Network


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

Hotspot


Dive into the research topics where Stacey J. Sukoff Rizzo is active.

Publication


Featured researches published by Stacey J. Sukoff Rizzo.


Science Translational Medicine | 2012

Negative Allosteric Modulation of the mGluR5 Receptor Reduces Repetitive Behaviors and Rescues Social Deficits in Mouse Models of Autism

Jill L. Silverman; Daniel G. Smith; Stacey J. Sukoff Rizzo; Michael N. Karras; Sarah M. Turner; Seda S. Tolu; Dianne K. Bryce; Deborah L. Smith; Kari R. Fonseca; Robert H. Ring; Jacqueline N. Crawley

Autism-like behaviors in mice were reversed by a negative modulator of a metabotropic glutamate receptor, suggesting a treatment for symptoms of autism spectrum disorders. Treatment of Autism Symptoms in Mice When they are 2 to 5 years old, children with autism start to show unusual social interactions and impaired communication. They may fail to develop relationships with their peers and be unable to interpret nuances of speech and body language. Most show repetitive motor behaviors and restricted interests and can have associated seizures, anxiety, or intellectual impairment. A large number of genes can put people at risk for this disorder, each in a small number of cases, and these genes point to connections between neurons as a vulnerable point in autism. Now, Silverman and colleagues have used two inbred strains of mice that display well-replicated behavioral abnormalities relevant to the diagnostic symptoms of autism and shown that some of these symptoms can be improved with a drug directed at a central glutamate receptor of the brain, mGluR5. The authors used two inbred strains of mice that display robust behaviors relevant to the diagnostic symptoms of autism. BTBR mice show deficits in many types of social interactions and high levels of repetitive self-grooming. C58 repetitively jumps. They used GRN-529, a compound developed by Pfizer that reduces the actions of glutamate, the main excitatory neurotransmitter in the brain. Other mGluR antagonists are showing promise in clinical trials for people with the fragile X mutation, who have both intellectual impairments and autism, so the authors reasoned that an mGluR5 compound might help autistic symptoms. GRN-529 reduced both the repetitive self-grooming in BTBR and the repetitive jumping in C58. Most intriguingly, GRN-529 also improved social behaviors in BTBR in two assays, one for social approach to an unfamiliar mouse and one for social interactions between freely moving pairs of mice. A particular strength of this study is that the authors replicated these beneficial actions of the mGluR5 compound in several separate groups of mice, in two laboratories. Although the path from target identification to effective human treatment is a long and winding road, the discovery of therapeutic efficacy for an mGluR5 negative allosteric modulator in both the repetitive and the social domains in two distinct mouse models is a promising beginning. This single biological target may offer a useful entry point to develop a pharmacological therapy that alleviates many symptoms of autism spectrum disorders. Neurodevelopmental disorders such as autism and fragile X syndrome were long thought to be medically untreatable, on the assumption that brain dysfunctions were immutably hardwired before diagnosis. Recent revelations that many cases of autism are caused by mutations in genes that control the ongoing formation and maturation of synapses have challenged this dogma. Antagonists of metabotropic glutamate receptor subtype 5 (mGluR5), which modulate excitatory neurotransmission, are in clinical trials for fragile X syndrome, a major genetic cause of intellectual disabilities. About 30% of patients with fragile X syndrome meet the diagnostic criteria for autism. Reasoning by analogy, we considered the mGluR5 receptor as a potential target for intervention in autism. We used BTBR T+tf/J (BTBR) mice, an established model with robust behavioral phenotypes relevant to the three diagnostic behavioral symptoms of autism—unusual social interactions, impaired communication, and repetitive behaviors—to probe the efficacy of a selective negative allosteric modulator of the mGluR5 receptor, GRN-529. GRN-529 reduced repetitive behaviors in three cohorts of BTBR mice at doses that did not induce sedation in control assays of open field locomotion. In addition, the same nonsedating doses reduced the spontaneous stereotyped jumping that characterizes a second inbred strain of mice, C58/J. Further, GRN-529 partially reversed the striking lack of sociability in BTBR mice on some parameters of social approach and reciprocal social interactions. These findings raise the possibility that a single targeted pharmacological intervention may alleviate multiple diagnostic behavioral symptoms of autism.


Neuropsychopharmacology | 2007

Increasing the Levels of Insulin-Like Growth Factor-I by an IGF Binding Protein Inhibitor Produces Anxiolytic and Antidepressant-Like Effects

Jessica Malberg; Brian Platt; Stacey J. Sukoff Rizzo; Robert H. Ring; Irwin Lucki; Lee E. Schechter; Sharon Rosenzweig-Lipson

The present studies were conducted to determine if increasing central levels of the neurotrophic factor insulin-like growth factor-1 (IGF-I) either directly or indirectly produces anxiolytic and antidepressant-like effects in the mouse. Central levels of IGF-I can be increased directly, by administering IGF-I, or indirectly by blocking the insulin-like growth factor binding proteins (IGFBPs). The IGFBP family has the unique ability to regulate IGF-I levels by sequestering IGF-I into an inactive complex. Therefore, an IGFBP inhibitor increases the level of IGF-I available to bind to its receptor. Intracerebroventricular (icv) administration of the nonspecific IGFBP inhibitor NBI-31772 (10–30 μg) increases the number of punished crossings in the four-plate test and NBI-31772 (0.3–10 μg) increases time spent in the open quadrant of the elevated zero maze (EZM), indicative of anxiolytic-like effects. NBI-31772 (3–30 μg) also decreases immobility time in the tail suspension test, indicative of antidepressant-like effects. Similarly, icv administration of IGF-I (0.1 μg) produces anxiolytic-like effects in the four-plate test and IGF-1 (0.3–1 μg) produces anxiolytic-like effects in the EZM. IGF-I (10 μg) also produces antidepressant-like effects in the tail suspension test. Coadministration of the IGF-I receptor antagonist JB1 with NBI-31772 or IGF-I blocks the anxiolytic-like and antidepressant-like effects of these compounds. These results suggest that NBI-31772 produces behavioral effects by increasing levels of IGF-I that in turn activate the IGF-I receptor. The present studies demonstrate that an IGFBP inhibitor mimics the behavioral effects of IGF-I and that IGFBP inhibition may represent a novel mechanism by which to increase IGF-I to treat depression and anxiety.


Journal of Pharmacology and Experimental Therapeutics | 2011

The Metabotropic Glutamate Receptor 7 Allosteric Modulator AMN082: A Monoaminergic Agent in Disguise?

Stacey J. Sukoff Rizzo; Sarah K. Leonard; Adam M. Gilbert; Paul Jeffrey Dollings; Deborah L. Smith; Mei-Yi Zhang; Li Di; Brian Platt; Sarah Neal; Jason M. Dwyer; Corey N. Bender; Jean Zhang; Tim Lock; Dianne Kowal; Angela Kramer; Andrew D. Randall; Christine Huselton; Karthick Vishwanathan; Susanna Y. Tse; John A. Butera; Robert H. Ring; Sharon Rosenzweig-Lipson; Zoë A. Hughes; John Dunlop

Metabotropic glutamate receptor 7 (mGluR7) remains the most elusive of the eight known mGluRs primarily because of the limited availability of tool compounds to interrogate its potential therapeutic utility. The discovery of N,N′-dibenzhydrylethane-1,2-diamine dihydrochloride (AMN082) as the first orally active, brain-penetrable, mGluR7-selective allosteric agonist by Mitsukawa and colleagues (Proc Natl Acad Sci USA 102:18712–18717, 2005) provides a means to investigate this receptor system directly. AMN082 demonstrates mGluR7 agonist activity in vitro and interestingly has a behavioral profile that supports utility across a broad spectrum of psychiatric disorders including anxiety and depression. The present studies were conducted to extend the in vitro and in vivo characterization of AMN082 by evaluating its pharmacokinetic and metabolite profile. Profiling of AMN082 in rat liver microsomes revealed rapid metabolism (t1/2 < 1 min) to a major metabolite, N-benzhydrylethane-1,2-diamine (Met-1). In vitro selectivity profiling of Met-1 demonstrated physiologically relevant transporter binding affinity at serotonin transporter (SERT), dopamine transporter (DAT), and norepinephrine transporter (NET) (323, 3020, and 3410 nM, respectively); whereas the parent compound AMN082 had appreciable affinity at NET (1385 nM). AMN082 produced antidepressant-like activity and receptor occupancy at SERT up to 4 h postdose, a time point at which AMN082 is significantly reduced in brain and plasma while the concentration of Met-1 continues to increase in brain. Acute Met-1 administration produced antidepressant-like activity as would be expected from its in vitro profile as a mixed SERT, NET, DAT inhibitor. Taken together, these data suggest that the reported in vivo actions of AMN082 should be interpreted with caution, because they may involve other mechanisms in addition to mGluR7.


PLOS ONE | 2013

Behavioral Characterization of A53T Mice Reveals Early and Late Stage Deficits Related to Parkinson’s Disease

Katrina L. Paumier; Stacey J. Sukoff Rizzo; Zdenek Berger; Yi Chen; Cathleen Gonzales; Edward Kaftan; Li Li; Susan M. Lotarski; Michael Monaghan; Wei Shen; Polina Stolyar; Dmytro Vasilyev; Margaret Zaleska; Warren D. Hirst; John Dunlop

Parkinsons disease (PD) pathology is characterized by the formation of intra-neuronal inclusions called Lewy bodies, which are comprised of alpha-synuclein (α-syn). Duplication, triplication or genetic mutations in α-syn (A53T, A30P and E46K) are linked to autosomal dominant PD; thus implicating its role in the pathogenesis of PD. In both PD patients and mouse models, there is increasing evidence that neuronal dysfunction occurs before the accumulation of protein aggregates (i.e., α-syn) and neurodegeneration. Characterization of the timing and nature of symptomatic dysfunction is important for understanding the impact of α-syn on disease progression. Furthermore, this knowledge is essential for identifying pathways and molecular targets for therapeutic intervention. To this end, we examined various functional and morphological endpoints in the transgenic mouse model expressing the human A53T α-syn variant directed by the mouse prion promoter at specific ages relating to disease progression (2, 6 and 12 months of age). Our findings indicate A53T mice develop fine, sensorimotor, and synaptic deficits before the onset of age-related gross motor and cognitive dysfunction. Results from open field and rotarod tests show A53T mice develop age-dependent changes in locomotor activity and reduced anxiety-like behavior. Additionally, digigait analysis shows these mice develop an abnormal gait by 12 months of age. A53T mice also exhibit spatial memory deficits at 6 and 12 months, as demonstrated by Y-maze performance. In contrast to gross motor and cognitive changes, A53T mice display significant impairments in fine- and sensorimotor tasks such as grooming, nest building and acoustic startle as early as 1–2 months of age. These mice also show significant abnormalities in basal synaptic transmission, paired-pulse facilitation and long-term depression (LTD). Combined, these data indicate the A53T model exhibits early- and late-onset behavioral and synaptic impairments similar to PD patients and may provide useful endpoints for assessing novel therapeutic interventions for PD.


Proceedings of the National Academy of Sciences of the United States of America | 2010

Phosphodiesterase 11A in brain is enriched in ventral hippocampus and deletion causes psychiatric disease-related phenotypes

Michele P. Kelly; Sheree F. Logue; Jonathon P. Day; Subha Lakkaraju; Lixin Jiang; Xiaotian Zhong; May Tam; Stacey J. Sukoff Rizzo; Brian Platt; Jason M. Dwyer; Sarah Neal; Virginia L. Pulito; Michael J. Agostino; Steven M. Grauer; Rachel Navarra; Cody Kelley; Thomas A. Comery; Richard J. Murrills; Miles D. Houslay; Nicholas J. Brandon

Phosphodiesterase 11A (PDE11A) is the most recently identified family of phosphodiesterases (PDEs), the only known enzymes to break down cyclic nucleotides. The tissue expression profile of this dual specificity PDE is controversial, and little is understood of its biological function, particularly in the brain. We seek here to determine if PDE11A is expressed in the brain and to understand its function, using PDE11A−/− knockout (KO) mice. We show that PDE11A mRNA and protein are largely restricted to hippocampus CA1, subiculum, and the amygdalohippocampal area, with a two- to threefold enrichment in the ventral vs. dorsal hippocampus, equal distribution between cytosolic and membrane fractions, and increasing levels of protein expression from postnatal day 7 through adulthood. Interestingly, PDE11A KO mice show subtle psychiatric-disease–related deficits, including hyperactivity in an open field, increased sensitivity to the glutamate N-methyl-D-aspartate receptor antagonist MK-801, as well as deficits in social behaviors (social odor recognition memory and social avoidance). In addition, PDE11A KO mice show enlarged lateral ventricles and increased activity in CA1 (as per increased Arc mRNA), phenotypes associated with psychiatric disease. The increased sensitivity to MK-801 exhibited by PDE11A KO mice may be explained by the biochemical dysregulation observed around the glutamate α-amino-3-hydroxy-5-methyl-4-isozazolepropionic (AMPA) receptor, including decreased levels of phosphorylated-GluR1 at Ser845 and the prototypical transmembrane AMPA-receptor–associated proteins stargazin (γ2) and γ8. Together, our data provide convincing evidence that PDE11A expression is restricted in the brain but plays a significant role in regulating brain function.


Journal of Pharmacology and Experimental Therapeutics | 2008

Correlating Efficacy in Rodent Cognition Models with in Vivo 5-Hydroxytryptamine1A Receptor Occupancy by a Novel Antagonist, (R)-N-(2-Methyl-(4-indolyl-1-piperazinyl)ethyl)-N-(2-pyridinyl)-cyclohexane Carboxamide (WAY-101405)

Warren D. Hirst; Terrance H. Andree; Suzan Aschmies; Wayne E. Childers; Thomas A. Comery; Lee A. Dawson; Mark L. Day; Irene B. Feingold; Steven M. Grauer; Boyd L. Harrison; Zoe A. Hughes; John Kao; Michael G. Kelly; Heidi van der Lee; Sharon Rosenzweig-Lipson; Annmarie Saab; Deborah L. Smith; Kelly Sullivan; Stacey J. Sukoff Rizzo; Cesario O. Tio; Mei-Yi Zhang; Lee E. Schechter

5-Hydroxytryptamine (5-HT)1A receptors play an important role in multiple cognitive processes, and compelling evidence suggests that 5-HT1A antagonists can reverse cognitive impairment. We have examined the therapeutic potential of a potent (Ki = 1.1 nM), selective (>100-fold), orally bioavailable, silent 5-HT1A receptor antagonist (KB = 1.3 nM) (R)-N-(2-methyl-(4-indolyl-1-piperazinyl)-ethyl)-N-(2-pyridinyl)-cyclohexane carboxamide (WAY-101405). Oral administration of WAY-101405 was shown to be effective in multiple rodent models of learning and memory. In a novel object recognition paradigm, 1 mg/kg enhanced retention (memory) for previously learned information, and it was able to reverse the memory deficits induced by scopolamine. WAY-101405 (1 mg/kg) was also able to reverse scopolamine-induced deficits in a rat contextual fear conditioning model. In the Morris water maze, WAY-101405 (3 mg/kg) significantly improved learning in a paradigm of increasing task difficulty. In vivo microdialysis studies in the dorsal hippocampus of freely moving adult rats demonstrated that acute administration of WAY-101405 (10 mg/kg) increased extracellular acetylcholine levels. The selective radioligand [3H]WAY-100635, administered i.v., was used for in vivo receptor occupancy studies, where WAY-101405 occupied 5-HT1A receptors in the rat cortex, with an ED50 value of 0.1 mg/kg p.o. Taken together, these studies demonstrate that WAY-101405 is a potent and selective, brain penetrant, orally bioavailable 5-HT1A receptor “silent” antagonist that is effective in preclinical memory paradigms at doses where approximately 90% of the postsynaptic 5-HT1A receptors are occupied. These results further support the rationale for use of this compound class in the treatment of cognitive dysfunction associated with psychiatric and neurological conditions.


American Journal of Human Genetics | 2016

Biallelic Mutations in PDE10A Lead to Loss of Striatal PDE10A and a Hyperkinetic Movement Disorder with Onset in Infancy

Christine P. Diggle; Stacey J. Sukoff Rizzo; Michael Popiolek; Reetta Hinttala; Jan-Philip Schülke; Manju A. Kurian; Ian M. Carr; Alexander F. Markham; David T. Bonthron; Christopher M. Watson; Saghira Malik Sharif; Veronica Reinhart; Larry C. James; Michelle Vanase-Frawley; Erik Charych; Melanie Allen; John F. Harms; Christopher J. Schmidt; Joanne Ng; Karen Pysden; Christine A. Strick; Päivi Vieira; Katariina Mankinen; Hannaleena Kokkonen; Matti Kallioinen; Raija Sormunen; Juha O. Rinne; Jarkko Johansson; Kati Alakurtti; Laura Huilaja

Deficits in the basal ganglia pathways modulating cortical motor activity underlie both Parkinson disease (PD) and Huntington disease (HD). Phosphodiesterase 10A (PDE10A) is enriched in the striatum, and animal data suggest that it is a key regulator of this circuitry. Here, we report on germline PDE10A mutations in eight individuals from two families affected by a hyperkinetic movement disorder due to homozygous mutations c.320A>G (p.Tyr107Cys) and c.346G>C (p.Ala116Pro). Both mutations lead to a reduction in PDE10A levels in recombinant cellular systems, and critically, positron-emission-tomography (PET) studies with a specific PDE10A ligand confirmed that the p.Tyr107Cys variant also reduced striatal PDE10A levels in one of the affected individuals. A knock-in mouse model carrying the homologous p.Tyr97Cys variant had decreased striatal PDE10A and also displayed motor abnormalities. Striatal preparations from this animal had an impaired capacity to degrade cyclic adenosine monophosphate (cAMP) and a blunted pharmacological response to PDE10A inhibitors. These observations highlight the critical role of PDE10A in motor control across species.


Journal of Biomolecular Screening | 2013

Future Viable Models of Psychiatry Drug Discovery in Pharma

Stacey J. Sukoff Rizzo; Jeremy R. Edgerton; Zoë A. Hughes; Nicholas J. Brandon

The unmet need for the treatment of disorders of the nervous system is growing, and as highlighted in the media and elsewhere, the results of an aging population will ensure this continues with an upward trajectory. Incredibly, the efforts within industry to identify new drugs to treat these conditions have seemingly disappeared despite the growing need. There has been a run of extraordinary failure in the later stages of the drug discovery process for neurological and psychiatric disorders, which has many causes. We believe, though, that we have to confront this dire situation, both by using learnings from the post hoc analysis of our historical failure, as well as harnessing the bewildering array of new technologies and data now available to us, to ensure we are making the right decisions along the very complicated path of drug discovery to registration.


Current protocols in mouse biology | 2016

Methodological Considerations for Optimizing and Validating Behavioral Assays.

Stacey J. Sukoff Rizzo; Jill L. Silverman

Preclinical animal models are indispensable tools for translational research for which behavioral characterization and phenotyping are essential to testing hypotheses and for evaluating the potential of novel therapeutic agents to treat diseases. The methods employed for comprehensive behavioral phenotyping and pharmacological experiments are complex and should be conducted exclusively by trained technicians with demonstrated proficiency. The ultimate goal is to identify disease‐relevant and translational behavioral endpoints that are robust, reliable, and reproducible, and that can be employed to evaluate potential of novel therapeutic agents to treat disease. The intent of the present article is to provide a pragmatic outline for establishing and optimizing behavioral assays and phenotyping batteries, ensuring that the assays and the data are reliable such that they can be reproduced within and across technicians and laboratories and, more importantly, that the data is translatable to the clinic.


Annual Review of Animal Biosciences | 2017

Behavioral Phenotyping Assays for Genetic Mouse Models of Neurodevelopmental, Neurodegenerative, and Psychiatric Disorders

Stacey J. Sukoff Rizzo; Jacqueline N. Crawley

Animal models offer heuristic research tools to understand the causes of human diseases and to identify potential treatments. With rapidly evolving genetic engineering technologies, mutations identified in a human disorder can be generated in the mouse genome. Phenotypic outcomes of the mutation are then explicated to confirm hypotheses about causes and to discover effective therapeutics. Most neurodevelopmental, neurodegenerative, and psychiatric disorders are diagnosed primarily by their prominent behavioral symptoms. Mouse behavioral assays analogous to the human symptoms have been developed to analyze the consequences of mutations and to evaluate proposed therapeutics preclinically. Here we describe the range of mouse behavioral tests available in the established behavioral neuroscience literature, along with examples of their translational applications. Concepts presented have been successfully used in other species, including flies, worms, fish, rats, pigs, and nonhuman primates. Identical strategies can be employed to test hypotheses about environmental causes and gene × environment interactions.

Collaboration


Dive into the Stacey J. Sukoff Rizzo's collaboration.

Top Co-Authors

Avatar
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