Network


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

Hotspot


Dive into the research topics where Diane Gesty-Palmer is active.

Publication


Featured researches published by Diane Gesty-Palmer.


Journal of Biological Chemistry | 2006

Distinct β-Arrestin- and G Protein-dependent Pathways for Parathyroid Hormone Receptor-stimulated ERK1/2 Activation

Diane Gesty-Palmer; Minyong Chen; Eric Reiter; Seungkirl Ahn; Christopher D. Nelson; Shuntai Wang; Allen E. Eckhardt; Conrad L. Cowan; Robert F. Spurney; Louis M. Luttrell; Robert J. Lefkowitz

Parathyroid hormone (PTH) regulates calcium homeostasis via the type I PTH/PTH-related peptide (PTH/PTHrP) receptor (PTH1R). The purpose of the present study was to identify the contributions of distinct signaling mechanisms to PTH-stimulated activation of the mitogen-activated protein kinases (MAPK) ERK1/2. In Human embryonic kidney 293 (HEK293) cells transiently transfected with hPTH1R, PTH stimulated a robust increase in ERK activity. The time course of ERK1/2 activation was biphasic with an early peak at 10 min and a later sustained ERK1/2 activation persisting for greater than 60 min. Pretreatment of HEK293 cells with the PKA inhibitor H89 or the PKC inhibitor GF109203X, individually or in combination reduced the early component of PTH-stimulated ERK activity. However, these inhibitors of second messenger dependent kinases had little effect on the later phase of PTH-stimulated ERK1/2 phosphorylation. This later phase of ERK1/2 activation at 30–60 min was blocked by depletion of cellular β-arrestin 2 and β-arrestin 1 by small interfering RNA. Furthermore, stimulation of hPTH1R with PTH analogues, [Trp1]PTHrp-(1–36) and [d-Trp12,Tyr34]PTH-(7–34), selectively activated Gs/PKA-mediated ERK1/2 activation or G protein-independent/β-arrestin-dependent ERK1/2 activation, respectively. It is concluded that PTH stimulates ERK1/2 through several distinct signal transduction pathways: an early G protein-dependent pathway meditated by PKA and PKC and a late pathway independent of G proteins mediated through β-arrestins. These findings imply the existence of distinct active conformations of the hPTH1R responsible for the two pathways, which can be stimulated by unique ligands. Such ligands may have distinct and valuable therapeutic properties.


Pharmacological Reviews | 2010

Beyond Desensitization: Physiological Relevance of Arrestin-Dependent Signaling

Louis M. Luttrell; Diane Gesty-Palmer

Heptahelical G protein-coupled receptors are the most diverse and therapeutically important family of receptors in the human genome. Ligand binding activates heterotrimeric G proteins that transmit intracellular signals by regulating effector enzymes or ion channels. G protein signaling is terminated, in large part, by arrestin binding, which uncouples the receptor and G protein and targets the receptor for internalization. It is clear, however, that heptahelical receptor signaling does not end with desensitization. Arrestins bind a host of catalytically active proteins and serve as ligand-regulated scaffolds that recruit protein and lipid kinase, phosphatase, phosphodiesterase, and ubiquitin ligase activity into the receptor-arrestin complex. Although many of these arrestin-bound effectors serve to modulate G protein signaling, degrading second messengers and regulating endocytosis and trafficking, other signals seem to extend beyond the receptor-arrestin complex to regulate such processes as protein translation and gene transcription. Although these findings have led to a re-envisioning of heptahelical receptor signaling, little is known about the physiological roles of arrestin-dependent signaling. In vivo, the duality of arrestin function makes it difficult to dissociate the consequences of arrestin-dependent desensitization from those that might be ascribed to arrestin-mediated signaling. Nonetheless, recent evidence generated using arrestin knockouts, G protein-uncoupled receptor mutants, and arrestin pathway-selective “biased agonists” is beginning to reveal that arrestin signaling plays important roles in the retina, central nervous system, cardiovascular system, bone remodeling, immune system, and cancer. Understanding the signaling roles of arrestins may foster the development of pathway-selective drugs that exploit these pathways for therapeutic benefit.


Proceedings of the National Academy of Sciences of the United States of America | 2008

Distinct conformational changes in β-arrestin report biased agonism at seven-transmembrane receptors

Arun K. Shukla; Jonathan D. Violin; Erin J. Whalen; Diane Gesty-Palmer; Sudha K. Shenoy; Robert J. Lefkowitz

β-arrestins critically regulate G protein-coupled receptors (GPCRs), also known as seven-transmembrane receptors (7TMRs), both by inhibiting classical G protein signaling and by initiating distinct β-arrestin-mediated signaling. The recent discovery of β-arrestin-biased ligands and receptor mutants has allowed characterization of these independent “G protein-mediated” and “β-arrestin-mediated” signaling mechanisms of 7TMRs. However, the molecular mechanisms underlying the dual functions of β-arrestins remain unclear. Here, using an intramolecular BRET (bioluminescence resonance energy transfer)-based biosensor of β-arrestin 2 and a combination of biased ligands and/or biased mutants of three different 7TMRs, we provide evidence that β-arrestin can adopt multiple “active” conformations. Surprisingly, phosphorylation-deficient mutants of the receptors are also capable of directing similar conformational changes in β-arrestin as is the wild-type receptor. This indicates that distinct receptor conformations induced and/or stabilized by different ligands can promote distinct and functionally specific conformations in β-arrestin even in the absence of receptor phosphorylation. Our data thus highlight another interesting aspect of 7TMR signaling—i.e., functionally specific receptor conformations can be translated to downstream effectors such as β-arrestins, thereby governing their functional specificity.


Science Translational Medicine | 2009

A β-Arrestin–Biased Agonist of the Parathyroid Hormone Receptor (PTH1R) Promotes Bone Formation Independent of G Protein Activation

Diane Gesty-Palmer; Pat Flannery; Ling Yuan; Leonor Corsino; Robert F. Spurney; Robert J. Lefkowitz; Louis M. Luttrell

A biased agonist binds to the seven-transmembrane parathyroid hormone receptor to trigger certain downstream effects but not others, potentially avoiding undesirable side effects. Building Better Bones What do β-blockers, antihistamines, and opiate pain drugs have in common? All act on constituents of the ~1000-member family of seven-transmembrane receptors (7TMRs). Arrayed on the surfaces of all cells in the body, these diverse proteins regulate myriad critical functions, including heart beat, immune response, secretion, and stomach acid production. Almost half of today’s prescribed drugs act to modulate the action of members of this huge family of receptors. When bound by their natural trigger—or a drug—these receptors activate two downstream cellular pathways: one mediated by G proteins and the other, discovered in the last 10 years, mediated by a small protein called β-arrestin, which can both terminate G protein–dependent signaling and activate other cellular processes. Finding the β-arrestin pathway raised the possibility that one could discover drugs that modulate the two signaling pathways independently and so produce better outcomes for patients, with fewer side effects. Gesty-Palmer et al. now show that a so-called biased agonist, PTH-βarr, selectively activates the β-arrestin (but not the G protein) pathway in mouse bone cells by binding to the 7TMR for parathyroid hormone (PTH), a key regulator of bone growth. By stimulating only the β-arrestin arm of the PTH-activated responses, PTH-βarr leads to bone growth in living mice without simultaneous bone degradation, which usually occurs through the G protein–linked pathway. Bone is remodeled throughout life, being continuously broken down by one type of bone cell (osteoclasts) and then rebuilt by another (osteoblasts). One regulator of the balance between the two processes is PTH. When this normal equilibrium goes awry, such that there is more bone destruction than production, bones become structurally weak, and bone diseases such as osteoporosis result. Patients with osteoporosis often are treated with bisphosphonate drugs, which prevent bone loss by binding to the calcium hydroxyapatite in bone and inhibiting the bone-degrading osteoclasts. Although bisphosphonates decrease the risk of fracture, the microarchitecture of bisphosphonate-induced bone is not normal, and long-term use can lead to adynamic bone fractures. Better bone microstructure can be built by daily injections of a fragment of PTH, which directly stimulates osteoblasts, but the PTH fragment also indirectly stimulates osteoclasts and must be injected daily to prevent the osteoclast-driven breakdown from reversing the induced bone growth. Because its effects are tilted more toward anabolic bone growth without simultaneous catabolism, PTH-βarr or similar biased agonists may prove to be superior treatments for osteoporosis. The results in Gesty-Palmer et al. illustrate that, by activating exclusively the β-arrestin pathway downstream of the PTH receptor, a biased agonist can stimulate a desirable physiological outcome (bone growth) while minimizing an undesirable effect (bone degradation). The benefits of biased agonists with differential signaling properties may extend to any therapeutic agent that acts through a 7TMR. About 40% of the therapeutic agents in use today exert their effects through seven-transmembrane receptors (7TMRs). When activated by ligands, these receptors trigger two pathways that independently transduce signals to the cell: one through heterotrimeric GTP-binding proteins (G proteins) and one through β-arrestins; so-called biased agonists can selectively activate these distinct pathways. Here, we investigate selective activation of these pathways through the use of a biased agonist for the type 1 parathyroid hormone (PTH)–PTH-related protein receptor (PTH1R), (d-Trp12,Tyr34)-PTH(7–34) (PTH-βarr), which activates β-arrestin but not classic G protein signaling. In mice, PTH-βarr induces anabolic bone formation, as does the nonselective agonist PTH(1–34), which activates both mechanisms. In β-arrestin2–null mice, the increase in bone mineral density evoked by PTH(1–34) is attenuated and that stimulated by PTH-βarr is ablated. The β-arrestin2–dependent pathway contributes primarily to trabecular bone formation and does not stimulate bone resorption. These results show that a biased agonist selective for the β-arrestin pathway can elicit a response in vivo distinct from that elicited by nonselective agonists. Ligands with these properties may form the basis for improved 7TMR-directed pharmacologic agents with enhanced therapeutic specificity.


The Journal of Infectious Diseases | 1999

Recurrent Staphylococcus auveus Bacteremia: Pulsed-Field Gel Electrophoresis Findings in 29 Patients

Vance G. Fowler; Li Kuo Kong; G. Ralph Corey; Geoffrey S. Gottlieb; R. Scott McClelland; Daniel J. Sexton; Diane Gesty-Palmer; Lizzie J. Harrell

To identify risk factors for relapse among 309 prospectively identified cases of Staphylococcus aureus bacteremia, patients with recurrent S. aureus bacteremia were identified, and pulsed-field gel electrophoresis (PFGE) was performed on isolates from both episodes. PFGE banding patterns from both isolates were identical in 23 patients, consistent with relapsed infection. Patients with PFGE-confirmed relapse were more likely by both univariate and multivariate analyses to have an indwelling foreign body (odds ratio [OR]=18.2, 95% confidence interval [CI]=7. 6-43.6; P<.001), to have received vancomycin therapy (OR=4.1, 95% CI=1.5-11.6; P=.008), or be hemodialysis-dependent (OR=4.1, 95% CI=1. 8-9.3; P=.002) than patients who did not develop recurrent bacteremia. These results suggest that recurrent episodes of S. aureus bacteremia are primarily relapses and are associated with an indwelling foreign body, receiving vancomycin therapy, and hemodialysis dependence.


Endocrinology | 2010

The Endogenous Selective Estrogen Receptor Modulator 27-Hydroxycholesterol Is a Negative Regulator of Bone Homeostasis

Carolyn D. DuSell; Erik R. Nelson; Xiaojuan Wang; Jennifer Abdo; Ulrike I. Mödder; Michihisa Umetani; Diane Gesty-Palmer; Norman B. Javitt; Sundeep Khosla; Donald P. McDonnell

Osteoporosis is an important clinical problem, affecting more than 50% of people over age 50 yr. Estrogen signaling is critical for maintaining proper bone density, and the identification of an endogenous selective estrogen receptor (ER) modulator, 27-hydroxycholesterol (27HC), suggests a mechanism by which nutritional/metabolic status can influence bone biology. With its levels directly correlated with cholesterol, a new possibility emerges wherein 27HC links estrogen and cholesterol signaling to bone homeostasis. In these studies, we found that increasing concentrations of 27HC, both by genetic and pharmacological means, led to decreased bone mineral density that was associated with decreased bone formation and increased bone resorption. Upon manipulation of endogenous estrogen levels, many of the responses to elevated 27HC were altered in such a way as to implicate ER as a likely mediator. In a model of postmenopausal bone loss, some pathologies associated with elevated 27HC were exacerbated by the absence of endogenous estrogens, suggesting that 27HC may act both in concert with and independently from classic ER signaling. These data provide evidence for interactions between estrogen signaling, cholesterol and metabolic disease, and osteoporosis. Patients with high cholesterol likely also have higher than average 27HC, perhaps putting them at a higher risk for bone loss and fracture. More studies are warranted to fully elucidate the mechanism of action of 27HC in bone and to identify ways to modulate this pathway therapeutically.


Endocrinology | 2011

The oxysterol, 27-hydroxycholesterol, links cholesterol metabolism to bone homeostasis through its actions on the estrogen and liver X receptors.

Erik R. Nelson; Carolyn D. DuSell; Xiaojuan Wang; Matthew K. Howe; Glenda L. Evans; Ryan D. Michalek; Michihisa Umetani; Jeffrey C. Rathmell; Sundeep Khosla; Diane Gesty-Palmer; Donald P. McDonnell

Osteoporosis and age-related bone loss are important public health concerns. Therefore, there is a high level of interest in the development of medical interventions and lifestyle changes that reduce the incidence of osteoporosis and age-related bone loss. Decreased bone mineral density is associated with high cholesterol, and patients on statins have increased bone mineral densities, strongly implicating cholesterol as a negative regulator of bone homeostasis. In this study, using both molecular and pharmacological approaches, we have been able to demonstrate that the primary cholesterol metabolite, 27-hydroxycholesterol, through its actions on both estrogen receptors and liver X receptors, decreases osteoblast differentiation and enhances osteoclastogenesis, resulting in increased bone resorbtion in mice. Induction of the short heterodimer partner protein by estrogens in osteoblasts can attenuate the liver X receptor-mediated actions of 27-hydroxycholesterol in bone. These data establish a mechanistic link between cholesterol and bone quality, highlight an unexpected target of estrogens in osteoblasts, and define a signaling axis, the therapeutic exploitation of which is likely to yield novel antiosteoporotic drugs.


Journal of Clinical Oncology | 2000

Prospective Analysis of Staphylococcus aureus Bacteremia in Nonneutropenic Adults With Malignancy

Ajay K. Gopal; Vance G. Fowler; Manish A. Shah; Diane Gesty-Palmer; Kieren A. Marr; R. Scott McClelland; Li Kuo Kong; Geoffrey S. Gottlieb; Lanclos K; Jennifer S. Li; Daniel J. Sexton; G. Ralph Corey

PURPOSE To determine the primary sources and secondary complications of Staphylococcus aureus bacteremia (SAB) in cancer patients, as well as predictors of outcome in cancer patients with SAB. PATIENTS AND METHODS Fifty-two patients at Duke University Medical Center met entry criteria between September 1994 and December 1996 for this prospective cohort study involving hospitalized nonneutropenic adult cancer patients with SAB. All subjects were observed throughout initial hospitalization and were evaluated again at 6 and 12 weeks or until death. RESULTS SAB was intravascular device-related in 42%, tissue infection-related (TIR) in 44%, and unidentifiable focus-related (UFR) in 13%. Seventeen patients (33%) were found to have metastatic infections or conditions, with eight (15%) developing infectious endocarditis (IE). Patients with TIR bacteremia were less likely than other patients to develop IE (4% v 24%, P =.06). The overall mortality rate was 38%, the SAB-related mortality rate was 15%, and the rate of SAB relapse was 12%. Methicillin resistance was not associated with adverse outcome. Inability to identify a point of entry (UFR bacteremia), however, was associated with a higher overall mortality rate (100% v 24%, P =.0006). Furthermore, a 72-hour surveillance blood culture positive for organisms was associated with an increased incidence of IE (P =.0006), metastatic infections or conditions (P =.0002), SAB relapse (P =.038), and SAB-related death (P =.038). CONCLUSION SAB in cancer patients is associated with significant morbidity from frequent metastatic infections or conditions including IE, as well as considerable mortality. Unknown initial infection site and 72-hour surveillance cultures positive for organisms were predictive of a complicated course and poor final outcome.


Molecular Endocrinology | 2013

β-Arrestin-Selective G Protein-Coupled Receptor Agonists Engender Unique Biological Efficacy in Vivo

Diane Gesty-Palmer; Ling Yuan; Bronwen Martin; William H. Wood; Mi Hye Lee; Michael G. Janech; Lam C. Tsoi; W. Jim Zheng; Louis M. Luttrell; Stuart Maudsley

Biased G protein-coupled receptor agonists are orthosteric ligands that possess pathway-selective efficacy, activating or inhibiting only a subset of the signaling repertoire of their cognate receptors. In vitro, D-Trp(12),Tyr(34)-bPTH(7-34) [bPTH(7-34)], a biased agonist for the type 1 PTH receptor, antagonizes receptor-G protein coupling but activates arrestin-dependent signaling. In vivo, both bPTH(7-34) and the conventional agonist hPTH(1-34) stimulate anabolic bone formation. To understand how two PTH receptor ligands with markedly different in vitro efficacy could elicit similar in vivo responses, we analyzed transcriptional profiles from calvarial bone of mice treated for 8 wk with vehicle, bPTH(7-34) or hPTH(1-34). Treatment of wild-type mice with bPTH(7-34) primarily affected pathways that promote expansion of the osteoblast pool, notably cell cycle regulation, cell survival, and migration. These responses were absent in β-arrestin2-null mice, identifying them as downstream targets of β-arrestin2-mediated signaling. In contrast, hPTH(1-34) primarily affected pathways classically associated with enhanced bone formation, including collagen synthesis and matrix mineralization. hPTH(1-34) actions were less dependent on β-arrestin2, as might be expected of a ligand capable of G protein activation. In vitro, bPTH(7-34) slowed the rate of preosteoblast proliferation, enhanced osteoblast survival when exposed to an apoptotic stimulus, and stimulated cell migration in wild-type, but not β-arrestin2-null, calvarial osteoblasts. These results suggest that bPTH(7-34) and hPTH(1-34) affect bone mass in vivo through predominantly separate genomic mechanisms created by largely distinct receptor-signaling networks and demonstrate that functional selectivity can be exploited to change the quality of G protein-coupled receptor efficacy.


PLOS ONE | 2010

Growth Hormone Mitigates against Lethal Irradiation and Enhances Hematologic and Immune Recovery in Mice and Nonhuman Primates

Benny J. Chen; Divino Deoliveira; Ivan Spasojevic; Gregory D. Sempowski; Chen Jiang; Kouros Owzar; Xiaojuan Wang; Diane Gesty-Palmer; J. Mark Cline; J. Daniel Bourland; Greg Dugan; Sarah K. Meadows; Pamela Daher; Garrett G. Muramoto; John P. Chute; Nelson J. Chao

Medications that can mitigate against radiation injury are limited. In this study, we investigated the ability of recombinant human growth hormone (rhGH) to mitigate against radiation injury in mice and nonhuman primates. BALB/c mice were irradiated with 7.5 Gy and treated post-irradiation with rhGH intravenously at a once daily dose of 20 µg/dose for 35 days. rhGH protected 17 out of 28 mice (60.7%) from lethal irradiation while only 3 out of 28 mice (10.7%) survived in the saline control group. A shorter course of 5 days of rhGH post-irradiation produced similar results. Compared with the saline control group, treatment with rhGH on irradiated BALB/c mice significantly accelerated overall hematopoietic recovery. Specifically, the recovery of total white cells, CD4 and CD8 T cell subsets, B cells, NK cells and especially platelets post radiation exposure were significantly accelerated in the rhGH-treated mice. Moreover, treatment with rhGH increased the frequency of hematopoietic stem/progenitor cells as measured by flow cytometry and colony forming unit assays in bone marrow harvested at day 14 after irradiation, suggesting the effects of rhGH are at the hematopoietic stem/progenitor level. rhGH mediated the hematopoietic effects primarily through their niches. Similar data with rhGH were also observed following 2 Gy sublethal irradiation of nonhuman primates. Our data demonstrate that rhGH promotes hematopoietic engraftment and immune recovery post the exposure of ionizing radiation and mitigates against the mortality from lethal irradiation even when administered after exposure.

Collaboration


Dive into the Diane Gesty-Palmer's collaboration.

Top Co-Authors

Avatar

Louis M. Luttrell

Medical University of South Carolina

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Bronwen Martin

Medical University of South Carolina

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Stuart Maudsley

Medical University of South Carolina

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge