1
|
Li JZ, Ramalingam N, Li S. Targeting epigenetic mechanisms in amyloid-β-mediated Alzheimer's pathophysiology: unveiling therapeutic potential. Neural Regen Res 2025; 20:54-66. [PMID: 38767476 PMCID: PMC11246147 DOI: 10.4103/nrr.nrr-d-23-01827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 01/03/2024] [Accepted: 02/07/2024] [Indexed: 05/22/2024] Open
Abstract
Alzheimer's disease is a prominent chronic neurodegenerative condition characterized by a gradual decline in memory leading to dementia. Growing evidence suggests that Alzheimer's disease is associated with accumulating various amyloid-β oligomers in the brain, influenced by complex genetic and environmental factors. The memory and cognitive deficits observed during the prodromal and mild cognitive impairment phases of Alzheimer's disease are believed to primarily result from synaptic dysfunction. Throughout life, environmental factors can lead to enduring changes in gene expression and the emergence of brain disorders. These changes, known as epigenetic modifications, also play a crucial role in regulating the formation of synapses and their adaptability in response to neuronal activity. In this context, we highlight recent advances in understanding the roles played by key components of the epigenetic machinery, specifically DNA methylation, histone modification, and microRNAs, in the development of Alzheimer's disease, synaptic function, and activity-dependent synaptic plasticity. Moreover, we explore various strategies, including enriched environments, exposure to non-invasive brain stimulation, and the use of pharmacological agents, aimed at improving synaptic function and enhancing long-term potentiation, a process integral to epigenetic mechanisms. Lastly, we deliberate on the development of effective epigenetic agents and safe therapeutic approaches for managing Alzheimer's disease. We suggest that addressing Alzheimer's disease may require distinct tailored epigenetic drugs targeting different disease stages or pathways rather than relying on a single drug.
Collapse
Affiliation(s)
- Jennie Z. Li
- Ann Romney Center for Neurologic Diseases, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Nagendran Ramalingam
- Ann Romney Center for Neurologic Diseases, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Shaomin Li
- Ann Romney Center for Neurologic Diseases, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
2
|
Osaki T, Delepine C, Osako Y, Kranz D, Levin A, Nelson C, Fagiolini M, Sur M. Early differential impact of MeCP2 mutations on functional networks in Rett syndrome patient-derived human cerebral organoids. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.10.607464. [PMID: 39149328 PMCID: PMC11326256 DOI: 10.1101/2024.08.10.607464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
Human cerebral organoids derived from induced pluripotent stem cells can recapture early developmental processes and reveal changes involving neurodevelopmental disorders. Mutations in the X-linked methyl-CpG binding protein 2 (MECP2) gene are associated with Rett syndrome, and disease severity varies depending on the location and type of mutation. Here, we focused on neuronal activity in Rett syndrome patient-derived organoids, analyzing two types of MeCP2 mutations - a missense mutation (R306C) and a truncating mutation (V247X) - using calcium imaging with three-photon microscopy. Compared to isogenic controls, we found abnormal neuronal activity in Rett organoids and altered network function based on graph theoretic analyses, with V247X mutations impacting functional responses and connectivity more severely than R306C mutations. These changes paralleled EEG data obtained from patients with comparable mutations. Labeling DLX promoter-driven inhibitory neurons demonstrated differences in activity and functional connectivity of inhibitory and excitatory neurons in the two types of mutation. Transcriptomic analyses revealed HDAC2-associated impairment in R306C organoids and decreased GABAA receptor expression in excitatory neurons in V247X organoids. These findings demonstrate mutation-specific mechanisms of vulnerability in Rett syndrome and suggest targeted strategies for their treatment.
Collapse
Affiliation(s)
- Tatsuya Osaki
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Whitehead Institute for Biomedical Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Chloe Delepine
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Yuma Osako
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Devorah Kranz
- Boston Children’s Hospital, Brookline, MA 02445, USA
- Harvard University, Cambridge, MA 02139, USA
| | - April Levin
- Boston Children’s Hospital, Brookline, MA 02445, USA
| | - Charles Nelson
- Boston Children’s Hospital, Brookline, MA 02445, USA
- Harvard University, Cambridge, MA 02139, USA
| | - Michela Fagiolini
- Boston Children’s Hospital, Brookline, MA 02445, USA
- Harvard University, Cambridge, MA 02139, USA
| | - Mriganka Sur
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| |
Collapse
|
3
|
Ciceri G, Studer L. Epigenetic control and manipulation of neuronal maturation timing. Curr Opin Genet Dev 2024; 85:102164. [PMID: 38412562 PMCID: PMC11175593 DOI: 10.1016/j.gde.2024.102164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 01/29/2024] [Accepted: 01/30/2024] [Indexed: 02/29/2024]
Abstract
During brain development, the sequence of developmental steps and the underlying transcriptional regulatory logic are largely conserved across species. However, the temporal unfolding of developmental programs varies dramatically across species and within a given species varies across brain regions and cell identities. The maturation of neurons in the human cerebral cortex is particularly slow and lasts for many years compared with only a few weeks for the corresponding mouse neurons. The mechanisms setting the 'schedule' of neuronal maturation remain unclear but appear to be linked to a cell-intrinsic 'clock'. Here, we discuss recent findings that highlight a role for epigenetic factors in the timing of neuronal maturation. Manipulations of those factors in stem cell-based models can override the intrinsic pace of neuronal maturation, including its protracted nature in human cortical neurons. We then contextualize the epigenetic regulation of maturation programs with findings from other model systems and propose potential interactions between epigenetic pathways and other drivers of developmental rates.
Collapse
Affiliation(s)
- Gabriele Ciceri
- The Center for Stem Cell Biology and Developmental Biology program, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| | - Lorenz Studer
- The Center for Stem Cell Biology and Developmental Biology program, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
4
|
Fan W, Zhou J. Icariside II protects dopaminergic neurons from 1‑methyl‑4‑phenylpyridinium‑induced neurotoxicity by downregulating HDAC2 to restore mitochondrial function. Exp Ther Med 2024; 27:40. [PMID: 38125349 PMCID: PMC10731403 DOI: 10.3892/etm.2023.12328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 08/18/2023] [Indexed: 12/23/2023] Open
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disease after Alzheimer's disease (AD). Icariside II (ICS II) is known to confer notable therapeutic effects against a variety of neurodegenerative diseases, such as AD. Therefore, the present study aimed to evaluate the possible effects of ICS II on 1-methyl-4-phenylpyridinium (MPP+)-induced SK-N-SH cell injury, in addition to understanding the underlying mechanism of action. The MPP+-induced SK-N-SH cell model was used to simulate PD in vitro. The viability and mitochondrial membrane potential of SK-N-SH cells were detected by MTT assay and JC-1 staining, respectively. Lactate dehydrogenase (LDH) release, ATP levels and complex I activity in treated SK-N-SH cells were measured using LDH activity, ATP and Complex I assay kits, respectively. The protein expression levels of histone deacetylase 2 (HDAC2) and γ-H2A histone family member X and the copy number of mitochondrial DNA were measured by western blotting or reverse transcription-quantitative PCR, respectively. Autodock 4.2 was used to predict the molecular docking site of ICS II on HDAC2. The results of the present study demonstrated that ICS II mitigated SK-N-SH cytotoxicity induced by MPP+. Specifically, ICS II alleviated DNA damage and restored mitochondrial function in SK-N-SH cells treated with MPP+. In addition, ICS II reduced the HDAC2 protein expression levels in MPP+-induced SK-N-SH cells. However, overexpression of HDAC2 reversed the protective effects of ICS II on DNA damage and mitochondrial dysfunction in MPP+-induced SK-N-SH cells. In conclusion, the results of the present study suggest that ICS II can protect dopaminergic neurons from MPP+-induced neurotoxicity by downregulating HDAC2 expression to restore mitochondrial function.
Collapse
Affiliation(s)
- Wenbo Fan
- Pharmaceutical Technology Department, Chemical Engineering School, Jiuquan Vocational Technical College, Jiuquan, Gansu 735000, P.R. China
| | - Jianwu Zhou
- Medical Laboratory of Qinghai Provincial People's Hospital, Xining, Qinghai 810000, P.R. China
| |
Collapse
|
5
|
Marinho D, Ferreira IL, Lorenzoni R, Cardoso SM, Santana I, Rego AC. Reduction of class I histone deacetylases ameliorates ER-mitochondria cross-talk in Alzheimer's disease. Aging Cell 2023; 22:e13895. [PMID: 37358017 PMCID: PMC10410063 DOI: 10.1111/acel.13895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 04/30/2023] [Accepted: 05/12/2023] [Indexed: 06/27/2023] Open
Abstract
Several molecular mechanisms have been described in Alzheimer's disease (AD), including repressed gene transcription and mitochondrial and endoplasmic reticulum (ER) dysfunction. In this study, we evaluate the potential efficacy of transcriptional modifications exerted by inhibition or knockdown of class I histone deacetylases (HDACs) in ameliorating ER-mitochondria cross-talk in AD models. Data show increased HDAC3 protein levels and decreased acetyl-H3 in AD human cortex, and increased HDAC2-3 in MCI peripheral human cells, HT22 mouse hippocampal cells exposed to Aβ1-42 oligomers (AβO) and APP/PS1 mouse hippocampus. Tacedinaline (Tac, a selective class I HDAC inhibitor) counteracted the increase in ER-Ca2+ retention and mitochondrial Ca2+ accumulation, mitochondrial depolarization and impaired ER-mitochondria cross-talk, as observed in 3xTg-AD mouse hippocampal neurons and AβO-exposed HT22 cells. We further demonstrated diminished mRNA levels of proteins involved in mitochondrial-associated ER membranes (MAM) in cells exposed to AβO upon Tac treatment, along with reduction in ER-mitochondria contacts (MERCS) length. HDAC2 silencing reduced ER-mitochondria Ca2+ transfer and mitochondrial Ca2+ retention, while knockdown of HDAC3 decreased ER-Ca2+ accumulation in AβO-treated cells. APP/PS1 mice treated with Tac (30 mg/kg/day) also showed regulation of mRNA levels of MAM-related proteins, and reduced Aβ levels. These data demonstrate that Tac normalizes Ca2+ signaling between mitochondria and ER, involving the tethering between the two organelles in AD hippocampal neural cells. Tac-mediated AD amelioration occurs through the regulation of protein expression at MAM, as observed in AD cells and animal models. Data support transcriptional regulation of ER-mitochondria communication as a promising target for innovative therapeutics in AD.
Collapse
Affiliation(s)
- Daniela Marinho
- CNC‐Center for Neuroscience and Cell BiologyUniversity of CoimbraCoimbraPortugal
- IIIUC‐Institute for Interdisciplinary ResearchUniversity of CoimbraCoimbraPortugal
- CIBB‐Center for Innovative Biomedicine and BiotechnologyUniversity of CoimbraCoimbraPortugal
| | - Ildete Luísa Ferreira
- CNC‐Center for Neuroscience and Cell BiologyUniversity of CoimbraCoimbraPortugal
- IIIUC‐Institute for Interdisciplinary ResearchUniversity of CoimbraCoimbraPortugal
- CIBB‐Center for Innovative Biomedicine and BiotechnologyUniversity of CoimbraCoimbraPortugal
| | - Ricardo Lorenzoni
- CNC‐Center for Neuroscience and Cell BiologyUniversity of CoimbraCoimbraPortugal
- IIIUC‐Institute for Interdisciplinary ResearchUniversity of CoimbraCoimbraPortugal
- CIBB‐Center for Innovative Biomedicine and BiotechnologyUniversity of CoimbraCoimbraPortugal
| | - Sandra M. Cardoso
- CNC‐Center for Neuroscience and Cell BiologyUniversity of CoimbraCoimbraPortugal
- CIBB‐Center for Innovative Biomedicine and BiotechnologyUniversity of CoimbraCoimbraPortugal
- FMUC‐Faculty of MedicineUniversity of CoimbraCoimbraPortugal
| | - Isabel Santana
- CNC‐Center for Neuroscience and Cell BiologyUniversity of CoimbraCoimbraPortugal
- CIBB‐Center for Innovative Biomedicine and BiotechnologyUniversity of CoimbraCoimbraPortugal
- FMUC‐Faculty of MedicineUniversity of CoimbraCoimbraPortugal
- Neurology DepartmentCHUC‐Centro Hospitalar e Universitário de CoimbraCoimbraPortugal
| | - A. Cristina Rego
- CNC‐Center for Neuroscience and Cell BiologyUniversity of CoimbraCoimbraPortugal
- CIBB‐Center for Innovative Biomedicine and BiotechnologyUniversity of CoimbraCoimbraPortugal
- FMUC‐Faculty of MedicineUniversity of CoimbraCoimbraPortugal
| |
Collapse
|
6
|
Single and repeated bisphenol A treatment induces ROS, Aβ and hyperphosphorylated-tau accumulation, and insulin pathways disruption, through HDAC2 and PTP1B overexpression, leading to SN56 cholinergic apoptotic cell death. Food Chem Toxicol 2022; 170:113500. [DOI: 10.1016/j.fct.2022.113500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 10/11/2022] [Accepted: 10/24/2022] [Indexed: 11/11/2022]
|
7
|
Epigenetic Regulation in Neurodegeneration Disease. Int J Mol Sci 2022; 23:ijms23116185. [PMID: 35682863 PMCID: PMC9181079 DOI: 10.3390/ijms23116185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 05/31/2022] [Indexed: 01/27/2023] Open
|
8
|
Cho C, Zeigler M, Mizuno S, Morrison RS, Totah RA, Barker-Haliski M. Reductions in Hydrogen Sulfide and Changes in Mitochondrial Quality Control Proteins Are Evident in the Early Phases of the Corneally Kindled Mouse Model of Epilepsy. Int J Mol Sci 2022; 23:ijms23031434. [PMID: 35163358 PMCID: PMC8835945 DOI: 10.3390/ijms23031434] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 01/21/2022] [Accepted: 01/24/2022] [Indexed: 12/14/2022] Open
Abstract
Epilepsy is a heterogenous neurological disorder characterized by recurrent unprovoked seizures, mitochondrial stress, and neurodegeneration. Hydrogen sulfide (H2S) is a gasotransmitter that promotes mitochondrial function and biogenesis, elicits neuromodulation and neuroprotection, and may acutely suppress seizures. A major gap in knowledge remains in understanding the role of mitochondrial dysfunction and progressive changes in H2S levels following acute seizures or during epileptogenesis. We thus sought to quantify changes in H2S and its methylated metabolite (MeSH) via LC-MS/MS following acute maximal electroshock and 6 Hz 44 mA seizures in mice, as well as in the early phases of the corneally kindled mouse model of chronic seizures. Plasma H2S was acutely reduced after a maximal electroshock seizure. H2S or MeSH levels and expressions of related genes in whole brain homogenates from corneally kindled mice were not altered. However, plasma H2S levels were significantly lower during kindling, but not after established kindling. Moreover, we demonstrated a time-dependent increase in expression of mitochondrial membrane integrity-related proteins, OPA1, MFN2, Drp1, and Mff during kindling, which did not correlate with changes in gene expression. Taken together, short-term reductions in plasma H2S could be a novel biomarker for seizures. Future studies should further define the role of H2S and mitochondrial stress in epilepsy.
Collapse
Affiliation(s)
- Christi Cho
- Department of Medicinal Chemistry, University of Washington, Seattle, WA 98195, USA; (C.C.); (M.Z.); (R.A.T.)
| | - Maxwell Zeigler
- Department of Medicinal Chemistry, University of Washington, Seattle, WA 98195, USA; (C.C.); (M.Z.); (R.A.T.)
| | - Stephanie Mizuno
- Department of Pharmacy, University of Washington, Seattle, WA 98195, USA;
| | | | - Rheem A. Totah
- Department of Medicinal Chemistry, University of Washington, Seattle, WA 98195, USA; (C.C.); (M.Z.); (R.A.T.)
| | - Melissa Barker-Haliski
- Department of Pharmacy, University of Washington, Seattle, WA 98195, USA;
- Correspondence: ; Tel.: +1-206-685-1783
| |
Collapse
|
9
|
Potjewyd FM, Annor‐Gyamfi JK, Aubé J, Chu S, Conlon IL, Frankowski KJ, Guduru SKR, Hardy BP, Hopkins MD, Kinoshita C, Kireev DB, Mason ER, Moerk CT, Nwogbo F, Pearce KH, Richardson TI, Rogers DA, Soni DM, Stashko M, Wang X, Wells C, Willson TM, Frye SV, Young JE, Axtman AD. Use of AD Informer Set compounds to explore validity of novel targets in Alzheimer's disease pathology. ALZHEIMER'S & DEMENTIA: TRANSLATIONAL RESEARCH & CLINICAL INTERVENTIONS 2022; 8:e12253. [PMID: 35434254 PMCID: PMC9005681 DOI: 10.1002/trc2.12253] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 11/29/2021] [Accepted: 12/15/2021] [Indexed: 12/03/2022]
Abstract
Introduction A chemogenomic set of small molecules with annotated activities and implicated roles in Alzheimer's disease (AD) called the AD Informer Set was recently developed and made available to the AD research community: https://treatad.org/data‐tools/ad‐informer‐set/. Methods Small subsets of AD Informer Set compounds were selected for AD‐relevant profiling. Nine compounds targeting proteins expressed by six AD‐implicated genes prioritized for study by Target Enablement to Accelerate Therapy Development for Alzheimer's Disease (TREAT‐AD) teams were selected for G‐protein coupled receptor (GPCR), amyloid beta (Aβ) and tau, and pharmacokinetic (PK) studies. Four non‐overlapping compounds were analyzed in microglial cytotoxicity and phagocytosis assays. Results The nine compounds targeting CAPN2, EPHX2, MDK, MerTK/FLT3, or SYK proteins were profiled in 46 to 47 primary GPCR binding assays. Human induced pluripotent stem cell (iPSC)‐derived neurons were treated with the same nine compounds and secretion of Aβ peptides (Aβ40 and Aβ42) as well as levels of phosphophorylated tau (p‐tau, Thr231) and total tau (t‐tau) peptides measured at two concentrations and two timepoints. Finally, CD1 mice were dosed intravenously to determine preliminary PK and/or brain‐specific penetrance values for these compounds. As a final cell‐based study, a non‐overlapping subset of four compounds was selected based on single‐concentration screening for analysis of both cytotoxicity and phagocytosis in murine and human microglia cells. Discussion We have demonstrated the utility of the AD Informer Set in the validation of novel AD hypotheses using biochemical, cellular (primary and immortalized), and in vivo studies. The selectivity for their primary targets versus essential GPCRs in the brain was established for our compounds. Statistical changes in tau, p‐tau, Aβ40, and/or Aβ42 and blood–brain barrier penetrance were observed, solidifying the utility of specific compounds for AD. Single‐concentration phagocytosis results were validated as predictive of dose–response findings. These studies established workflows, validated assays, and illuminated next steps for protein targets and compounds.
Collapse
Affiliation(s)
- Frances M. Potjewyd
- UNC Eshelman School of Pharmacy Division of Chemical Biology and Medicinal Chemistry Structural Genomics Consortium Chapel Hill North Carolina USA
| | - Joel K. Annor‐Gyamfi
- UNC Eshelman School of Pharmacy Division of Chemical Biology and Medicinal Chemistry Structural Genomics Consortium Chapel Hill North Carolina USA
| | - Jeffrey Aubé
- UNC Eshelman School of Pharmacy Division of Chemical Biology and Medicinal Chemistry Center for Integrative Chemical Biology and Drug Discovery Chapel Hill North Carolina USA
| | - Shaoyou Chu
- Department of Laboratory Medicine and Pathology University of Washington Seattle Washington USA
| | - Ivie L. Conlon
- UNC Eshelman School of Pharmacy Division of Chemical Biology and Medicinal Chemistry Center for Integrative Chemical Biology and Drug Discovery Chapel Hill North Carolina USA
| | - Kevin J. Frankowski
- UNC Eshelman School of Pharmacy Division of Chemical Biology and Medicinal Chemistry Center for Integrative Chemical Biology and Drug Discovery Chapel Hill North Carolina USA
| | - Shiva K. R. Guduru
- UNC Eshelman School of Pharmacy Division of Chemical Biology and Medicinal Chemistry Center for Integrative Chemical Biology and Drug Discovery Chapel Hill North Carolina USA
| | - Brian P. Hardy
- UNC Eshelman School of Pharmacy Division of Chemical Biology and Medicinal Chemistry Center for Integrative Chemical Biology and Drug Discovery Chapel Hill North Carolina USA
| | - Megan D. Hopkins
- UNC Eshelman School of Pharmacy Division of Chemical Biology and Medicinal Chemistry Center for Integrative Chemical Biology and Drug Discovery Chapel Hill North Carolina USA
| | - Chizuru Kinoshita
- Department of Laboratory Medicine and Pathology University of Washington Seattle Washington USA
- Institute for Stem Cell and Regenerative Medicine University of Washington Seattle Washington USA
| | - Dmitri B. Kireev
- UNC Eshelman School of Pharmacy Division of Chemical Biology and Medicinal Chemistry Center for Integrative Chemical Biology and Drug Discovery Chapel Hill North Carolina USA
| | - Emily R. Mason
- Department of Medicine Division of Clinical Pharmacology Indiana University School of Medicine Indianapolis Indiana USA
| | - Charles T. Moerk
- Department of Laboratory Medicine and Pathology University of Washington Seattle Washington USA
- Institute for Stem Cell and Regenerative Medicine University of Washington Seattle Washington USA
| | - Felix Nwogbo
- UNC Eshelman School of Pharmacy Division of Chemical Biology and Medicinal Chemistry Center for Integrative Chemical Biology and Drug Discovery Chapel Hill North Carolina USA
| | - Kenneth H. Pearce
- UNC Eshelman School of Pharmacy Division of Chemical Biology and Medicinal Chemistry Center for Integrative Chemical Biology and Drug Discovery Chapel Hill North Carolina USA
| | - Timothy I. Richardson
- Department of Medicine Division of Clinical Pharmacology Indiana University School of Medicine Indianapolis Indiana USA
| | - David A. Rogers
- UNC Eshelman School of Pharmacy Division of Chemical Biology and Medicinal Chemistry Center for Integrative Chemical Biology and Drug Discovery Chapel Hill North Carolina USA
| | - Disha M. Soni
- Department of Medicine Division of Clinical Pharmacology Indiana University School of Medicine Indianapolis Indiana USA
| | - Michael Stashko
- UNC Eshelman School of Pharmacy Division of Chemical Biology and Medicinal Chemistry Center for Integrative Chemical Biology and Drug Discovery Chapel Hill North Carolina USA
| | - Xiaodong Wang
- UNC Eshelman School of Pharmacy Division of Chemical Biology and Medicinal Chemistry Center for Integrative Chemical Biology and Drug Discovery Chapel Hill North Carolina USA
| | - Carrow Wells
- UNC Eshelman School of Pharmacy Division of Chemical Biology and Medicinal Chemistry Structural Genomics Consortium Chapel Hill North Carolina USA
| | - Timothy M. Willson
- UNC Eshelman School of Pharmacy Division of Chemical Biology and Medicinal Chemistry Structural Genomics Consortium Chapel Hill North Carolina USA
| | - Stephen V. Frye
- UNC Eshelman School of Pharmacy Division of Chemical Biology and Medicinal Chemistry Center for Integrative Chemical Biology and Drug Discovery Chapel Hill North Carolina USA
| | - Jessica E. Young
- Department of Laboratory Medicine and Pathology University of Washington Seattle Washington USA
- Institute for Stem Cell and Regenerative Medicine University of Washington Seattle Washington USA
| | - Alison D. Axtman
- UNC Eshelman School of Pharmacy Division of Chemical Biology and Medicinal Chemistry Structural Genomics Consortium Chapel Hill North Carolina USA
| |
Collapse
|