Network


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

Hotspot


Dive into the research topics where Leticia Toledo-Sherman is active.

Publication


Featured researches published by Leticia Toledo-Sherman.


Nature Chemical Biology | 2015

The promise and peril of chemical probes

C.H. Arrowsmith; James E. Audia; Christopher M. Austin; Jonathan B. Baell; Jonathan Bennett; Julian Blagg; C. Bountra; Paul E. Brennan; Peter J. Brown; Mark Edward Bunnage; Carolyn Buser-Doepner; Robert M. Campbell; Adrian Carter; Philip Cohen; Robert A. Copeland; Ben Cravatt; Jayme L. Dahlin; Dashyant Dhanak; A. Edwards; Mathias Frederiksen; Stephen V. Frye; Nathanael S. Gray; Charles E. Grimshaw; David Hepworth; Trevor Howe; Kilian Huber; Jian Jin; Stefan Knapp; Joanne Kotz; Ryan G. Kruger

Chemical probes are powerful reagents with increasing impacts on biomedical research. However, probes of poor quality or that are used incorrectly generate misleading results. To help address these shortcomings, we will create a community-driven wiki resource to improve quality and convey current best practice.


Journal of Medicinal Chemistry | 2012

Discovery and Structure–Activity Relationship of Potent and Selective Covalent Inhibitors of Transglutaminase 2 for Huntington’s Disease

Michael Prime; Ole Andreas Andersen; John J. Barker; Mark Brooks; Robert K. Y. Cheng; Ian Toogood-Johnson; Stephen Martin Courtney; Frederick Arthur Brookfield; Christopher John Yarnold; Richard W. Marston; Peter Johnson; Siw Johnsen; Jordan J. Palfrey; Darshan Vaidya; Sayeh Erfan; Osamu Ichihara; Brunella Felicetti; Shilpa Palan; Anna Pedret-Dunn; Sabine Schaertl; Ina Sternberger; Andreas Ebneth; Andreas Scheel; Dirk Winkler; Leticia Toledo-Sherman; Maria Beconi; Douglas Macdonald; Ignacio Munoz-Sanjuan; Celia Dominguez; John Wityak

Tissue transglutaminase 2 (TG2) is a multifunctional protein primarily known for its calcium-dependent enzymatic protein cross-linking activity via isopeptide bond formation between glutamine and lysine residues. TG2 overexpression and activity have been found to be associated with Huntingtons disease (HD); specifically, TG2 is up-regulated in the brains of HD patients and in animal models of the disease. Interestingly, genetic deletion of TG2 in two different HD mouse models, R6/1 and R6/2, results in improved phenotypes including a reduction in neuronal death and prolonged survival. Starting with phenylacrylamide screening hit 7d, we describe the SAR of this series leading to potent and selective TG2 inhibitors. The suitability of the compounds as in vitro tools to elucidate the biology of TG2 was demonstrated through mode of inhibition studies, characterization of druglike properties, and inhibition profiles in a cell lysate assay.


Journal of Medicinal Chemistry | 2015

Development of a Series of Aryl Pyrimidine Kynurenine Monooxygenase Inhibitors as Potential Therapeutic Agents for the Treatment of Huntington's Disease

Leticia Toledo-Sherman; Michael Prime; Ladislav Mrzljak; Maria Beconi; Alan Beresford; Frederick Arthur Brookfield; Christopher John Brown; Isabell Cardaun; Stephen Martin Courtney; Ulrike Dijkman; Estelle Hamelin-Flegg; Peter Johnson; Valerie Kempf; Kathy Lyons; Kimberly Matthews; William Leonard Mitchell; Catherine O’Connell; Paula Pena; Kendall Powell; Arash Rassoulpour; Laura Reed; Wolfgang Reindl; Suganathan Selvaratnam; Weslyn Ward Friley; Derek Weddell; Naomi Went; Patricia Wheelan; Christin Winkler; Dirk Winkler; John Wityak

We report on the development of a series of pyrimidine carboxylic acids that are potent and selective inhibitors of kynurenine monooxygenase and competitive for kynurenine. We describe the SAR for this novel series and report on their inhibition of KMO activity in biochemical and cellular assays and their selectivity against other kynurenine pathway enzymes. We describe the optimization process that led to the identification of a program lead compound with a suitable ADME/PK profile for therapeutic development. We demonstrate that systemic inhibition of KMO in vivo with this lead compound provides pharmacodynamic evidence for modulation of kynurenine pathway metabolites both in the periphery and in the central nervous system.


Drug Metabolism and Disposition | 2012

Metabolism and Pharmacokinetics of JM6 in Mice: JM6 Is Not a Prodrug for Ro-61-8048

Maria Beconi; Dawn Yates; Kathryn Lyons; Kim Matthews; Steve Clifton; Tania Mead; Michael Prime; Dirk Winkler; Catherine O'Connell; Daryl Walter; Leticia Toledo-Sherman; Ignacio Munoz-Sanjuan; Celia Dominguez

Understanding whether regulation of tryptophan metabolites can ameliorate neurodegeneration is of high interest to investigators. A recent publication describes 3,4-dimethoxy-N-(4-(3-nitrophenyl)-5-(piperidin-1-ylmethyl)thiazol-2-yl)benzenesulfonamide (JM6) as a novel prodrug for the kynurenine 3-monooxygenase (KMO) inhibitor 3,4-dimethoxy-N-(4-(3-nitrophenyl)thiazol-2-yl)benzenesulfonamide (Ro-61-8048) that elicits therapeutic effects in mouse models of Huntingtons and Alzheimers diseases (Cell 145:863–874, 2011). Our evaluation of the metabolism and pharmacokinetics of JM6 and Ro-61-8048 indicate instead that Ro-61-8048 concentrations in mouse plasma after JM6 administration originate from a Ro-61-8048 impurity (<0.1%) in JM6. After a 0.05 mg/kg Ro-61-8048 oral dose alone or coadministered with 10 mg/kg JM6 to mice, the Ro-61-8048 areas under the concentration-time curves (AUCs) from 0 to infinity were similar (4300 and 4900 nM × h, respectively), indicating no detectable contributions of JM6 metabolism to the Ro-61-8048 AUCs. JM6 was stable in incubations under acidic conditions and Ro-61-8048 was not a product of JM6 metabolism in vitro (plasma, blood, or hepatic models). Species differences in the quantitative rate of oxidative metabolism indicate that major circulating JM6 metabolite(s) in mice are unlikely to be major in humans: JM6 is rapidly metabolized via the piperidyl moiety in mouse (forming an iminium ion reactive intermediate) but is slowly metabolized in human (in vitro), primarily via O-dealkylation at the phenyl ring. Our data indicate that JM6 is not a prodrug for Ro-61-8048 and is not a potent KMO inhibitor.


ACS Medicinal Chemistry Letters | 2012

Irreversible 4-Aminopiperidine Transglutaminase 2 Inhibitors for Huntington's Disease.

Michael Prime; Frederick Arthur Brookfield; Stephen Martin Courtney; Simon Gaines; Richard W. Marston; Osamu Ichihara; Marie Li; Darshan Vaidya; Helen Williams; Anna Pedret-Dunn; Laura Reed; Sabine Schaertl; Leticia Toledo-Sherman; Maria Beconi; Douglas Macdonald; Ignacio Munoz-Sanjuan; Celia Dominguez; John Wityak

A new series of potent TG2 inhibitors are reported that employ a 4-aminopiperidine core bearing an acrylamide warhead. We establish the structure-activity relationship of this new series and report on the transglutaminase selectivity and in vitro ADME properties of selected compounds. We demonstrate that the compounds do not conjugate glutathione in an in vitro setting and have superior plasma stability over our previous series.


Experimental Neurology | 2016

The novel KMO inhibitor CHDI-340246 leads to a restoration of electrophysiological alterations in mouse models of Huntington's disease.

Vahri Beaumont; Ladislav Mrzljak; Ulrike Dijkman; Robert Freije; Mariette Heins; Arash Rassoulpour; Geoffrey Tombaugh; Simon Gelman; Amyaouch Bradaia; Esther Steidl; Melanie Gleyzes; Taneli Heikkinen; Kimmo Lehtimäki; Jukka Puoliväli; Outi Kontkanen; Robyn M. Javier; Ioana Neagoe; Heike Deisemann; Dirk Winkler; Andreas Ebneth; Vinod Khetarpal; Leticia Toledo-Sherman; Celia Dominguez; Larry Park; Ignacio Munoz-Sanjuan

Dysregulation of the kynurenine (Kyn) pathway has been associated with the progression of Huntingtons disease (HD). In particular, elevated levels of the kynurenine metabolites 3-hydroxy kynurenine (3-OH-Kyn) and quinolinic acid (Quin), have been reported in the brains of HD patients as well as in rodent models of HD. The production of these metabolites is controlled by the activity of kynurenine mono-oxygenase (KMO), an enzyme which catalyzes the synthesis of 3-OH-Kyn from Kyn. In order to determine the role of KMO in the phenotype of mouse models of HD, we have developed a potent and selective KMO inhibitor termed CHDI-340246. We show that this compound, when administered orally to transgenic mouse models of HD, potently and dose-dependently modulates the Kyn pathway in peripheral tissues and in the central nervous system. The administration of CHDI-340246 leads to an inhibition of the formation of 3-OH-Kyn and Quin, and to an elevation of Kyn and Kynurenic acid (KynA) levels in brain tissues. We show that administration of CHDI-340246 or of Kyn and of KynA can restore several electrophysiological alterations in mouse models of HD, both acutely and after chronic administration. However, using a comprehensive panel of behavioral tests, we demonstrate that the chronic dosing of a selective KMO inhibitor does not significantly modify behavioral phenotypes or natural progression in mouse models of HD.


Journal of Biomolecular Screening | 2013

Development of LC/MS/MS, High-Throughput Enzymatic and Cellular Assays for the Characterization of Compounds That Inhibit Kynurenine Monooxygenase (KMO)

Dirk Winkler; Maria Beconi; Leticia Toledo-Sherman; Michael Prime; Andreas Ebneth; Celia Dominguez; Ignacio Munoz-Sanjuan

Kynurenine monooxygenase (KMO) catalyzes the conversion of kynurenine to 3-hydroxykynurenine. Modulation of KMO activity has been implicated in several neurodegenerative diseases, including Huntington disease. Our goal is to develop potent and selective small-molecule KMO inhibitors with suitable pharmacokinetic characteristics for in vivo proof-of-concept studies and subsequent clinical development. We developed a comprehensive panel of biochemical and cell-based assays that use liquid chromatography/tandem mass spectrometry to quantify unlabeled kynurenine and 3-hydroxykynurenine. We describe assays to measure KMO inhibition in cell and tissue extracts, as well as cellular assays including heterologous cell lines and primary rat microglia and human peripheral blood mononuclear cells.


Archives of Biochemistry and Biophysics | 2017

Nuclear factor (erythroid-derived 2)-like 2 (NRF2) drug discovery: Biochemical toolbox to develop NRF2 activators by reversible binding of Kelch-like ECH-associated protein 1 (KEAP1)

Alberto Bresciani; Antonino Missineo; Mariana Gallo; Mauro Cerretani; Paola Fezzardi; Licia Tomei; Daniel O. Cicero; Sergio Altamura; Alessia Santoprete; Raffaele Ingenito; Elisabetta Bianchi; Robert Pacifici; Celia Dominguez; Ignacio Munoz-Sanjuan; Steven Harper; Leticia Toledo-Sherman; Larry Park

Mechanisms that activate innate antioxidant responses, as a way to mitigate oxidative stress at the site of action, hold much therapeutic potential in diseases, such as Parkinsons disease, Alzheimers disease and Huntingtons disease, where the use of antioxidants as monotherapy has not yielded positive results. The nuclear factor NRF2 is a transcription factor whose activity upregulates the expression of cell detoxifying enzymes in response to oxidative stress. NRF2 levels are modulated by KEAP1, a sensor of oxidative stress. KEAP1 binds NRF2 and facilitates its ubiquitination and subsequent degradation. Recently, compounds that reversibly disrupt the NRF2-KEAP1 interaction have been described, opening the field to a new era of safer NRF2 activators. This paper describes a set of new, robust and informative biochemical assays that enable the selection and optimization of non-covalent KEAP1 binders. These include a time-resolved fluorescence resonance energy transfer (TR-FRET) primary assay with high modularity and robustness, a surface plasmon resonance (SPR) based KEAP1 direct binding assay that enables the quantification and analysis of full kinetic binding parameters and finally a 1H-15N heteronuclear single quantum coherence (HSQC) NMR assay suited to study the interaction surface of KEAP1 with residue-specific information to validate the interaction of ligands in the KEAP1 binding site.


Journal of Pharmaceutical and Biomedical Analysis | 2015

A single-run liquid chromatography mass spectrometry method to quantify neuroactive kynurenine pathway metabolites in rat plasma.

Laura Orsatti; Roberto Speziale; Maria Vittoria Orsale; Fulvia Caretti; Maria Veneziano; Matteo Zini; Edith Monteagudo; Kathryn A. Lyons; Maria Beconi; Kelvin Chan; Todd Herbst; Leticia Toledo-Sherman; Ignacio Munoz-Sanjuan; Fabio Bonelli; Celia Dominguez

Neuroactive metabolites in the kynurenine pathway of tryptophan catabolism are associated with neurodegenerative disorders. Tryptophan is transported across the blood-brain barrier and converted via the kynurenine pathway to N-formyl-L-kynurenine, which is further degraded to L-kynurenine. This metabolite can then generate a group of metabolites called kynurenines, most of which have neuroactive properties. The association of tryptophan catabolic pathway alterations with various central nervous system (CNS) pathologies has raised interest in analytical methods to accurately quantify kynurenines in body fluids. We here describe a rapid and sensitive reverse-phase HPLC-MS/MS method to quantify L-kynurenine (KYN), kynurenic acid (KYNA), 3-hydroxy-L-kynurenine (3HK) and anthranilic acid (AA) in rat plasma. Our goal was to quantify these metabolites in a single run; given their different physico-chemical properties, major efforts were devoted to develop a chromatography suitable for all metabolites that involves plasma protein precipitation with acetonitrile followed by chromatographic separation by C18 RP chromatography, detected by electrospray mass spectrometry. Quantitation range was 0.098-100 ng/ml for 3HK, 9.8-20,000 ng/ml for KYN, 0.49-1000 ng/ml for KYNA and AA. The method was linear (r>0.9963) and validation parameters were within acceptance range (calibration standards and QC accuracy within ±30%).


Journal of Medicinal Chemistry | 2015

Lead Optimization toward Proof-of-Concept Tools for Huntington’s Disease within a 4-(1H-Pyrazol-4-yl)pyrimidine Class of Pan-JNK Inhibitors

John Wityak; Kevin McGee; Michael Conlon; Ren Hua Song; Bryan Cordell Duffy; Brent Clayton; Michael P. Lynch; Gwen Wang; Emily Freeman; James C. Haber; Douglas B. Kitchen; David D. Manning; Jiffry Ismail; Yuri L. Khmelnitsky; Peter C. Michels; Jeff Webster; Macarena Irigoyen; Michele Luche; Monica Hultman; Mei Bai; IokTeng D. Kuok; Ryan Newell; Marieke Lamers; Philip M. Leonard; Dawn Yates; Kim L. Matthews; Lynette Ongeri; Steve Clifton; Tania Mead; Susan Deupree

Through medicinal chemistry lead optimization studies focused on calculated properties and guided by X-ray crystallography and computational modeling, potent pan-JNK inhibitors were identified that showed submicromolar activity in a cellular assay. Using in vitro ADME profiling data, 9t was identified as possessing favorable permeability and a low potential for efflux, but it was rapidly cleared in liver microsomal incubations. In a mouse pharmacokinetics study, compound 9t was brain-penetrant after oral dosing, but exposure was limited by high plasma clearance. Brain exposure at a level expected to support modulation of a pharmacodynamic marker in mouse was achieved when the compound was coadministered with the pan-cytochrome P450 inhibitor 1-aminobenzotriazole.

Collaboration


Dive into the Leticia Toledo-Sherman's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Cheryl Arrowsmith

University of Oklahoma Health Sciences Center

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