Network


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

Hotspot


Dive into the research topics where Maria F. Sassano is active.

Publication


Featured researches published by Maria F. Sassano.


Nature | 2012

Automated design of ligands to polypharmacological profiles

Jérémy Besnard; Gian Filippo Ruda; Vincent Setola; Keren Abecassis; Ramona M. Rodriguiz; Xi Ping Huang; Suzanne Norval; Maria F. Sassano; Antony I. Shin; Lauren A. Webster; Frederick R. C. Simeons; Laste Stojanovski; Annik Prat; Nabil G. Seidah; Daniel B. Constam; G. Richard Bickerton; Kevin D. Read; William C. Wetsel; Ian H. Gilbert; Bryan L. Roth; Andrew L. Hopkins

The clinical efficacy and safety of a drug is determined by its activity profile across many proteins in the proteome. However, designing drugs with a specific multi-target profile is both complex and difficult. Therefore methods to design drugs rationally a priori against profiles of several proteins would have immense value in drug discovery. Here we describe a new approach for the automated design of ligands against profiles of multiple drug targets. The method is demonstrated by the evolution of an approved acetylcholinesterase inhibitor drug into brain-penetrable ligands with either specific polypharmacology or exquisite selectivity profiles for G-protein-coupled receptors. Overall, 800 ligand–target predictions of prospectively designed ligands were tested experimentally, of which 75% were confirmed to be correct. We also demonstrate target engagement in vivo. The approach can be a useful source of drug leads when multi-target profiles are required to achieve either selectivity over other drug targets or a desired polypharmacology.


Proceedings of the National Academy of Sciences of the United States of America | 2011

Discovery of β-Arrestin–Biased Dopamine D2 Ligands for Probing Signal Transduction Pathways Essential for Antipsychotic Efficacy

John A. Allen; Julianne M. Yost; Vincent Setola; Xin Chen; Maria F. Sassano; Meng Chen; Sean M. Peterson; Prem N. Yadav; Xi Ping Huang; Bo Feng; Niels H. Jensen; Xin Che; Xu Bai; Stephen V. Frye; William C. Wetsel; Marc G. Caron; Jonathan A. Javitch; Bryan L. Roth; Jian Jin

Elucidating the key signal transduction pathways essential for both antipsychotic efficacy and side-effect profiles is essential for developing safer and more effective therapies. Recent work has highlighted noncanonical modes of dopamine D2 receptor (D2R) signaling via β-arrestins as being important for the therapeutic actions of both antipsychotic and antimanic agents. We thus sought to create unique D2R agonists that display signaling bias via β-arrestin–ergic signaling. Through a robust diversity-oriented modification of the scaffold represented by aripiprazole (1), we discovered UNC9975 (2), UNC0006 (3), and UNC9994 (4) as unprecedented β-arrestin–biased D2R ligands. These compounds also represent unprecedented β-arrestin–biased ligands for a Gi-coupled G protein–coupled receptor (GPCR). Significantly, UNC9975, UNC0006, and UNC9994 are simultaneously antagonists of Gi-regulated cAMP production and partial agonists for D2R/β-arrestin-2 interactions. Importantly, UNC9975 displayed potent antipsychotic-like activity without inducing motoric side effects in inbred C57BL/6 mice in vivo. Genetic deletion of β-arrestin-2 simultaneously attenuated the antipsychotic actions of UNC9975 and transformed it into a typical antipsychotic drug with a high propensity to induce catalepsy. Similarly, the antipsychotic-like activity displayed by UNC9994, an extremely β-arrestin–biased D2R agonist, in wild-type mice was completely abolished in β-arrestin-2 knockout mice. Taken together, our results suggest that β-arrestin signaling and recruitment can be simultaneously a significant contributor to antipsychotic efficacy and protective against motoric side effects. These functionally selective, β-arrestin–biased D2R ligands represent valuable chemical probes for further investigations of D2R signaling in health and disease.


Nature | 2016

Structure-based discovery of opioid analgesics with reduced side effects

Aashish Manglik; Henry Lin; Dipendra K. Aryal; John D. McCorvy; Daniela Dengler; Gregory Corder; Anat Levit; Ralf C. Kling; Viachaslau Bernat; Harald Hübner; Xi-Ping Huang; Maria F. Sassano; Patrick M. Giguère; Stefan Löber; Da Duan; Grégory Scherrer; Brian K. Kobilka; Peter Gmeiner; Bryan L. Roth; Brian K. Shoichet

Morphine is an alkaloid from the opium poppy used to treat pain. The potentially lethal side effects of morphine and related opioids—which include fatal respiratory depression—are thought to be mediated by μ-opioid-receptor (μOR) signalling through the β-arrestin pathway or by actions at other receptors. Conversely, G-protein μOR signalling is thought to confer analgesia. Here we computationally dock over 3 million molecules against the μOR structure and identify new scaffolds unrelated to known opioids. Structure-based optimization yields PZM21—a potent Gi activator with exceptional selectivity for μOR and minimal β-arrestin-2 recruitment. Unlike morphine, PZM21 is more efficacious for the affective component of analgesia versus the reflexive component and is devoid of both respiratory depression and morphine-like reinforcing activity in mice at equi-analgesic doses. PZM21 thus serves as both a probe to disentangle μOR signalling and a therapeutic lead that is devoid of many of the side effects of current opioids.


Nature Methods | 2013

A pharmacological organization of G protein–coupled receptors

Henry Lin; Maria F. Sassano; Bryan L. Roth; Brian K. Shoichet

Protein classification typically uses structural, sequence or functional similarity. Here we introduce an orthogonal method that organizes proteins by ligand similarity, focusing on the class A G-protein–coupled receptor (GPCR) protein family. Comparing a ligand-based dendrogram to a sequence-based one, we identified GPCRs that were distantly linked by sequence but were neighbors by ligand similarity. Experimental testing of the ligands predicted to link three of these new pairs confirmed the predicted association, with potencies ranging from low nanomolar to low micromolar. We also predicted hundreds of non-GPCRs closely related to GPCRs by ligand similarity and confirmed several cases experimentally. Ligand similarities among these targets may reflect the conservation of identical ligands among unrelated receptors, which signal in different time domains. Our method integrates these apparently disparate receptors into chemically coherent circuits and suggests which of these receptors may be targeted by individual ligands.


ACS Chemical Biology | 2013

Conformation guides molecular efficacy in docking screens of activated β-2 adrenergic G protein coupled receptor

Dahlia R. Weiss; Seungkirl Ahn; Maria F. Sassano; Andrew Kleist; Xiao Zhu; Ryan T. Strachan; Bryan L. Roth; Robert J. Lefkowitz; Brian K. Shoichet

A prospective, large library virtual screen against an activated β2-adrenergic receptor (β2AR) structure returned potent agonists to the exclusion of inverse-agonists, providing the first complement to the previous virtual screening campaigns against inverse-agonist-bound G protein coupled receptor (GPCR) structures, which predicted only inverse-agonists. In addition, two hits recapitulated the signaling profile of the co-crystal ligand with respect to the G protein and arrestin mediated signaling. This functional fidelity has important implications in drug design, as the ability to predict ligands with predefined signaling properties is highly desirable. However, the agonist-bound state provides an uncertain template for modeling the activated conformation of other GPCRs, as a dopamine D2 receptor (DRD2) activated model templated on the activated β2AR structure returned few hits of only marginal potency.


Journal of Medicinal Chemistry | 2013

Colloidal Aggregation Causes Inhibition of G Protein-Coupled Receptors

Maria F. Sassano; Allison K. Doak; Bryan L. Roth; Brian K. Shoichet

Colloidal aggregation is the dominant mechanism for artifactual inhibition of soluble proteins, and controls against it are now widely deployed. Conversely, investigating this mechanism for membrane-bound receptors has proven difficult. Here we investigate the activity of four well-characterized aggregators against three G protein-coupled receptors (GPCRs) recognizing peptide and protein ligands. Each of the aggregators was active at micromolar concentrations against the three GPCRs in cell-based assays. This activity could be attenuated by either centrifugation of the inhibitor stock solution or by addition of Tween-80 detergent. In the absence of agonist, the aggregators acted as inverse agonists, consistent with a direct receptor interaction. Meanwhile, several literature GPCR ligands that resemble aggregators themselves formed colloids, by both physical and enzymological tests. These observations suggest that some GPCRs may be artifactually antagonized by colloidal aggregates, an effect that merits the attention of investigators in this field.


ACS Medicinal Chemistry Letters | 2013

Discovery of β2 Adrenergic Receptor Ligands Using Biosensor Fragment Screening of Tagged Wild-Type Receptor

Tonia Aristotelous; Seungkirl Ahn; Arun K. Shukla; Sylwia Gawron; Maria F. Sassano; Alem W. Kahsai; Laura M. Wingler; Xiao Zhu; Xi Ping Huang; Jennifer Riley; Jérémy Besnard; Kevin D. Read; Bryan L. Roth; Ian H. Gilbert; Andrew L. Hopkins; Robert J. Lefkowitz; Iva Navratilova

G-protein coupled receptors (GPCRs) are the primary target class of currently marketed drugs, accounting for about a quarter of all drug targets of approved medicines. However, almost all the screening efforts for novel ligand discovery rely exclusively on cellular systems overexpressing the receptors. An alternative ligand discovery strategy is a fragment-based drug discovery, where low molecular weight compounds, known as fragments, are screened as initial starting points for optimization. However, the screening of fragment libraries usually employs biophysical screening methods, and as such, it has not been routinely applied to membrane proteins. We present here a surface plasmon resonance biosensor approach that enables, cell-free, label-free, fragment screening that directly measures fragment interactions with wild-type GPCRs. We exemplify the method by the discovery of novel, selective, high affinity antagonists of human β2 adrenoceptor.


Structure | 2014

The TLQP-21 Peptide Activates the G-Protein-Coupled Receptor C3aR1 via a Folding-upon-Binding Mechanism.

Cheryl Cero; Vitaly V. Vostrikov; Raffaello Verardi; Cinzia Severini; T. Gopinath; Patrick D. Braun; Maria F. Sassano; Allison Gurney; Bryan L. Roth; Lucy Vulchanova; Roberta Possenti; Gianluigi Veglia; Alessandro Bartolomucci

TLQP-21, a VGF-encoded peptide is emerging as a novel target for obesity-associated disorders. TLQP-21 is found in the sympathetic nerve terminals in the adipose tissue and targets the G-protein-coupled receptor complement-3a receptor1 (C3aR1). The mechanisms of TLQP-21-induced receptor activation remain unexplored. Here, we report that TLQP-21 is intrinsically disordered and undergoes a disorder-to-order transition, adopting an α-helical conformation upon targeting cells expressing the C3aR1. We determined that the hot spots for TLQP-21 are located at the C terminus, with mutations in the last four amino acids progressively reducing the bioactivity and, a single site mutation (R21A) or C-terminal amidation abolishing its function completely. Additionally, the human TLQP-21 sequence carrying a S20A substitution activates the human C3aR1 receptor with lower potency compared to the rodent sequence. These studies reveal the mechanism of action of TLQP-21 and provide molecular templates for designing agonists and antagonists to modulate C3aR1 functions.


PLOS Biology | 2013

In Silico Molecular Comparisons of C. elegans and Mammalian Pharmacology Identify Distinct Targets That Regulate Feeding

George A. Lemieux; Michael J. Keiser; Maria F. Sassano; Christian Laggner; Fahima Mayer; Roland J. Bainton; Zena Werb; Bryan L. Roth; Brian K. Shoichet; Kaveh Ashrafi

This paper takes advantage of similarities between the C. elegans and human pharmacopeia to identify and validate pharmacological targets that regulate C. elegans feeding rates.


PLOS ONE | 2016

G Protein Coupled Receptor Kinase 3 Regulates Breast Cancer Migration, Invasion, and Metastasis

Matthew J. Billard; David J. Fitzhugh; Joel S. Parker; Jaime M. Brozowski; Marcus W. McGinnis; Roman G. Timoshchenko; D. Stephen Serafin; Ruth A. Lininger; Nancy Klauber-DeMore; G. Gary Sahagian; Young K. Truong; Maria F. Sassano; Jonathan S. Serody; Teresa K. Tarrant

Triple negative breast cancer (TNBC) is a heterogeneous disease that has a poor prognosis and limited treatment options. Chemokine receptor interactions are important modulators of breast cancer metastasis; however, it is now recognized that quantitative surface expression of one important chemokine receptor, CXCR4, may not directly correlate with metastasis and that its functional activity in breast cancer may better inform tumor pathogenicity. G protein coupled receptor kinase 3 (GRK3) is a negative regulator of CXCR4 activity, and we show that GRK expression correlates with tumorigenicity, molecular subtype, and metastatic potential in human tumor microarray analysis. Using established human breast cancer cell lines and an immunocompetent in vivo mouse model, we further demonstrate that alterations in GRK3 expression levels in tumor cells directly affect migration and invasion in vitro and the establishment of distant metastasis in vivo. The effects of GRK3 modulation appear to be specific to chemokine-mediated migration behaviors without influencing tumor cell proliferation or survival. These data demonstrate that GRK3 dysregulation may play an important part in TNBC metastasis.

Collaboration


Dive into the Maria F. Sassano's collaboration.

Top Co-Authors

Avatar

Bryan L. Roth

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Vincent Setola

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Xi Ping Huang

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Jian Jin

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

John A. Allen

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Wesley K. Kroeze

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge