Network


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

Hotspot


Dive into the research topics where Guemalli R. Cardona is active.

Publication


Featured researches published by Guemalli R. Cardona.


Journal of Biological Chemistry | 1997

TRAM-1, A Novel 160-kDa Thyroid Hormone Receptor Activator Molecule, Exhibits Distinct Properties from Steroid Receptor Coactivator-1

Akira Takeshita; Guemalli R. Cardona; Noriyuki Koibuchi; Chen-Shian Suen; William W. Chin

Nuclear hormone receptors (NRs) are ligand-dependent transcription factors that regulate target gene transcription. We report the molecular cloning and characterization of a novel human cDNA encoding TRAM-1, athyroid hormone receptor activatormolecule, a ∼160-kDa protein homologous with SRC-1/TIF2, by far-Western-based expression screening. TRAM-1 binds to thyroid hormone receptor (TR) and other NRs in a ligand-dependent manner and enhances ligand-induced transcriptional activity of TR. The AF-2 region in NRs has been thought to play a critical role in mediating ligand-dependent transactivation by the interaction with coactivators. Surprisingly, TRAM-1 retains strong ligand-dependent interaction with an AF-2 mutant of TR (E457A), while SRC-1 fails to interact with this mutant. Furthermore, we identified a critical TRAM-1 binding site in rat TRβ1 outside of AF-2, as TRAM-1 shows weak ligand-dependent interaction with a helix 3 ligand binding domain TR mutant (K288A), compared with SRC-1. These results suggest that TRAM-1 is a coactivator that may exhibit its activity by interacting with subdomains of NRs other than the AF-2 region, in contrast to SRC-1/TIF2.


Biochemical and Biophysical Research Communications | 1992

Isolation and characterization of cDNAs encoding the rat pituitary gonadotropin-releasing hormone receptor

Ursula B. Kaiser; Dayao Zhao; Guemalli R. Cardona; William W. Chin

Rat pituitary cDNAs encoding the full peptide coding sequence of the rat gonadotropin-releasing hormone receptor were isolated and characterized. The deduced amino acid sequence encodes a protein of 327 residues with seven putative transmembrane domains characteristic of the family of G-protein coupled receptors. It is 95% identical at the amino acid level with the mouse gonadotropin-releasing hormone receptor. An mRNA of 4.5 Kb was identified in the rat pituitary, ovary, and testis, and in murine alpha T3 cells. In addition, a larger mRNA species of 5.0-5.5 Kb was present in these rat tissues, and a smaller mRNA species of 1.8 Kb was present in the rat pituitary and ovary, and in alpha T3 cells. The receptor mRNA levels were increased in the female rat pituitary after ovariectomy compared to levels in intact female rats.


Journal of Biological Chemistry | 1999

Identification and Characterization of the Gonadotropin-releasing Hormone Response Elements in the Mouse Gonadotropin-releasing Hormone Receptor Gene

Errol R. Norwitz; Guemalli R. Cardona; Kyeong-Hoon Jeong; William W. Chin

The response of the pituitary gonadotrope to gonadotropin-releasing hormone (GnRH) correlates directly with the concentration of GnRH receptors (GnRHR) on the cell surface, which is mediated in part at the level of GnRHR gene expression. Several hormones have been implicated in this regulation, most notably GnRH itself. Despite these observations and the central role that GnRH is known to play in reproductive development and function, the molecular mechanism(s) by which GnRH regulates transcription of the GnRHR gene has not been well elucidated. Previous studies in this laboratory have identified and partially characterized the promoter region of the mouse GnRHR gene and demonstrated that the regulatory elements for tissue-specific expression as well as for GnRH regulation are present within the 1.2-kilobase 5′-flanking sequence. By using deletion and mutational analysis as well as functional transfection studies in the murine gonadotrope-derived αT3-1 cell line, we have localized GnRH responsiveness of the mouse GnRHR gene to two DNA sequences at −276/−269 (designated Sequence UnderlyingResponsiveness to GnRH-2 (SURG-2), which contains the consensus sequence for the activating protein-1-binding site) and −292/−285 (a novel element designated SURG-1), and demonstrated that this response is mediated via protein kinase C. By using the electrophoretic mobility shift assay, we further demonstrate that a member(s) of the Fos/Jun heterodimer superfamily is responsible in part for the DNA-protein complexes formed on SURG-2, using αT3-1 nuclear extracts. These data define a bipartite GnRH response element in the mouse GnRHR 5′-flanking sequence and suggest that the activating protein-1 complex plays a central role in conferring GnRH responsiveness to the murine GnRHR gene.


Molecular and Cellular Biology | 2002

Identification and Characterization of a Tissue-Specific Coactivator, GT198, That Interacts with the DNA-Binding Domains of Nuclear Receptors

Lan Ko; Guemalli R. Cardona; Alexandra Henrion-Caude; William W. Chin

ABSTRACT Gene activation mediated by nuclear receptors is regulated in a tissue-specific manner and requires interactions between nuclear receptors and their cofactors. Here, we identified and characterized a tissue-specific coactivator, GT198, that interacts with the DNA-binding domains of nuclear receptors. GT198 was originally described as a genomic transcript that mapped to the human breast cancer susceptibility locus 17q12-q21 with unknown function. We show that GT198 exhibits a tissue-specific expression pattern in which its mRNA is elevated in testis, spleen, thymus, pituitary cells, and several cancer cell lines. GT198 is a 217-amino-acid nuclear protein that contains a leucine zipper required for its dimerization. In vitro binding and yeast two-hybrid assays indicated that GT198 interacted with nuclear receptors through their DNA-binding domains. GT198 potently stimulated transcription mediated by estrogen receptor α and β, thyroid hormone receptor β1, androgen receptor, glucocorticoid receptor, and progesterone receptor. However, the action of GT198 was distinguishable from that of the ligand-binding domain-interacting nuclear receptor coactivators, such as TRBP, CBP, and SRC-1, with respect to basal activation and hormone sensitivity. Furthermore, protein kinase A, protein kinase C, and mitogen-activated protein kinase can phosphorylate GT198 in vitro, and cotransfection of these kinases regulated the transcriptional activity of GT198. These data suggest that GT198 is a tissue-specific, kinase-regulated nuclear receptor coactivator that interacts with the DNA-binding domains of nuclear receptors.


Journal of Biological Chemistry | 1998

Thyroid Hormone Response Elements Differentially Modulate the Interactions of Thyroid Hormone Receptors with Two Receptor Binding Domains in the Steroid Receptor Coactivator-1

Akira Takeshita; Paul M. Yen; Masato Ikeda; Guemalli R. Cardona; Ying Liu; Noriyuki Koibuchi; Errol R. Norwitz; William W. Chin

Ligand-dependent transcriptional activation by nuclear receptors is mediated by interactions with coactivators. Recently, a consensus interaction motif (LXXLL) has been identified in a number of coactivators such as steroid receptor coactivator-1 (SRC-1). SRC-1 contains three such motifs in the central (nuclear receptor binding domain-1, NBD-1) and a single one in the C-terminal (NBD-2) regions. To define the nature and role of the two NBDs in SRC-1, interaction studies between the two NBDs and thyroid hormone receptor (TR) were performed. Although NBD-1 and NBD-2 showed similar ligand- and AF-2-dependent interactions with TR in solution, these two NBDs possessed distinct interaction properties with TR when TR is bound to a thyroid hormone-response element (TRE). Both in vitro and in vivo interaction studies demonstrate that NBD-1, but not NBD-2, exhibits ligand-dependent interaction with TR in the presence of TREs. In addition, a natural isoform of SRC-1, SRC-1E, which lacks NBD-2, preserved TR as well as progesterone receptor-mediated coactivator function on reporter gene expression. Finally, we found that NBD-1 failed to interact with a TR and retinoid X receptor heterodimer complex on a transcriptionally inactive direct repeat +4 TRE in electrophoretic mobility shift assays. These observations indicate that DNA-induced, as well as ligand-induced, conformational change(s) of TR may influence the nature of its binding to SRC-1, and that the two NBDs of SRC-1 may play different roles to regulate ligand-dependent transactivation of TRs.


Journal of Cellular Biochemistry | 1999

EXPRESSION OF MULTIPLE THYROID HORMONE RECEPTOR ISOFORMS IN RAT FEMORAL AND VERTEBRAL BONE AND IN BONE MARROW OSTEOGENIC CULTURES

Moira Milne; Moo-Il Kang; Guemalli R. Cardona; John M. Quail; Lewis E. Braverman; William W. Chin; Daniel T. Baran

Thyroid hormones influence both bone formation and bone resorption. Clinical data and animal studies provide evidence of skeletal site heterogeneity (hip vs. spine) of bone responses to thyroid hormones. In vitro studies also demonstrate direct effects of thyroid hormones on cells of the osteoblast lineage. Transcriptional regulation by thyroid hormone is mediated by ligand‐dependent transcription factors called thyroid hormone receptors (TRs). Two genes, c‐ErbAα and c‐ErbAβ, generate at least four TR isoforms in the rat: TRα1, c‐erbAα2, TRβ1, and TRβ2. Although functional TRs have been identified in cells of the osteoblast lineage, it is still not known if TR isoform expression in bone differs depending upon which skeletal site is examined. We have used ribonuclease protection assay and Northern blot analysis to simultaneously examine the expression of TR isoform mRNAs in adult rat femoral and vertebral bone. TRα1, c‐erbAα2, and TRβ1 are expressed in both femur and vertebra whole bone. Bone marrow cells from both skeletal sites were also cultured under conditions whereby the osteoprogenitors differentiated into osteoblasts and formed a mineralized extracellular matrix. TRα1, c‐erbAα2, and TRβ1 mRNAs are each expressed in both femoral and vertebral osteoblast cultures. The presence of TRα1, c‐erbAα2, and β1 proteins was confirmed by Western analysis of nuclear protein extracts from femoral and vertebral cell cultures. These results indicate that the three predominant TR isoforms are highly expressed in bone and osteoblasts from femurs and vertebrae. Whether there are distinct mechanisms of thyroid hormone action mediated by TRα1, c‐erbAα2, and TRβ1 at these separate skeletal sites remain to be shown. J. Cell. Biochem. 74:684–693, 1999.


Journal of Medicinal Chemistry | 2016

The Discovery, Preclinical, and Early Clinical Development of Potent and Selective GPR40 Agonists for the Treatment of Type 2 Diabetes Mellitus (LY2881835, LY2922083, and LY2922470)

Chafiq Hamdouchi; Steven D. Kahl; Anjana Patel Lewis; Guemalli R. Cardona; Richard W. Zink; Keyue Chen; Thomas E. Eessalu; James Ficorilli; Marialuisa C. Marcelo; Keith A. Otto; Kelly L. Wilbur; Jayana P. Lineswala; Jared L. Piper; D. Scott Coffey; Stephanie Ann Sweetana; Joseph Haas; Dawn A. Brooks; Edward J. Pratt; Ruth M. Belin; Mark A. Deeg; Xiaosu Ma; Ellen A. Cannady; Jason T. Johnson; Nathan Yumibe; Qi Chen; Pranab Maiti; Chahrzad Montrose-Rafizadeh; Yanyun Chen; Anne Reifel Miller

The G protein-coupled receptor 40 (GPR40) also known as free fatty acid receptor 1 (FFAR1) is highly expressed in pancreatic, islet β-cells and responds to endogenous fatty acids, resulting in amplification of insulin secretion only in the presence of elevated glucose levels. Hypothesis driven structural modifications to endogenous FFAs, focused on breaking planarity and reducing lipophilicity, led to the identification of spiropiperidine and tetrahydroquinoline acid derivatives as GPR40 agonists with unique pharmacology, selectivity, and pharmacokinetic properties. Compounds 1 (LY2881835), 2 (LY2922083), and 3 (LY2922470) demonstrated potent, efficacious, and durable dose-dependent reductions in glucose levels along with significant increases in insulin and GLP-1 secretion during preclinical testing. A clinical study with 3 administered to subjects with T2DM provided proof of concept of 3 as a potential glucose-lowering therapy. This manuscript summarizes the scientific rationale, medicinal chemistry, preclinical, and early development data of this new class of GPR40 agonists.


Bioorganic & Medicinal Chemistry Letters | 2015

Design and synthesis of a novel series of [1-(4-hydroxy-benzyl)-1H-indol-5-yloxy]-acetic acid compounds as potent, selective, thyroid hormone receptor β agonists

Timothy Paul Burkholder; Brian Eugene Cunningham; Joshua Ryan Clayton; Peter Ambrose Lander; Matthew L. Brown; Robert Anthony Doti; Gregory L. Durst; Chahrzad Montrose-Rafizadeh; Constance King; Harold E. Osborne; Robert M. Amos; Richard W. Zink; Lawrence E. Stramm; Thomas P. Burris; Guemalli R. Cardona; Debra L. Konkol; Charles Reidy; Michael E. Christe; Michael James Genin

The design, synthesis, and structure activity relationships for a novel series of indoles as potent, selective, thyroid hormone receptor β (TRβ) agonists is described. Compounds with >50× binding selectivity for TRβ over TRα were generated and evaluation of compound 1c from this series in a model of dyslipidemia demonstrated positive effects on plasma lipid endpoints in vivo.


Journal of Medicinal Chemistry | 2018

Discovery of LY3104607: A Potent and Selective G Protein-Coupled Receptor 40 (GPR40) Agonist with Optimized Pharmacokinetic Properties to Support Once Daily Oral Treatment in Patients with Type 2 Diabetes Mellitus

Chafiq Hamdouchi; Pranab Maiti; Alan M. Warshawsky; Amy C. DeBaillie; Keith A. Otto; Kelly L. Wilbur; Steven D. Kahl; Anjana Patel Lewis; Guemalli R. Cardona; Richard W. Zink; Keyue Chen; Siddaramaiah Cr; Jayana P. Lineswala; Grace L. Neathery; Cecilia Bouaichi; Benjamin A. Diseroad; Alison N. Campbell; Stephanie Ann Sweetana; Lisa A. Adams; Over Cabrera; Xiaosu Ma; Nathan Yumibe; Chahrzad Montrose-Rafizadeh; Yanyun Chen; Anne Reifel Miller

As a part of our program to identify potent GPR40 agonists capable of being dosed orally once daily in humans, we incorporated fused heterocycles into our recently disclosed spiropiperidine and tetrahydroquinoline acid derivatives 1, 2, and 3 with the intention of lowering clearance and improving the maximum absorbable dose (Dabs). Hypothesis-driven structural modifications focused on moving away from the zwitterion-like structure. and mitigating the N-dealkylation and O-dealkylation issues led to triazolopyridine acid derivatives with unique pharmacology and superior pharmacokinetic properties. Compound 4 (LY3104607) demonstrated functional potency and glucose-dependent insulin secretion (GDIS) in primary islets from rats. Potent, efficacious, and durable dose-dependent reductions in glucose levels were seen during glucose tolerance test (GTT) studies. Low clearance, volume of distribution, and high oral bioavailability were observed in all species. The combination of enhanced pharmacology and pharmacokinetic properties supported further development of this compound as a potential glucose-lowering drug candidate.


Endocrinology | 1999

Escape from the Acute Wolff-Chaikoff Effect Is Associated with a Decrease in Thyroid Sodium/Iodide Symporter Messenger Ribonucleic Acid and Protein

Peter H. K. Eng; Guemalli R. Cardona; Shih-Lieh Fang; Michael Previti; Sharon Alex; Nancy Carrasco; William W. Chin; Lewis E. Braverman

Collaboration


Dive into the Guemalli R. Cardona's collaboration.

Top Co-Authors

Avatar

William W. Chin

Singapore General Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Lan Ko

Eli Lilly and Company

View shared research outputs
Top Co-Authors

Avatar

Lewis E. Braverman

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge