Network


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

Hotspot


Dive into the research topics where Wen-Ning Zhao is active.

Publication


Featured researches published by Wen-Ning Zhao.


Journal of the American Chemical Society | 2010

An aldol-based build/couple/pair strategy for the synthesis of medium- and large-sized rings: discovery of macrocyclic histone deacetylase inhibitors.

Lisa A. Marcaurelle; Eamon Comer; Sivaraman Dandapani; Jeremy R. Duvall; Baudouin Gerard; Sarathy Kesavan; Maurice D. Lee; Haibo Liu; Jason T. Lowe; Jean-Charles Marie; Carol Mulrooney; Bhaumik A. Pandya; Ann Rowley; Troy D. Ryba; Byung-Chul Suh; Jingqiang Wei; Damian W. Young; Lakshmi B. Akella; Nathan T. Ross; Yan-Ling Zhang; Daniel M. Fass; Surya A. Reis; Wen-Ning Zhao; Stephen J. Haggarty; Michelle Palmer; Michael A. Foley

An aldol-based build/couple/pair (B/C/P) strategy was applied to generate a collection of stereochemically and skeletally diverse small molecules. In the build phase, a series of asymmetric syn- and anti-aldol reactions were performed to produce four stereoisomers of a Boc-protected γ-amino acid. In addition, both stereoisomers of O-PMB-protected alaninol were generated to provide a chiral amine coupling partner. In the couple step, eight stereoisomeric amides were synthesized by coupling the chiral acid and amine building blocks. The amides were subsequently reduced to generate the corresponding secondary amines. In the pair phase, three different reactions were employed to enable intramolecular ring-forming processes: nucleophilic aromatic substitution (S(N)Ar), Huisgen [3+2] cycloaddition, and ring-closing metathesis (RCM). Despite some stereochemical dependencies, the ring-forming reactions were optimized to proceed with good to excellent yields, providing a variety of skeletons ranging in size from 8- to 14-membered rings. Scaffolds resulting from the RCM pairing reaction were diversified on the solid phase to yield a 14 400-membered library of macrolactams. Screening of this library led to the discovery of a novel class of histone deacetylase inhibitors, which display mixed enzyme inhibition, and led to increased levels of acetylation in a primary mouse neuron culture. The development of stereo-structure/activity relationships was made possible by screening all 16 stereoisomers of the macrolactams produced through the aldol-based B/C/P strategy.


PLOS ONE | 2013

A Selective HDAC 1/2 Inhibitor Modulates Chromatin and Gene Expression in Brain and Alters Mouse Behavior in Two Mood-Related Tests

Frederick A. Schroeder; Michael C. Lewis; Daniel M. Fass; Florence F. Wagner; Yan-Ling Zhang; Krista M. Hennig; Jennifer Gale; Wen-Ning Zhao; Surya A. Reis; Douglas Barker; Erin Berry-Scott; Sung Won Kim; Elizabeth L. Clore; Jacob M. Hooker; Edward B. Holson; Stephen J. Haggarty; Tracey L. Petryshen

Psychiatric diseases, including schizophrenia, bipolar disorder and major depression, are projected to lead global disease burden within the next decade. Pharmacotherapy, the primary – albeit often ineffective – treatment method, has remained largely unchanged over the past 50 years, highlighting the need for novel target discovery and improved mechanism-based treatments. Here, we examined in wild type mice the impact of chronic, systemic treatment with Compound 60 (Cpd-60), a slow-binding, benzamide-based inhibitor of the class I histone deacetylase (HDAC) family members, HDAC1 and HDAC2, in mood-related behavioral assays responsive to clinically effective drugs. Cpd-60 treatment for one week was associated with attenuated locomotor activity following acute amphetamine challenge. Further, treated mice demonstrated decreased immobility in the forced swim test. These changes are consistent with established effects of clinical mood stabilizers and antidepressants, respectively. Whole-genome expression profiling of specific brain regions (prefrontal cortex, nucleus accumbens, hippocampus) from mice treated with Cpd-60 identified gene expression changes, including a small subset of transcripts that significantly overlapped those previously reported in lithium-treated mice. HDAC inhibition in brain was confirmed by increased histone acetylation both globally and, using chromatin immunoprecipitation, at the promoter regions of upregulated transcripts, a finding consistent with in vivo engagement of HDAC targets. In contrast, treatment with suberoylanilide hydroxamic acid (SAHA), a non-selective fast-binding, hydroxamic acid HDAC 1/2/3/6 inhibitor, was sufficient to increase histone acetylation in brain, but did not alter mood-related behaviors and had dissimilar transcriptional regulatory effects compared to Cpd-60. These results provide evidence that selective inhibition of HDAC1 and HDAC2 in brain may provide an epigenetic-based target for developing improved treatments for mood disorders and other brain disorders with altered chromatin-mediated neuroplasticity.


Nature Chemical Biology | 2011

A screen for regulators of survival of motor neuron protein levels

Nina R. Makhortova; Monica Hayhurst; Antonio Cerqueira; Amy D Sinor-Anderson; Wen-Ning Zhao; Patrick W. Heiser; Arvanites Arvanites; Lance S. Davidow; Zachary Waldon; Judith A. Steen; Kelvin Lam; Hien D. Ngo; Lee L. Rubin

The motor neuron disease spinal muscular atrophy (SMA) results from mutations that lead to low levels of the ubiquitously expressed protein survival of motor neuron (SMN). An ever-increasing collection of data suggests that therapeutics that elevate SMN may be effective in treating SMA. We executed an image-based screen of annotated chemical libraries and discovered several classes of compounds that were able to increase cellular SMN. Among the most important was the RTK-PI3K-AKT-GSK-3 signaling cascade. Chemical inhibitors of glycogen synthase kinase 3 (GSK-3) and short hairpin RNAs (shRNAs) directed against this target elevated SMN levels primarily by stabilizing the protein. It was particularly notable that GSK-3 chemical inhibitors were also effective in motor neurons, not only in elevating SMN levels, but also in blocking the death that was produced when SMN was acutely reduced by an SMN-specific shRNA. Thus, we have established a screen capable of detecting drug-like compounds that correct the main phenotypic change underlying SMA.


Proceedings of the National Academy of Sciences of the United States of America | 2017

Probing the lithium-response pathway in hiPSCs implicates the phosphoregulatory set-point for a cytoskeletal modulator in bipolar pathogenesis

Brian Tobe; Andrew M. Crain; Alicia M. Winquist; Barbara Calabrese; Hiroko Makihara; Wen-Ning Zhao; Jasmin Lalonde; Haruko Nakamura; Glenn T. Konopaske; Michelle M. Sidor; Cameron D. Pernia; Naoya Yamashita; Moyuka Wada; Yuuka Inoue; Fumio Nakamura; Steven D. Sheridan; Ryan W. Logan; Michael Brandel; Dongmei Wu; Joshua G. Hunsberger; Laurel Dorsett; Cordulla Duerr; Ranor C. B. Basa; Michael McCarthy; Namrata D. Udeshi; Philipp Mertins; Steven A. Carr; Guy A. Rouleau; Lina Mastrangelo; Jianxue Li

Significance One-third of bipolar disorder (BPD) patients are lithium-responsive (LiR) for unknown reasons. Were lithium’s target to be identified, then BPD’s pathogenesis might be unraveled. We identified and mapped the “lithium-response pathway,” which governs the phosphorylation of CRMP2, a cytoskeleton regulator, particularly for dendritic spines: hence, a neural network modulator. Although “toggling” between inactive (phosphorylated) and active (nonphosphorylated) CRMP2 is physiologic, the “set-point” in LiR BPD is abnormal. Lithium (and other pathway-modulators) normalize that set-point. Hence, BPD is a disorder not of a gene but of the posttranslational regulation of a developmentally critical molecule. Such knowledge should enable better mechanistically based treatments and bioassays. Instructively, lithium was our “molecular can-opener” for “prying” intracellularly to reveal otherwise inscrutable pathophysiology in this complex polygenic disorder. The molecular pathogenesis of bipolar disorder (BPD) is poorly understood. Using human-induced pluripotent stem cells (hiPSCs) to unravel such mechanisms in polygenic diseases is generally challenging. However, hiPSCs from BPD patients responsive to lithium offered unique opportunities to discern lithiums target and hence gain molecular insight into BPD. By profiling the proteomics of BDP–hiPSC-derived neurons, we found that lithium alters the phosphorylation state of collapsin response mediator protein-2 (CRMP2). Active nonphosphorylated CRMP2, which binds cytoskeleton, is present throughout the neuron; inactive phosphorylated CRMP2, which dissociates from cytoskeleton, exits dendritic spines. CRMP2 elimination yields aberrant dendritogenesis with diminished spine density and lost lithium responsiveness (LiR). The “set-point” for the ratio of pCRMP2:CRMP2 is elevated uniquely in hiPSC-derived neurons from LiR BPD patients, but not with other psychiatric (including lithium-nonresponsive BPD) and neurological disorders. Lithium (and other pathway modulators) lowers pCRMP2, increasing spine area and density. Human BPD brains show similarly elevated ratios and diminished spine densities; lithium therapy normalizes the ratios and spines. Consistent with such “spine-opathies,” human LiR BPD neurons with abnormal ratios evince abnormally steep slopes for calcium flux; lithium normalizes both. Behaviorally, transgenic mice that reproduce lithiums postulated site-of-action in dephosphorylating CRMP2 emulate LiR in BPD. These data suggest that the “lithium response pathway” in BPD governs CRMP2s phosphorylation, which regulates cytoskeletal organization, particularly in spines, modulating neural networks. Aberrations in the posttranslational regulation of this developmentally critical molecule may underlie LiR BPD pathogenesis. Instructively, examining the proteomic profile in hiPSCs of a functional agent—even one whose mechanism-of-action is unknown—might reveal otherwise inscrutable intracellular pathogenic pathways.


Journal of Biological Chemistry | 2015

Unbiased Cell-based Screening in a Neuronal Cell Model of Batten Disease Highlights an Interaction between Ca2+ Homeostasis, Autophagy, and CLN3 Protein Function

Uma Chandrachud; Mathew Walker; Alexandra M. Simas; Sasja Heetveld; Anton Petcherski; Madeleine Klein; Hyejin Oh; Pavlina Wolf; Wen-Ning Zhao; Stephanie Norton; Stephen J. Haggarty; Emyr Lloyd-Evans; Susan L. Cotman

Background: CLN3 protein function is still unknown, but its loss causes Batten disease. Results: Drug screening in a Batten disease model was developed to identify modifiers of altered cellular pathways. Conclusion: Alterations in Ca2+ handling are implicated in Batten disease, which may negatively influence the intracellular pathways regulated by Ca2+. Significance: A proof-of-concept is established for the application of drug screening to Batten disease research. Abnormal accumulation of undigested macromolecules, often disease-specific, is a major feature of lysosomal and neurodegenerative disease and is frequently attributed to defective autophagy. The mechanistic underpinnings of the autophagy defects are the subject of intense research, which is aided by genetic disease models. To gain an improved understanding of the pathways regulating defective autophagy specifically in juvenile neuronal ceroid lipofuscinosis (JNCL or Batten disease), a neurodegenerative disease of childhood, we developed and piloted a GFP-microtubule-associated protein 1 light chain 3 (GFP-LC3) screening assay to identify, in an unbiased fashion, genotype-sensitive small molecule autophagy modifiers, employing a JNCL neuronal cell model bearing the most common disease mutation in CLN3. Thapsigargin, a sarco/endoplasmic reticulum Ca2+-ATPase (SERCA) Ca2+ pump inhibitor, reproducibly displayed significantly more activity in the mouse JNCL cells, an effect that was also observed in human-induced pluripotent stem cell-derived JNCL neural progenitor cells. The mechanism of thapsigargin sensitivity was Ca2+-mediated, and autophagosome accumulation in JNCL cells could be reversed by Ca2+ chelation. Interrogation of intracellular Ca2+ handling highlighted alterations in endoplasmic reticulum, mitochondrial, and lysosomal Ca2+ pools and in store-operated Ca2+ uptake in JNCL cells. These results further support an important role for the CLN3 protein in intracellular Ca2+ handling and in autophagic pathway flux and establish a powerful new platform for therapeutic screening.


Bioorganic & Medicinal Chemistry | 2016

Kinetic and structural insights into the binding of histone deacetylase 1 and 2 (HDAC1, 2) inhibitors.

Florence F. Wagner; Michel Weiwer; Stefan Steinbacher; Adrian Schomburg; Peter Reinemer; Jennifer Gale; Arthur J. Campbell; Stewart L. Fisher; Wen-Ning Zhao; Surya A. Reis; Krista M. Hennig; Méryl Thomas; Peter Müller; Martin R. Jefson; Daniel M. Fass; Stephen J. Haggarty; Yan-Ling Zhang; Edward B. Holson

The structure-activity and structure-kinetic relationships of a series of novel and selective ortho-aminoanilide inhibitors of histone deacetylases (HDACs) 1 and 2 are described. Different kinetic and thermodynamic selectivity profiles were obtained by varying the moiety occupying an 11Å channel leading to the Zn(2+) catalytic pocket of HDACs 1 and 2, two paralogs with a high degree of structural similarity. The design of these novel inhibitors was informed by two ligand-bound crystal structures of truncated hHDAC2. BRD4884 and BRD7232 possess kinetic selectivity for HDAC1 versus HDAC2. We demonstrate that the binding kinetics of HDAC inhibitors can be tuned for individual isoforms in order to modulate target residence time while retaining functional activity and increased histone H4K12 and H3K9 acetylation in primary mouse neuronal cell culture assays. These chromatin modifiers, with tuned binding kinetic profiles, can be used to define the relation between target engagement requirements and the pharmacodynamic response of HDACs in different disease applications.


Bioorganic & Medicinal Chemistry Letters | 2016

Dissecting structure–activity-relationships of crebinostat: Brain penetrant HDAC inhibitors for neuroepigenetic regulation

Balaram Ghosh; Wen-Ning Zhao; Surya A. Reis; Debasis Patnaik; Daniel M. Fass; Li-Huei Tsai; Ralph Mazitschek; Stephen J. Haggarty

Targeting chromatin-mediated epigenetic regulation has emerged as a potential avenue for developing novel therapeutics for a wide range of central nervous system disorders, including cognitive disorders and depression. Histone deacetylase (HDAC) inhibitors have been pursued as cognitive enhancers that impact the regulation of gene expression and other mechanisms integral to neuroplasticity. Through systematic modification of the structure of crebinostat, a previously discovered cognitive enhancer that affects genes critical to memory and enhances synaptogenesis, combined with biochemical and neuronal cell-based screening, we identified a novel hydroxamate-based HDAC inhibitor, here named neurinostat, with increased potency compared to crebinostat in inducing neuronal histone acetylation. In addition, neurinostat was found to have a pharmacokinetic profile in mouse brain modestly improved over that of crebinostat. This discovery of neurinostat and demonstration of its effects on neuronal HDACs adds to the available pharmacological toolkit for dissecting the molecular and cellular mechanisms of neuroepigenetic regulation in health and disease.


Molecular Neuropsychiatry | 2017

WNT/β-Catenin Pathway and Epigenetic Mechanisms Regulate the Pitt-Hopkins Syndrome and Schizophrenia Risk Gene TCF4

Krista M. Hennig; Daniel M. Fass; Wen-Ning Zhao; Steven D. Sheridan; Ting Fu; Serkan Erdin; Alexei Stortchevoi; Diane Lucente; Jannine D. Cody; David Sweetser; James F. Gusella; Michael E. Talkowski; Stephen J. Haggarty

Genetic variation within the transcription factor TCF4 locus can cause the intellectual disability and developmental disorder Pitt-Hopkins syndrome (PTHS), whereas single-nucleotide polymorphisms within noncoding regions are associated with schizophrenia. These genetic findings position TCF4 as a link between transcription and cognition; however, the neurobiology of TCF4 remains poorly understood. Here, we quantitated multiple distinct TCF4 transcript levels in human induced pluripotent stem cell-derived neural progenitors and differentiated neurons, and PTHS patient fibroblasts. We identify two classes of pharmacological treatments that regulate TCF4 expression: WNT pathway activation and inhibition of class I histone deacetylases. In PTHS fibroblasts, both of these perturbations upregulate a subset of TCF4 transcripts. Finally, using chromatin immunoprecipitation sequencing in conjunction with genome-wide transcriptome analysis, we identified TCF4 target genes that may mediate the effect of TCF4 loss on neuroplasticity. Our studies identify new pharmacological assays, tools, and targets for the development of therapeutics for cognitive disorders.


PLOS ONE | 2018

Traditional and systems biology based drug discovery for the rare tumor syndrome neurofibromatosis type 2

Robert J. Allaway; Steve Angus; Roberta L. Beauchamp; Jaishri O. Blakeley; Marga Bott; Sarah S. Burns; Annemarie Carlstedt; Long-Sheng Chang; Xin Chen; D. Wade Clapp; Patrick DeSouza; Serkan Erdin; Cristina Fernandez-Valle; Justin Guinney; James F. Gusella; Stephen J. Haggarty; Gary L. Johnson; Salvatore La Rosa; Helen Morrison; Alejandra M. Petrilli; Scott R. Plotkin; Abhishek Pratap; Vijaya Ramesh; Noah Sciaky; Anat Stemmer-Rachamimov; Tim J. Stuhlmiller; Michael E. Talkowski; D. Bradley Welling; Charles W. Yates; Jon S. Zawistowski

Neurofibromatosis 2 (NF2) is a rare tumor suppressor syndrome that manifests with multiple schwannomas and meningiomas. There are no effective drug therapies for these benign tumors and conventional therapies have limited efficacy. Various model systems have been created and several drug targets have been implicated in NF2-driven tumorigenesis based on known effects of the absence of merlin, the product of the NF2 gene. We tested priority compounds based on known biology with traditional dose-concentration studies in meningioma and schwann cell systems. Concurrently, we studied functional kinome and gene expression in these cells pre- and post-treatment to determine merlin deficient molecular phenotypes. Cell viability results showed that three agents (GSK2126458, Panobinostat, CUDC-907) had the greatest activity across schwannoma and meningioma cell systems, but merlin status did not significantly influence response. In vivo, drug effect was tumor specific with meningioma, but not schwannoma, showing response to GSK2126458 and Panobinostat. In culture, changes in both the transcriptome and kinome in response to treatment clustered predominantly based on tumor type. However, there were differences in both gene expression and functional kinome at baseline between meningioma and schwannoma cell systems that may form the basis for future selective therapies. This work has created an openly accessible resource (www.synapse.org/SynodosNF2) of fully characterized isogenic schwannoma and meningioma cell systems as well as a rich data source of kinome and transcriptome data from these assay systems before and after treatment that enables single and combination drug discovery based on molecular phenotype.


ACS Chemical Neuroscience | 2018

Class I Histone Deacetylase Inhibition by Tianeptinaline Modulates Neuroplasticity and Enhances Memory

Wen-Ning Zhao; Balaram Ghosh; Marshall W. Tyler; Jasmin Lalonde; Nadine F. Joseph; Nina Kosaric; Daniel M. Fass; Li-Huei Tsai; Ralph Mazitschek; Stephen J. Haggarty

Through epigenetic and other regulatory functions, the histone deacetylase (HDAC) family of enzymes has emerged as a promising therapeutic target for central nervous system and other disorders. Here we report on the synthesis and functional characterization of new HDAC inhibitors based structurally on tianeptine, a drug used primarily to treat major depressive disorder (MDD) that has a poorly understood mechanism of action. Since the chemical structure of tianeptine resembles certain HDAC inhibitors, we profiled the in vitro HDAC inhibitory activity of tianeptine and demonstrated its ability to inhibit the lysine deacetylase activity of a subset of class I HDACs. Consistent with a model of active site Zn2+ chelation by the carboxylic acid present in tianeptine, newly synthesized analogues containing either a hydroxamic acid or ortho-aminoanilide exhibited increased potency and selectivity among the HDAC family. This in vitro potency translated to improved efficacy in a panel of high-content imaging assays designed to assess HDAC target engagement and functional effects on critical pathways involved in neuroplasticity in both primary mouse neurons and, for the first time, human neurons differentiated from pluripotent stem cells. Most notably, tianeptinaline, a class I HDAC-selective analogue of tianeptine, but not tianeptine itself, increased histone acetylation, and enhanced CREB-mediated transcription and the expression of Arc (activity-regulated cytoskeleton-associated protein). Systemic in vivo administration of tianeptinaline to mice confirmed its brain penetration and was found to enhance contextual fear conditioning, a behavioral test of hippocampal-dependent memory. Tianeptinaline and its derivatives provide new pharmacological tools to dissect chromatin-mediated neuroplasticity underlying memory and other epigenetically related processes implicated in health and disease.

Collaboration


Dive into the Wen-Ning Zhao'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

Balaram Ghosh

Birla Institute of Technology and Science

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge