chain (MHC) isogene. However, a molecular signal for beta-MHC isogene induction has not been identified. We examined cardiac MHC isogene expression in a cell culture model for hypertrophy. alpha-MHC and beta-MHC iso-protein and iso-mRNA levels in cultured cardiac myocytes were quantified during hypertrophy stimulated by the alpha 1-adrenergic agonist, norepinephrine (NE). beta-MHC iso-protein content was increased 3.2-fold vs. control (P less than 0.001), whereas alpha-MHC isoprotein content was not changed significantly (1.4-fold vs. control, P = NS). MHC iso-mRNA levels were quantified by nuclease S1 analysis, using a single oligonucleotide probe. NE increased beta-MHC iso-mRNA content by 3.9-fold vs. control (P less than 0.001), but there was no change in alpha-MHC iso-mRNA (1.1-fold vs. control, P = NS). The NE-stimulated increase in beta-MHC iso-mRNA preceded in time the increase in beta-MHC isoprotein accumulation. The EC50 for NE induction of beta-MHC was 40 nM, and pharmacologic experiments indicated alpha 1-adrenergic receptor specificity. alpha-MHC isogene expression was predominant in control myocytes (68% alpha-isoprotein and 60% alpha-iso-mRNA). In contrast, beta-MHC expression was equal to alpha-MHC or predominant after treatment with NE (51% beta-isoprotein and 69% beta-iso-mRNA). Thus, alpha 1-adrenergic receptor stimulation increases the cellular contents of beta-MHC iso-mRNA and beta-MHC isoprotein during hypertrophy of cultured neonatal rat cardiac myocytes, but does not change the levels of alpha-MHC iso-mRNA or isoprotein. The effect on beta-MHC is mediated primarily at the level of mRNA steady-state level (pretranslational). Activation of the alpha 1-adrenergic receptor is the first identified molecular signal for increased beta-MHC isogene expression in a model of cardiac hypertrophy.


Journal of Clinical Investigation | 1987

Induction of the skeletal alpha-actin gene in alpha 1-adrenoceptor-mediated hypertrophy of rat cardiac myocytes.

Nanette H. Bishopric; Paul C. Simpson; Charles P. Ordahl

Myocardial hypertrophy in vivo is associated with reexpression of contractile protein isogenes characteristic of fetal and neonatal development. The molecular signals for hypertrophy and isogene switching are unknown. We studied alpha (sarcomeric)-actin messenger RNA (mRNA) expression in cultured cardiac myocytes from the neonatal rat. In the cultured cells, as in the adult heart in vivo, expression of cardiac alpha-actin (cACT) predominated over that of skeletal alpha-actin (sACT) mRNA, the fetal/neonatal isoform. alpha 1-Adrenergic receptor stimulation induced hypertrophy of these cells, increasing total RNA and cytoskeletal actin mRNA by 1.8-fold over control, and total alpha-actin mRNA by 4.3 fold. This disproportionate increase in total alpha-actin mRNA was produced by a preferential induction of sACT mRNA, which increased by 10.6-fold over control versus only 2.6-fold for cACT mRNA. The alpha 1-adrenoceptor is the first identified molecular mediator of early developmental isogene reexpression in cardiac myocyte hypertrophy.


Journal of Molecular and Cellular Cardiology | 1988

Differential acute and chronic response of protein kinase C in cultured neonatal rat heart myocytes to α1-adrenergic and phorbol ester stimulation ☆

Curtis J. Henrich; Paul C. Simpson

Both alpha 1-adrenergic agonists (e.g. norepinephrine, NE*) and tumor-promoting phorbol esters (e.g. phorbol myristate acetate, PMA) are known to activate protein kinase C (PKC) (Abdel-Latif, 1986, Niedel and Blackshear, 1986). However, alpha 1 agonists and PMA produce very different effects on cardiac function (see Simpson, 1985; Benfey, 1987; Meidell et al., 1986; Leatherman et al., 1987; Yuan et al., 1987; for examples). PKC activation in heart cells has been studied only for PMA treated perfused heart (Yuan et al., 1987). Therefore, acute activation and chronic regulation of PKC by NE and PMA were compared in cultured neonatal rat heart myocytes. NE acutely and transiently activated PKC, as measured by translocation of PKC activity to the cell particulate fraction (Niedel and Blackshear, 1986). Particulate PKC activity peaked at 23% of total after NE for 30 s, as compared with 8% for control (P less than 0.001). By contrast, acute PKC activation by PMA was more pronounced and persistent, with particulate PKC activity 62% of total at 5 min (P less than 0.001). Calcium/lipid-independent kinase activity increased acutely with PMA, but not with NE. Chronic treatment with NE (24 to 48 h) increased total per cell PKC activity and 3H-phorbol dibutyrate (PDB) binding sites, an index of the number of PKC molecules (Niedel and Blackshear, 1986), by 30 to 60% over control (all P less than 0.05 to 0.01). In contrast with NE, chronic treatment with PMA down-regulated PKC, reducing total per cell PKC activity and 3H-PDB binding sites to 3% and 12% of control, respectively (P less than 0.01).(ABSTRACT TRUNCATED AT 250 WORDS)


Journal of Biological Chemistry | 1996

-Adrenergic Receptor Subtype mRNAs Are Differentially Regulated by -Adrenergic and Other Hypertrophic Stimuli in Cardiac Myocytes in Culture and In Vivo REPRESSION OF α AND α BUT INDUCTION OF α

D. Gregg Rokosh; Alexandre F. R. Stewart; K. C. Chang; Beth A. Bailey; Joel S. Karliner; S. Albert Camacho; Carlin S. Long; Paul C. Simpson

The three cloned α-adrenergic receptor (AR) subtypes, α, α, and α, can all couple to the same effector, phospholipase C, and the reason(s) for conservation of multiple subtypes remain uncertain. All three α-ARs are expressed natively in cultured neonatal rat cardiac myocytes, where chronic exposure to the agonist catecholamine norepinephrine (NE) induces hypertrophic growth and gene transcription. We show here, using RNase protection, that the α-AR subtype mRNAs respond in distinctly different ways during prolonged NE exposure (12-72 h). α and α mRNA levels were repressed by NE, whereas α mRNA was induced. Changes in mRNA levels were mediated by an α-AR, were not explained by altered mRNA stability, and were reflected in receptor proteins by [3H]prazosin binding. α-AR-stimulated phosphoinositide hydrolysis and myocyte growth were not desensitized. Three other hypertrophic agonists in culture, endothelin-1, PGF2α, and phorbol 12-myristate 13-acetate, also induced α mRNA and repressed α mRNA. In myocytes from hearts with pressure overload hypertrophy, α mRNA changes were identical to those produced by NE in culture. These results provide the first example of a difference in regulation among α-AR subtypes expressed natively in the same cell. Transcriptional induction of the α-AR could be a mechanism for sustained growth signaling through this receptor and is a common feature of a hypertrophic phenotype in cardiac myocytes.

Collaboration


Dive into the Paul C. Simpson's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Brian C. Jensen

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Carlin S. Long

University of Colorado Denver

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

K.-I. Kariya

United States Department of Veterans Affairs

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge