Network


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

Hotspot


Dive into the research topics where Jeffrey R. Bender is active.

Publication


Featured researches published by Jeffrey R. Bender.


Circulation Research | 2000

Membrane Estrogen Receptor Engagement Activates Endothelial Nitric Oxide Synthase via the PI3-Kinase–Akt Pathway in Human Endothelial Cells

M. Page Haynes; Diviya Sinha; Kerry S. Russell; Mark Collinge; David Fulton; Manuel Morales-Ruiz; William C. Sessa; Jeffrey R. Bender

17&bgr;-Estradiol (E2) is a rapid activator of endothelial nitric oxide synthase (eNOS). The product of this activation event, NO, is a fundamental determinant of cardiovascular homeostasis. We previously demonstrated that E2-stimulated endothelial NO release can occur without an increase in cytosolic Ca2+. Here we demonstrate for the first time, to our knowledge, that E2 rapidly induces phosphorylation and activation of eNOS through the phosphatidylinositol 3 (PI3)-kinase–Akt pathway. E2 treatment (10 ng/mL) of the human endothelial cell line, EA.hy926, resulted in increased NO production, which was abrogated by the PI3-kinase inhibitor, LY294002, and the estrogen receptor antagonist ICI 182,780. E2 stimulated rapid Akt phosphorylation on serine 473. As has been shown for vascular endothelial growth factor, eNOS is an E2-activated Akt substrate, demonstrated by rapid eNOS phosphorylation on serine 1177, a critical residue for eNOS activation and enhanced sensitivity to resting cellular Ca2+ levels. Adenoviral-mediated EA.hy926 transduction confirmed functional involvement of Akt, because a kinase-deficient, dominant-negative Akt abolished E2-stimulated NO release. The membrane-impermeant E2BSA conjugate, shown to bind endothelial cell membrane sites, also induced rapid Akt and consequent eNOS phosphorylation. Thus, engagement of membrane estrogen receptors results in rapid endothelial NO release through a PI3-kinase–Akt-dependent pathway. This explains, in part, the reduced requirement for cytosolic Ca2+ fluxes and describes an important pathway relevant to cardiovascular pathophysiology.


Circulation Research | 1997

17β-Estradiol Regulation of Human Endothelial Cell Basal Nitric Oxide Release, Independent of Cytosolic Ca2+ Mobilization

Teresa Caulin-Glaser; Guillermo García-Cardeña; Phillip Sarrel; William C. Sessa; Jeffrey R. Bender

Estradiol retards the development of atherosclerosis. Animal models have suggested that NO may be a critical effector molecule in this cardiovascular protection. In this study, female human umbilical vein endothelial cells (HUVECs) were propagated in phenol red-free gonadal hormone-free medium and pretreated with 17 beta-estradiol (E2). Reduced NO2- and NO3- (NOx) concentration, determined by chemiluminescence, demonstrated a rapid increase in basal HUVEC NO release in response to physiological concentrations of E2. The estrogen receptor (ER) antagonist ICI 164,384 inhibited the augmented NO release, demonstrating an ER-mediated component of this response. Because endothelial NO synthase (eNOS) activity is largely regulated by cytosolic Ca2+, relative [Ca2+]i in response to E2 was determined in a fluorometric assay. E2 did not promote HUVEC Ca2+ fluxes. Furthermore, eNOS activity in E2-pretreated endothelial whole-cell lysates was not dependent on additional Ca2+. Despite involving the ER, this is a nongenomic effect E2, as demonstrated by maintained responses in transcriptionally inhibited cells and by the rapidly (10 minutes) of cGMP formation in an NO bioassay. We demonstrate, for the first time, that independent of cytosolic Ca2+ mobilization, there is augmentation of eNOS activity with a resultant increase in HUVEC basal NO release in response to short-term estradiol exposure. Implications for the cardiovascular protective role of estrogen are discussed.


Proceedings of the National Academy of Sciences of the United States of America | 2003

Plasma membrane localization and function of the estrogen receptor α variant (ER46) in human endothelial cells

Lei Li; M. Page Haynes; Jeffrey R. Bender

Estrogen receptor (ER) α variants have been identified in an array of nonendothelial cells. We previously demonstrated that estrogen rapidly induces nitric oxide release via a phosphatidylinositol 3-kinase/Akt/endothelial nitric-oxide synthase (eNOS) pathway in EA.hy926 cells (immortalized human endothelial cells), which express a 46-kDa ER. We now confirm that, due to alternative splicing, the 46-kDa endothelial cell protein (ER46) is an amino-terminal truncated product of full-length ERα (ER66). ER46 is expressed in the plasma membrane, cytosol, and nucleus of resting, estrogen-deprived cells. Flow cytometric and immunofluorescence microscopic analyses demonstrated that the ER46 C but not N terminus is Ab-accessible in the plasma membrane. Inhibition of palmitoylation with tunicamycin and [3H]palmitic acid labeling demonstrated an estrogen-induced, palmitoylation-dependent plasma membrane ER46 recruitment, with reorganization into caveolae. In reconstituted, estrogen-stimulated COS-7 (ER-null) cells, membrane ER46 more efficiently triggered membrane eNOS phosphorylation than ER66. Conversely, ER66 more efficiently mediated estrogen response element reporter-gene transactivation than ER46. These results demonstrate that ER46 is localized and further dynamically targeted to the plasma membrane in a palmitoylation-dependent manner. ER46 more efficiently modulates membrane-initiated estrogen actions, including eNOS activation, than full-length ER66. These findings may have important implications in vascular-specific targeting of estrogen receptor agonists.


Journal of Clinical Investigation | 1996

Effects of 17beta-estradiol on cytokine-induced endothelial cell adhesion molecule expression.

Teresa Caulin-Glaser; C. A. Watson; Ruggero Pardi; Jeffrey R. Bender

One of the earliest events in atherosclerosis is interaction of circulating mononuclear leukocytes and the endothelium. Endothelial cell (EC) activation by cytokines results in expression of adhesion molecules and production of chemotactic factors, augmenting leukocyte adhesion and recruitment, respectively. The incidence of atherosclerosis in premenopausal women is significantly less than that observed in age-matched males with similar risk profiles. Because estrogen has gene regulatory effects, we investigated whether 17beta-estradiol (E2) can inhibit cytokine-mediated EC adhesion molecule transcriptional activation. Cultured human umbilical vein EC (estrogen receptor-positive) were propagated in gonadal hormone-free medium and were E2-pretreated for 48 h before IL-1 activation. Detected by FACS analysis, E2 strongly (60-80%) inhibited IL-1-mediated membrane E-selectin and vascular cell adhesion molecule-1 induction, and intercellular adhesion molecule-1 hyperinduction. 17alpha-estradiol (an inactive E2 stereoisomer) had no effect. This inhibition correlated with similar reductions in steady state-induced E-selectin mRNA levels, and was abrogated by the E2 antagonist ICI 164,384, demonstrating a specific, estrogen receptor-mediated effect. Nuclear run-offs confirmed suppression at the transcriptional level. The implications of these results for the cardiovascular protective role of estrogen are discussed.


Immunology Today | 1992

Regulatory mechanisms in leukocyte adhesion: flexible receptors for sophisticated travelers

Ruggero Pardi; Luca Inverardi; Jeffrey R. Bender

Unstimulated leukocytes spend extended periods circulating in the blood, punctuated by migration through lymphoid areas and peripheral tissues. During transit, strong cell-cell interactions control immune surveillance and specialized effector functions. The structures and mechanisms that allow this flexible adhesion and migration behavior are the subject of this review.


Steroids | 2005

Vascular Cell Signaling by Membrane Estrogen Receptors

Kyung Hee Kim; Katie Moriarty; Jeffrey R. Bender

The definition of estrogens actions has expanded from transcriptional regulation to the rapid, membrane-initiated activation of numerous signal transduction cascades. Multiple biological effects of estrogen have been shown in numerous animals, cellular and molecular studies, which support the favorable effects of estrogen on vascular structure, function, and cell signaling. Work from several laboratories has shown that these effects are mediated by distinct forms of estrogen receptor (ER) alpha. This includes estrogen-stimulated rapid activation of endothelial nitric oxide synthase (eNOS), resulting in the elaboration of the athero-protective, angiogenesis-promoting product nitric oxide (NO). We have described the expression of ER46, an N-terminus truncated isoform of the ERalpha, in human endothelial cells (EC), and its critical role in membrane-initiated, rapid responses to 17beta-estradiol (E2). We have proposed an ER46-centered, eNOS activating molecular complex in human EC caveolar membranes, containing c-Src, phosphatidylinositol 3-kinase (PI3K), Akt and eNOS. Our previous studies support estrogen-induced rapid eNOS activation via a sequential c-Src/PI3K/Akt cascade in EC. In this review, we describe estrogen-induced, rapid, non-genomic actions in endothelium, driven by c-Src-ER46-caveolin-1 interactions, with consequent activation of eNOS. Amidst ongoing controversies in hormone replacement therapy, these molecular and cellular data, defining favorable estrogenic effects on the endothelium, provide a strong impetus to resolve these clinical questions.


American Journal of Physiology-heart and Circulatory Physiology | 1999

Neuregulin activation of ErbB receptors in vascular endothelium leads to angiogenesis

Kerry S. Russell; David F. Stern; Peter J. Polverini; Jeffrey R. Bender

The ErbB, or epidermal growth factor receptor (EGF-r), family of transmembrane tyrosine kinase receptors has been demonstrated to play an important role in growth regulation and intracellular signaling in a wide variety of cell types. Targeted deletion of neuregulin (an ErbB ligand) in mice results in endocardial cushion abnormalities, suggesting that these receptor-ligand interactions have important effects on vascular endothelial growth and development. To study the role of ErbB receptor signaling in vascular endothelium, we investigated the expression pattern of the various receptor family members and the effect of ErbB receptor stimulation in human umbilical vein endothelial cells (HUVEC). We demonstrate that ErbB2 (neu), ErbB3, and ErbB4 are highly expressed, whereas ErbB1 (EGF-r) is undetectable. Stimulation of HUVEC with recombinant neuregulin-beta (an ErbB3/4 ligand) induces rapid calcium fluxes, receptor tyrosine phosphorylation, and cell proliferation. We demonstrate marked in vitro and in vivo angiogenic responses to neuregulin-beta, which are independent of vascular endothelial cell growth factor. These findings support an important role for the ErbB family of receptors in endothelial cell signaling and function, including neuregulin-induced angiogenesis.The ErbB, or epidermal growth factor receptor (EGF-r), family of transmembrane tyrosine kinase receptors has been demonstrated to play an important role in growth regulation and intracellular signaling in a wide variety of cell types. Targeted deletion of neuregulin (an ErbB ligand) in mice results in endocardial cushion abnormalities, suggesting that these receptor-ligand interactions have important effects on vascular endothelial growth and development. To study the role of ErbB receptor signaling in vascular endothelium, we investigated the expression pattern of the various receptor family members and the effect of ErbB receptor stimulation in human umbilical vein endothelial cells (HUVEC). We demonstrate that ErbB2 (neu), ErbB3, and ErbB4 are highly expressed, whereas ErbB1 (EGF-r) is undetectable. Stimulation of HUVEC with recombinant neuregulin-β (an ErbB3/4 ligand) induces rapid calcium fluxes, receptor tyrosine phosphorylation, and cell proliferation. We demonstrate marked in vitro and in vivo angiogenic responses to neuregulin-β, which are independent of vascular endothelial cell growth factor. These findings support an important role for the ErbB family of receptors in endothelial cell signaling and function, including neuregulin-induced angiogenesis.


Journal of Immunology | 2000

Simvastatin Modulates Cytokine-Mediated Endothelial Cell Adhesion Molecule Induction: Involvement of an Inhibitory G Protein

Mehran M. Sadeghi; Mark Collinge; Ruggero Pardi; Jeffrey R. Bender

Endothelial cell adhesion molecules (CAMs) E-selectin, ICAM-1, and VCAM-1 play variably important roles in immune-mediated processes. They are induced by the proinflammatory cytokines IL-1 and TNF-α, and NF-κB is required for the regulated expression of all three genes. Regulators of this pathway could potentially be potent immune modulators. We studied the effect of a 3-hydroxy-3-methylglutaryl coenzyme A reductase inhibitor, simvastatin, on cytokine-induced expression of CAMs in HUVEC. Unexpectedly, pretreatment with simvastatin potentiated the induction of all three endothelial CAMs by IL-1 and TNF, but not LPS or PMA, as detected by flow cytometry. Northern blot analysis demonstrated an increase in steady state IL-1-induced E-selectin mRNA levels in cells pretreated with simvastatin. This was associated with an increase in nuclear translocation of NF-κB, as detected by EMSA. The effect of simvastatin was reversed by mevalonate and geranylgeranyl pyrophosphate but not squalene, indicating that an inhibitory prenylated protein is involved in endothelial responses to proinflammatory cytokines. Pertussis toxin mimicked the effect of simvastatin, and the G protein activator NaF inhibited the cytokine-induced expression of endothelial CAMs, indicating that a Giα protein is involved. These results demonstrate that cytokine-mediated activation of the endothelium, and specifically CAM induction, can be modulated by a heterotrimeric G protein-coupled pathway. This may represent a “basal tone” of endothelial inactivation, which can either be disinhibited or amplified, depending on the stimulus.


Immunology Today | 1997

Through and beyond the wall: late steps in leukocyte transendothelial migration

Elisabetta Bianchi; Jeffrey R. Bender; Francesco Blasi; Ruggero Pardi

A combination of approaches has provided firm experimental support for a step-wise model of leukocyte adhesion to vascular endothelium under hemodynamic shear stress. However, leukocyte adhesion to vascular endothelium acquires functional significance only if coupled to transmigration and progression into extravascular tissues. As discussed here by Elisabetta Bianchi and colleagues, these latter processes involve the sequential activation of adhesion-dependent functional programs, carried out by structurally diverse multimolecular complexes.


Circulation | 2004

Detection of Injury-Induced Vascular Remodeling by Targeting Activated αvβ3 Integrin In Vivo

Mehran M. Sadeghi; Svetlana Krassilnikova; Jiasheng Zhang; Amir A. Gharaei; Hooman Rastegar Fassaei; Leila Esmailzadeh; Ali Kooshkabadi; Scott Edwards; Padmaja Yalamanchili; Thomas D. Harris; Albert J. Sinusas; Barry L. Zaret; Jeffrey R. Bender

Background—The &agr;vβ3 integrin plays a critical role in cell proliferation and migration. We hypothesized that vascular cell proliferation, a hallmark of injury-induced remodeling, can be tracked by targeting &agr;vβ3 integrin expression in vivo. Methods and Results—RP748, a novel 111In-labeled &agr;vβ3-specific radiotracer, was evaluated for its cell-binding characteristics and ability to track injury-induced vascular proliferation in vivo. Three groups of experiments were performed. In cultured endothelial cells (ECs), TA145, a cy3-labeled homologue of RP748, localized to &agr;vβ3 at focal contacts. Activation of&agr;vβ3 by Mn 2+ led to increased EC binding of TA145. Left common carotid artery wire injury in apolipoprotein E−/− mice led to vascular wall expansion over a period of 4 weeks. RP748 (7.4 MBq) was injected into groups of 9 mice at 1, 3, or 4 weeks after left carotid injury, and carotids were harvested for autoradiography. Relative autographic intensity, defined as counts/pixel of the injured left carotid area divided by counts/pixel of the uninjured right carotid area, was higher at 1 and 3 weeks (1.8±0.1 and 1.9±0.2, respectively) and decreased significantly by 4 weeks after injury (1.4±0.1, P <0.05). Carotid &agr;v and β3 integrin expression was maximal at 1 week and decreased by 4 weeks after injury. The proliferation index, as determined by Ki67 staining, followed a temporal pattern similar to that of RP748 uptake. Dynamic gamma imaging demonstrated rapid renal clearance of RP748. Conclusions—RP748 has preferential binding to activated &agr;vβ3 integrin and can track the injury-induced vascular proliferative process in vivo.

Collaboration


Dive into the Jeffrey R. Bender's collaboration.

Top Co-Authors

Avatar

Ruggero Pardi

Vita-Salute San Raffaele University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Vinod S. Ramgolam

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge