Network


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

Hotspot


Dive into the research topics where Jerome C. Nwachukwu is active.

Publication


Featured researches published by Jerome C. Nwachukwu.


eLife | 2014

Resveratrol modulates the inflammatory response via an estrogen receptor-signal integration network

Jerome C. Nwachukwu; Sathish Srinivasan; Nelson E Bruno; Alexander A. Parent; Travis S. Hughes; Julie A. Pollock; Olsi Gjyshi; Valerie Cavett; Jason Nowak; Ruben D. Garcia-Ordonez; René Houtman; Patrick R. Griffin; Douglas J. Kojetin; John A. Katzenellenbogen; Michael D. Conkright; Kendall W. Nettles

Resveratrol has beneficial effects on aging, inflammation and metabolism, which are thought to result from activation of the lysine deacetylase, sirtuin 1 (SIRT1), the cAMP pathway, or AMP-activated protein kinase. In this study, we report that resveratrol acts as a pathway-selective estrogen receptor-α (ERα) ligand to modulate the inflammatory response but not cell proliferation. A crystal structure of the ERα ligand-binding domain (LBD) as a complex with resveratrol revealed a unique perturbation of the coactivator-binding surface, consistent with an altered coregulator recruitment profile. Gene expression analyses revealed significant overlap of TNFα genes modulated by resveratrol and estradiol. Furthermore, the ability of resveratrol to suppress interleukin-6 transcription was shown to require ERα and several ERα coregulators, suggesting that ERα functions as a primary conduit for resveratrol activity. DOI: http://dx.doi.org/10.7554/eLife.02057.001


Science Translational Medicine | 2015

Dual suppression of estrogenic and inflammatory activities for targeting of endometriosis

Yuechao Zhao; Ping Gong; Yiru Chen; Jerome C. Nwachukwu; Sathish Srinivasan; CheMyong Ko; Milan K. Bagchi; Robert N. Taylor; Kenneth S. Korach; Kendall W. Nettles; John A. Katzenellenbogen; Benita S. Katzenellenbogen

The estrogen-dependent inflammatory and neuroangiogenic activities that drive endometriosis can be suppressed by estrogen receptor ligands in mice. Fewer Lesions, More Little Mice Endometriosis is a poorly understood disorder of the female reproductive system, where collections of tissue that normally lines the uterus appear outside the uterus as well. These tissue deposits can be found anywhere in the abdominal cavity, where they cause inflammation and pain, and often also decreased fertility. Although some hormonal treatments for endometriosis exist, they are not always effective, have numerous side effects, and also suppress fertility. Now, Zhao et al. present some mechanistic explanations for the inflammatory phenomena seen in endometriosis. Even more importantly, the authors identified two new estrogen receptor ligands that can suppress endometriosis in mouse models safely and effectively, without disrupting the animals’ reproductive cycles and fertility. Estrogenic and inflammatory components play key roles in a broad range of diseases including endometriosis, a common estrogen-dependent gynecological disorder in which endometrial tissue creates inflammatory lesions at extrauterine sites, causing pelvic pain and reduced fertility. Current medical therapies focus primarily on reducing systemic levels of estrogens, but these are of limited effectiveness and have considerable side effects. We developed estrogen receptor (ER) ligands, chloroindazole (CLI) and oxabicycloheptene sulfonate (OBHS), which showed strong ER-dependent anti-inflammatory activity in a preclinical model of endometriosis that recapitulates the estrogen dependence and inflammatory responses of the disease in immunocompetent mice and in primary human endometriotic stromal cells in culture. Estrogen-dependent phenomena, including cell proliferation, cyst formation, vascularization, and lesion growth, were all arrested by CLI or OBHS, which prevented lesion expansion and also elicited regression of established lesions, suppressed inflammation, angiogenesis, and neurogenesis in the lesions, and interrupted crosstalk between lesion cells and infiltrating macrophages. Studies in ERα or ERβ knockout mice indicated that ERα is the major mediator of OBHS effectiveness and ERβ is dominant in CLI actions, implying involvement of both ERs in endometriosis. Neither ligand altered estrous cycling or fertility at doses that were effective for suppression of endometriosis. Hence, CLI and OBHS are able to restrain endometriosis by dual suppression of the estrogen-inflammatory axis. Our findings suggest that these compounds have the desired characteristics of preventive and therapeutic agents for clinical endometriosis and possibly other estrogen-driven and inflammation-promoted disorders.


Nature Chemical Biology | 2013

Ligand-binding dynamics rewire cellular signaling via estrogen receptor-α

Sathish Srinivasan; Jerome C. Nwachukwu; Alex A Parent; Valerie Cavett; Jason Nowak; Travis S. Hughes; Douglas J. Kojetin; John A. Katzenellenbogen; Kendall W. Nettles

Ligand-binding dynamics control allosteric signaling through the estrogen receptor-α (ERα), but the biological consequences of such dynamic binding orientations are unknown. Here, we compare a set of ER ligands having dynamic binding orientation (dynamic ligands) with a control set of isomers that are constrained to bind in a single orientation (constrained ligands). Proliferation of breast cancer cells directed by constrained ligands is associated with DNA binding, coactivator recruitment and activation of the estrogen-induced gene GREB1, reflecting a highly interconnected signaling network. In contrast, proliferation driven by dynamic ligands is associated with induction of ERα-mediated transcription in a DNA-binding domain (DBD)-dependent manner. Further, dynamic ligands displayed enhanced anti-inflammatory activity. The DBD-dependent profile was predictive of these signaling patterns in a larger diverse set of natural and synthetic ligands. Thus, ligand dynamics directs unique signaling pathways, and reveals a novel role of the DBD in allosteric control of ERα-mediated signaling.


Nature Communications | 2015

Structural mechanism for signal transduction in RXR nuclear receptor heterodimers

Douglas J. Kojetin; Edna Matta-Camacho; Travis S. Hughes; Sathish Srinivasan; Jerome C. Nwachukwu; Valerie Cavett; Jason Nowak; Michael J. Chalmers; David Marciano; Theodore M. Kamenecka; Andrew I. Shulman; Mark Rance; Patrick R. Griffin; John B. Bruning; Kendall W. Nettles

A subset of nuclear receptors (NRs) function as obligate heterodimers with retinoid X receptor (RXR), allowing integration of ligand-dependent signals across the dimer interface via an unknown structural mechanism. Using nuclear magnetic resonance (NMR) spectroscopy, x-ray crystallography and hydrogen/deuterium exchange (HDX) mass spectrometry, here we show an allosteric mechanism through which RXR co-operates with a permissive dimer partner, peroxisome proliferator-activated receptor (PPAR)-γ, while rendered generally unresponsive by a non-permissive dimer partner, thyroid hormone (TR) receptor. Amino acid residues that mediate this allosteric mechanism comprise an evolutionarily conserved network discovered by statistical coupling analysis (SCA). This SCA network acts as a signalling rheostat to integrate signals between dimer partners, ligands and coregulator-binding sites, thereby affecting signal transmission in RXR heterodimers. These findings define rules guiding how NRs integrate two ligand-dependent signalling pathways into RXR heterodimer-specific responses.


Journal of Medicinal Chemistry | 2013

Thiophene-Core Estrogen Receptor Ligands Having Superagonist Activity

Jian Min; Pengcheng Wang; Sathish Srinivasan; Jerome C. Nwachukwu; Pu Guo; Minjian Huang; Kathryn E. Carlson; John A. Katzenellenbogen; Kendall W. Nettles; Hai-Bing Zhou

To probe the importance of the heterocyclic core of estrogen receptor (ER) ligands, we prepared a series of thiophene-core ligands by Suzuki cross-coupling of aryl boronic acids with bromo-thiophenes and we assessed their receptor binding and cell biological activities. The disposition of the phenol substituents on the thiophene core, at alternate or adjacent sites, and the nature of substituents on these phenols, all contribute to binding affinity and subtype selectivity. Most of the bis(hydroxyphenyl)-thiophenes were ERβ selective, whereas the tris(hydroxyphenyl)-thiophenes were ERα selective; analogous furan-core compounds generally have lower affinity and less selectivity. Some diarylthiophenes show distinct superagonist activity in reporter gene assays, giving maximal activities 2-3 times that of estradiol, and modeling suggests that these ligands have a different interaction with a hydrogen-bonding residue in helix-11. Ligand-core modification may be a new strategy for developing ER ligands whose selectivity is based on having transcriptional activity greater than that of estradiol.


The EMBO Journal | 2014

Creb coactivators direct anabolic responses and enhance performance of skeletal muscle

Nelson E Bruno; Kimberly A Kelly; Richard Hawkins; Mariam Bramah-Lawani; Antonio L. Amelio; Jerome C. Nwachukwu; Kendall W. Nettles; Michael D. Conkright

During the stress response to intense exercise, the sympathetic nervous system (SNS) induces rapid catabolism of energy reserves through the release of catecholamines and subsequent activation of protein kinase A (PKA). Paradoxically, chronic administration of sympathomimetic drugs (β‐agonists) leads to anabolic adaptations in skeletal muscle, suggesting that sympathetic outflow also regulates myofiber remodeling. Here, we show that β‐agonists or catecholamines released during intense exercise induce Creb‐mediated transcriptional programs through activation of its obligate coactivators Crtc2 and Crtc3. In contrast to the catabolic activity normally associated with SNS function, activation of the Crtc/Creb transcriptional complex by conditional overexpression of Crtc2 in the skeletal muscle of transgenic mice fostered an anabolic state of energy and protein balance. Crtc2‐overexpressing mice have increased myofiber cross‐sectional area, greater intramuscular triglycerides and glycogen content. Moreover, maximal exercise capacity was enhanced after induction of Crtc2 expression in transgenic mice. Collectively these findings demonstrate that the SNS‐adrenergic signaling cascade coordinates a transient catabolic stress response during high‐intensity exercise, which is followed by transcriptional reprogramming that directs anabolic changes for recovery and that augments subsequent exercise performance.


ChemMedChem | 2012

Development of selective estrogen receptor modulator (SERM)-like activity through an indirect mechanism of estrogen receptor antagonism: defining the binding mode of 7-oxabicyclo[2.2.1]hept-5-ene scaffold core ligands.

Yangfan Zheng; Manghong Zhu; Sathish Srinivasan; Jerome C. Nwachukwu; Valerie Cavett; Jian Min; Kathryn E. Carlson; Pengcheng Wang; Chune Dong; John A. Katzenellenbogen; Kendall W. Nettles; Hai-Bing Zhou

Previously, we discovered estrogen receptor (ER) ligands with a novel three‐dimensional oxabicyclo[2.2.1]heptene core scaffold and good ER binding affinity act as partial agonists via small alkyl ester substitutions on the bicyclic core that indirectly modulate the critical switch helix in the ER ligand binding domain, helix 12, by interactions with helix 11. This contrasts with the mechanism of action of tamoxifen, which directly pushes helix 12 out of the conformation required for gene activation. We now report that a much larger substitution can be tolerated at this position of the bicyclic core scaffold, namely a phenyl sulfonate group, which defines a novel binding epitope for the estrogen receptor. We prepared an array of 14 oxabicycloheptene sulfonates, varying the phenyl sulfonate group. As with the parent compound, 5,6‐bis‐(4‐hydroxyphenyl)‐7‐oxabicyclo[2.2.1]hept‐5‐ene‐2‐sulfonic acid phenyl ester (OBHS), these compounds showed preferential affinity for ERα, and the disposition and size of the phenyl substituents were important determinants of the binding affinity and selectivity of these compounds, with those having ortho substituents giving the highest, and para substituents the lowest affinities for ERα. A few analogues exhibit ERα binding affinities that are comparable to or, in the case of the ortho‐chloro analogue, higher than that of OBHS itself. In cell‐based studies, we found several compounds with activity profiles comparable to tamoxifen, but acting entirely as indirect antagonists, allosterically interfering with recruitment of coactivator proteins to the receptor. Thus, the OBHS binding epitope represents a novel approach to the development of estrogen receptor antagonists via an indirect mechanism of antagonism.


Journal of Medicinal Chemistry | 2012

Identification and Structure-Activity Relationships of a Novel Series of Estrogen Receptor Ligands Based on 7-Thiabicyclo[2.2.1]hept-2-ene-7-oxide

Pengcheng Wang; Jian Min; Jerome C. Nwachukwu; Valerie Cavett; Kathryn E. Carlson; Pu Guo; Manghong Zhu; Yangfan Zheng; Chune Dong; John A. Katzenellenbogen; Kendall W. Nettles; Hai-Bing Zhou

To develop estrogen receptor (ER) ligands having novel structures and activities, we have explored compounds in which the central hydrophobic core has a more three-dimensional topology than typically found in estrogen ligands and thus exploits the unfilled space in the ligand-binding pocket. Here, we build upon our previous investigations of 7-oxabicyclo[2.2.1]heptene core ligands, by replacing the oxygen bridge with a sulfoxide. These new 7-thiabicyclo[2.2.1]hept-2-ene-7-oxides were conveniently prepared by a Diels-Alder reaction of 3,4-diarylthiophenes with dienophiles in the presence of an oxidant and give cycloadducts with endo stereochemistry. Several new compounds demonstrated high binding affinities with excellent ERα selectivity, but unlike oxabicyclic compounds, which are transcriptional antagonists, most thiabicyclic compounds are potent, ERα-selective agonists. Modeling suggests that the gain in activity of the thiabicyclic compounds arises from their endo stereochemistry that stabilizes an active ER conformation. Further, the disposition of methyl substituents in the phenyl groups attached to the bicyclic core unit contributes to their binding affinity and subtype selectivity.


Molecular and Cellular Biology | 2011

Regulation of Androgen Receptor-Mediated Transcription by RPB5 Binding Protein URI/RMP

Paolo Mita; Jeffrey N. Savas; Nabil Djouder; John R. Yates; Susan Ha; Rachel Ruoff; Eric D. Schafler; Jerome C. Nwachukwu; Naoko Tanese; Nicholas J. Cowan; Jiri Zavadil; Michael J. Garabedian; Susan K. Logan

ABSTRACT Androgen receptor (AR)-mediated transcription is modulated by interaction with coregulatory proteins. We demonstrate that the unconventional prefoldin RPB5 interactor (URI) is a new regulator of AR transcription and is critical for antagonist (bicalutamide) action. URI is phosphorylated upon androgen treatment, suggesting communication between the URI and AR signaling pathways. Whereas depletion of URI enhances AR-mediated gene transcription, overexpression of URI suppresses AR transcriptional activation and anchorage-independent prostate cancer cell growth. Repression of AR-mediated transcription is achieved, in part, by URI binding and regulation of androgen receptor trapped clone 27 (Art-27), a previously characterized AR corepressor. Consistent with this idea, genome-wide expression profiling in prostate cancer cells upon depletion of URI or Art-27 reveals substantially overlapping patterns of gene expression. Further, depletion of URI increases the expression of the AR target gene NKX-3.1, decreases the recruitment of Art-27, and increases AR occupancy at the NKX-3.1 promoter. While Art-27 can bind AR directly, URI is bound to chromatin prior to hormone-dependent recruitment of AR, suggesting a role for URI in modulating AR recruitment to target genes.


Nature Chemical Biology | 2017

Full antagonism of the estrogen receptor without a prototypical ligand side chain.

Sathish Srinivasan; Jerome C. Nwachukwu; Nelson E Bruno; Venkatasubramanian Dharmarajan; Devrishi Goswami; Irida Kastrati; Scott Novick; Jason Nowak; Valerie Cavett; Hai-Bing Zhou; Nittaya Boonmuen; Yuechao Zhao; Jian Min; Jonna Frasor; Benita S. Katzenellenbogen; Patrick R. Griffin; John A. Katzenellenbogen; Kendall W. Nettles

Resistance to endocrine therapies remains a significant clinical problem for estrogen receptor-α (ERα)-positive breast cancer. On-target side effects limit therapeutic compliance and use for chemoprevention, highlighting an unmet need for new therapies. Here we present a full-antagonist ligand series lacking the prototypical ligand side chain that has been universally used to engender antagonism of ERα through poorly understood structural mechanisms. A series of crystal structures and phenotypic assays reveal a structure-based design strategy with separate design elements for antagonism and degradation of the receptor and access to a structurally distinct space for further improvements in ligand design. Understanding structural rules that guide ligands to produce diverse ERα-mediated phenotypes has broad implications for the treatment of breast cancer and other estrogen-sensitive aspects of human health including bone homeostasis, energy metabolism, and autoimmunity.

Collaboration


Dive into the Jerome C. Nwachukwu's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jason Nowak

Scripps Research Institute

View shared research outputs
Top Co-Authors

Avatar

Valerie Cavett

Scripps Research Institute

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Nelson E Bruno

Scripps Research Institute

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge