1
|
Wu Q, Morrow EM, Gamsiz Uzun ED. A deep learning model for prediction of autism status using whole-exome sequencing data. PLoS Comput Biol 2024; 20:e1012468. [PMID: 39514604 DOI: 10.1371/journal.pcbi.1012468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 11/20/2024] [Accepted: 09/06/2024] [Indexed: 11/16/2024] Open
Abstract
Autism is a developmental disability. Research demonstrated that children with autism benefit from early diagnosis and early intervention. Genetic factors are considered major contributors to the development of autism. Machine learning (ML), including deep learning (DL), has been evaluated in phenotype prediction, but this method has been limited in its application to autism. We developed a DL model, the Separate Translated Autism Research Neural Network (STAR-NN) model to predict autism status. The model was trained and tested using whole exome sequencing data from 43,203 individuals (16,809 individuals with autism and 26,394 non-autistic controls). Polygenic scores from common variants and the aggregated count of rare variants on genes were used as input. In STAR-NN, protein truncating variants, possibly damaging missense variants and mild effect missense variants on the same gene were separated at the input level and merged to one gene node. In this way, rare variants with different level of pathogenic effects were treated separately. We further validated the performance of STAR-NN using an independent dataset, including 13,827 individuals with autism and 14,052 non-autistic controls. STAR-NN achieved a modest ROC-AUC of 0.7319 on the testing dataset and 0.7302 on the independent dataset. STAR-NN outperformed other traditional ML models. Gene Ontology analysis on the selected gene features showed an enrichment for potentially informative pathways including calcium ion transport.
Collapse
Affiliation(s)
- Qing Wu
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, Rhode Island, United States of America
- Center for Translational Neuroscience, Robert J. and Nancy D. Carney Institute for Brain Science and Brown Institute for Translational Science, Brown University, Providence, Rhode Island, United States of America
- Center for Computational Molecular Biology, Brown University, Providence, Rhode Island, United States of America
| | - Eric M Morrow
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, Rhode Island, United States of America
- Center for Translational Neuroscience, Robert J. and Nancy D. Carney Institute for Brain Science and Brown Institute for Translational Science, Brown University, Providence, Rhode Island, United States of America
- Center for Computational Molecular Biology, Brown University, Providence, Rhode Island, United States of America
- Developmental Disorders Genetics Research Program, Department of Psychiatry and Human Behavior, Emma Pendleton Bradley Hospital, East Providence, Rhode Island, United States of America
| | - Ece D Gamsiz Uzun
- Center for Translational Neuroscience, Robert J. and Nancy D. Carney Institute for Brain Science and Brown Institute for Translational Science, Brown University, Providence, Rhode Island, United States of America
- Center for Computational Molecular Biology, Brown University, Providence, Rhode Island, United States of America
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School of Brown University, Providence, Rhode Island, United States of America
- Department of Pathology and Laboratory Medicine, Rhode Island Hospital, Providence, Rhode Island, United States of America
| |
Collapse
|
2
|
Tian L, He M, Fan H, Zhang H, Dong X, Qiao M, Tang C, Yu Y, Chen T, Zhou N. COVID-19 of differing severity: from bulk to single-cell expression data analysis. Cell Cycle 2023; 22:1777-1797. [PMID: 37486005 PMCID: PMC10446813 DOI: 10.1080/15384101.2023.2239620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 04/03/2023] [Accepted: 06/24/2023] [Indexed: 07/25/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19) is raging worldwide and causes an immense disease burden. Despite this, the biomarkers and targeting drugs of COVID-19 of differing severity remain largely unknown. Based on the GSE164805 dataset, we identified modules most critical for mild COVID-19 (mCOVID-19) and severe COVID-19 (sCOVID-19) through WGCNA, respectively. We subsequently constructed a protein-protein interaction network, and detected 16 hub genes for mCOVID-19 and 10 hub genes for sCOVID-19, followed by the prediction of upstream transcription factors (TFs) and kinases. The enrichment analysis then showed downregulation of TNFA signaling via NFKB for mCOVID-19, as well as downregulation of MYC targets V1 for sCOVID-19. Infiltration degrees of many immune cells, such as macrophages, were also sharply different between mCOVID-19 and sCOVID-19 samples. Predicted protein targeting drugs with the highest scores nearly all belong to naturally derived or synthetic glucocorticoids. For the two single-cell RNA-seq datasets, we explored the expression distribution of hub genes for mCOVID-19/sCOVID-19 in each cell type. The expression levels of PRKCA, MCM5, TYMS, RBBP4, BCL6, FLOT1, KDM6B, and TLR2 were found to be cell-type-specific. Furthermore, the expression levels of 10 hub genes for mCOVID-19 were significantly upregulated in PBMCs between eight healthy controls and eight mCOVID-19 patients at our institution. Collectively, we detected critical modules, pathways, TFs, kinases, immune cells, targeting drugs, hub genes, and their expression distributions in different cell types that may involve the pathogenesis of COVID-19 of differing severity, which may propel earlier diagnosis and more effective treatment of this intractable disease in the future.
Collapse
Affiliation(s)
- Linlin Tian
- Nanjing Municipal Center for Disease Control and Prevention, Nanjing, Jiangsu, P.R. China
| | - Min He
- Nanjing Municipal Center for Disease Control and Prevention, Nanjing, Jiangsu, P.R. China
| | - Huafeng Fan
- Nanjing Municipal Center for Disease Control and Prevention, Nanjing, Jiangsu, P.R. China
| | - Hongying Zhang
- Nanjing Municipal Center for Disease Control and Prevention, Nanjing, Jiangsu, P.R. China
| | - Xiaoxiao Dong
- Nanjing Municipal Center for Disease Control and Prevention, Nanjing, Jiangsu, P.R. China
| | - Mengkai Qiao
- Nanjing Municipal Center for Disease Control and Prevention, Nanjing, Jiangsu, P.R. China
| | - Chenyu Tang
- Nanjing Municipal Center for Disease Control and Prevention, Nanjing, Jiangsu, P.R. China
| | - Yan Yu
- Nanjing Municipal Center for Disease Control and Prevention, Nanjing, Jiangsu, P.R. China
| | - Tong Chen
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, P.R. China
| | - Nan Zhou
- Nanjing Municipal Center for Disease Control and Prevention, Nanjing, Jiangsu, P.R. China
| |
Collapse
|
3
|
Yang J, Luly KM, Green JJ. Nonviral nanoparticle gene delivery into the CNS for neurological disorders and brain cancer applications. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2023; 15:e1853. [PMID: 36193561 PMCID: PMC10023321 DOI: 10.1002/wnan.1853] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 05/24/2022] [Accepted: 08/11/2022] [Indexed: 03/15/2023]
Abstract
Nonviral nanoparticles have emerged as an attractive alternative to viral vectors for gene therapy applications, utilizing a range of lipid-based, polymeric, and inorganic materials. These materials can either encapsulate or be functionalized to bind nucleic acids and protect them from degradation. To effectively elicit changes to gene expression, the nanoparticle carrier needs to undergo a series of steps intracellularly, from interacting with the cellular membrane to facilitate cellular uptake to endosomal escape and nucleic acid release. Adjusting physiochemical properties of the nanoparticles, such as size, charge, and targeting ligands, can improve cellular uptake and ultimately gene delivery. Applications in the central nervous system (CNS; i.e., neurological diseases, brain cancers) face further extracellular barriers for a gene-carrying nanoparticle to surpass, with the most significant being the blood-brain barrier (BBB). Approaches to overcome these extracellular challenges to deliver nanoparticles into the CNS include systemic, intracerebroventricular, intrathecal, and intranasal administration. This review describes and compares different biomaterials for nonviral nanoparticle-mediated gene therapy to the CNS and explores challenges and recent preclinical and clinical developments in overcoming barriers to nanoparticle-mediated delivery to the brain. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Neurological Disease Therapeutic Approaches and Drug Discovery > Emerging Technologies Nanotechnology Approaches to Biology > Nanoscale Systems in Biology.
Collapse
Affiliation(s)
- Joanna Yang
- Departments of Biomedical Engineering, Ophthalmology, Oncology, Neurosurgery, Materials Science & Engineering, and Chemical & Biomolecular Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Kathryn M Luly
- Departments of Biomedical Engineering, Ophthalmology, Oncology, Neurosurgery, Materials Science & Engineering, and Chemical & Biomolecular Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Jordan J Green
- Departments of Biomedical Engineering, Ophthalmology, Oncology, Neurosurgery, Materials Science & Engineering, and Chemical & Biomolecular Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
4
|
Ford C, Burd CG. GOPC facilitates the sorting of syndecan-1 in polarized epithelial cells. Mol Biol Cell 2022; 33:ar86. [PMID: 35830596 PMCID: PMC9582621 DOI: 10.1091/mbc.e22-05-0165] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 06/30/2022] [Accepted: 07/06/2022] [Indexed: 11/11/2022] Open
Abstract
The trans-Golgi network must coordinate sorting and secretion of proteins and lipids to intracellular organelles and the plasma membrane. During polarization of epithelial cells, changes in the lipidome and the expression and distribution of proteins contribute to the formation of apical and basolateral plasma membrane domains. Previous studies using HeLa cells show that the syndecan-1 transmembrane domain confers sorting within sphingomyelin-rich vesicles in a sphingomyelin secretion pathway. In polarized Madin-Darby canine kidney cells, we reveal differences in the sorting of syndecan-1, whereupon the correct trafficking of the protein is not dependent on its transmembrane domain and changes in sphingomyelin content of cells during polarization. Instead, we reveal that correct basolateral targeting of syndecan-1 requires a full-length PDZ motif in syndecan-1 and the PDZ domain golgin protein GOPC. Moreover, we reveal changes in Golgi morphology elicited by GOPC overexpression. These results suggest that the role of GOPC in sorting syndecan-1 is indirect and likely due to GOPC effects on Golgi organization.
Collapse
Affiliation(s)
- Charlotte Ford
- Department of Cell Biology, Yale School of Medicine, New Haven, CT 06520
| | | |
Collapse
|
5
|
Azam S, Jakaria M, Kim J, Ahn J, Kim IS, Choi DK. Group I mGluRs in Therapy and Diagnosis of Parkinson’s Disease: Focus on mGluR5 Subtype. Biomedicines 2022; 10:biomedicines10040864. [PMID: 35453614 PMCID: PMC9032558 DOI: 10.3390/biomedicines10040864] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 04/05/2022] [Accepted: 04/06/2022] [Indexed: 11/16/2022] Open
Abstract
Metabotropic glutamate receptors (mGluRs; members of class C G-protein-coupled receptors) have been shown to modulate excitatory neurotransmission, regulate presynaptic extracellular glutamate levels, and modulate postsynaptic ion channels on dendritic spines. mGluRs were found to activate myriad signalling pathways to regulate synapse formation, long-term potentiation, autophagy, apoptosis, necroptosis, and pro-inflammatory cytokines release. A notorious expression pattern of mGluRs has been evident in several neurodegenerative diseases, including Alzheimer’s disease, Parkinson’s disease, Huntington’s disease, and schizophrenia. Among the several mGluRs, mGluR5 is one of the most investigated types of considered prospective therapeutic targets and potential diagnostic tools in neurodegenerative diseases and neuropsychiatric disorders. Recent research showed mGluR5 radioligands could be a potential tool to assess neurodegenerative disease progression and trace respective drugs’ kinetic properties. This article provides insight into the group I mGluRs, specifically mGluR5, in the progression and possible therapy for PD.
Collapse
Affiliation(s)
- Shofiul Azam
- Department of Applied Life Science, Graduate School, BK21 Program, Konkuk University, Chungju 27478, Korea; (S.A.); (M.J.); (J.K.); (J.A.)
| | - Md. Jakaria
- Department of Applied Life Science, Graduate School, BK21 Program, Konkuk University, Chungju 27478, Korea; (S.A.); (M.J.); (J.K.); (J.A.)
- Melbourne Dementia Research Centre, The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC 3052, Australia
| | - JoonSoo Kim
- Department of Applied Life Science, Graduate School, BK21 Program, Konkuk University, Chungju 27478, Korea; (S.A.); (M.J.); (J.K.); (J.A.)
| | - Jaeyong Ahn
- Department of Applied Life Science, Graduate School, BK21 Program, Konkuk University, Chungju 27478, Korea; (S.A.); (M.J.); (J.K.); (J.A.)
| | - In-Su Kim
- Department of Biotechnology, College of Biomedical and Health Science, Research Institute of Inflammatory Disease (RID), Konkuk University, Chungju 27478, Korea
- Correspondence: (I.-S.K.); (D.-K.C.); Tel.: +82-43-840-3905 (I.-S.K.); +82-43-840-3610 (D.-K.C.); Fax: +82-43-840-3872 (D.-K.C.)
| | - Dong-Kug Choi
- Department of Applied Life Science, Graduate School, BK21 Program, Konkuk University, Chungju 27478, Korea; (S.A.); (M.J.); (J.K.); (J.A.)
- Department of Biotechnology, College of Biomedical and Health Science, Research Institute of Inflammatory Disease (RID), Konkuk University, Chungju 27478, Korea
- Correspondence: (I.-S.K.); (D.-K.C.); Tel.: +82-43-840-3905 (I.-S.K.); +82-43-840-3610 (D.-K.C.); Fax: +82-43-840-3872 (D.-K.C.)
| |
Collapse
|
6
|
Group I Metabotropic Glutamate Receptors and Interacting Partners: An Update. Int J Mol Sci 2022; 23:ijms23020840. [PMID: 35055030 PMCID: PMC8778124 DOI: 10.3390/ijms23020840] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 01/09/2022] [Accepted: 01/10/2022] [Indexed: 12/21/2022] Open
Abstract
Group I metabotropic glutamate (mGlu) receptors (mGlu1/5 subtypes) are G protein-coupled receptors and are broadly expressed in the mammalian brain. These receptors play key roles in the modulation of normal glutamatergic transmission and synaptic plasticity, and abnormal mGlu1/5 signaling is linked to the pathogenesis and symptomatology of various mental and neurological disorders. Group I mGlu receptors are noticeably regulated via a mechanism involving dynamic protein-protein interactions. Several synaptic protein kinases were recently found to directly bind to the intracellular domains of mGlu1/5 receptors and phosphorylate the receptors at distinct amino acid residues. A variety of scaffolding and adaptor proteins also interact with mGlu1/5. Constitutive or activity-dependent interactions between mGlu1/5 and their interacting partners modulate trafficking, anchoring, and expression of the receptors. The mGlu1/5-associated proteins also finetune the efficacy of mGlu1/5 postreceptor signaling and mGlu1/5-mediated synaptic plasticity. This review analyzes the data from recent studies and provides an update on the biochemical and physiological properties of a set of proteins or molecules that interact with and thus regulate mGlu1/5 receptors.
Collapse
|
7
|
Gu L, Luo WY, Xia N, Zhang JN, Fan JK, Yang HM, Wang MC, Zhang H. Upregulated mGluR5 induces ER stress and DNA damage by regulating the NMDA receptor subunit NR2B. J Biochem 2021; 171:349-359. [PMID: 34908130 DOI: 10.1093/jb/mvab140] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Accepted: 12/06/2021] [Indexed: 11/12/2022] Open
Abstract
Dysfunction caused by mGluR5 expression or activation is an important mechanism in the development of Parkinson's disease (PD). Early clinical studies on mGluR5 negative allosteric modulators have shown some limitations. It is therefore necessary to find a more specific approach to block mGluR5-mediated neurotoxicity. Here, we determined the role of NMDA receptor subunit NR2B in mGluR5-mediated ER stress and DNA damage. In vitro study, rotenone-induced ER stress and DNA damage were accompanied by an increase in mGluR5 expression, and overexpressed or activated mGluR5 with agonist CHPG induced ER stress and DNA damage, while blocking mGluR5 with antagonist MPEP alleviated the effect. Furthermore, the damage caused by CHPG was blocked by NMDA receptor antagonist MK-801. Additionally, rotenone or CHPG increased the p-Src and p-NR2B, which was inhibited by MPEP. Blocking p-Src or NR2B with PP2 or CP101,606 alleviated CHPG-induced ER stress and DNA damage. Overactivation of mGluR5 accompanied with the increase of p-Src and p-NR2B in the ER stress and DNA damage was found in rotenone-induced PD rat model. These findings suggest a new mechanism wherein mGluR5 induces ER stress and DNA damage through the NMDA receptor and propose NR2B as the molecular target for therapeutic strategy for PD.
Collapse
Affiliation(s)
- Li Gu
- Department of Neurobiology, School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Beijing Institute for Brain Disorders, Capital Medical University, Beijing 100069, China
| | - Wen-Yuan Luo
- Department of Neurobiology, School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Beijing Institute for Brain Disorders, Capital Medical University, Beijing 100069, China
| | - Ning Xia
- Department of Neurobiology, School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Beijing Institute for Brain Disorders, Capital Medical University, Beijing 100069, China.,Department of neurology, Massachusetts General Hospital, Boston, MA, USA
| | - Jian-Nan Zhang
- Department of Neurobiology, School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Beijing Institute for Brain Disorders, Capital Medical University, Beijing 100069, China
| | - Jing-Kai Fan
- Department of Neurobiology, School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Beijing Institute for Brain Disorders, Capital Medical University, Beijing 100069, China
| | - Hui-Min Yang
- Department of Neurobiology, School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Beijing Institute for Brain Disorders, Capital Medical University, Beijing 100069, China
| | - Meng-Chen Wang
- School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Hong Zhang
- Department of Neurobiology, School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Beijing Institute for Brain Disorders, Capital Medical University, Beijing 100069, China
| |
Collapse
|
8
|
Bashkatova V. Metabotropic glutamate receptors and nitric oxide in dopaminergic neurotoxicity. World J Psychiatry 2021; 11:830-840. [PMID: 34733645 PMCID: PMC8546773 DOI: 10.5498/wjp.v11.i10.830] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 04/11/2021] [Accepted: 08/03/2021] [Indexed: 02/06/2023] Open
Abstract
Dopaminergic neurotoxicity is characterized by damage and death of dopaminergic neurons. Parkinson's disease (PD) is a neurodegenerative disorder that primarily involves the loss of dopaminergic neurons in the substantia nigra. Therefore, the study of the mechanisms, as well as the search for new targets for the prevention and treatment of neurodegenerative diseases, is an important focus of modern neuroscience. PD is primarily caused by dysfunction of dopaminergic neurons; however, other neurotransmitter systems are also involved. Research reports have indicated that the glutamatergic system is involved in different pathological conditions, including dopaminergic neurotoxicity. Over the last two decades, the important functional interplay between dopaminergic and glutamatergic systems has stimulated interest in the possible role of metabotropic glutamate receptors (mGluRs) in the development of extrapyramidal disorders. However, the specific mechanisms driving these processes are presently unclear. The participation of the universal neuronal messenger nitric oxide (NO) in the mechanisms of dopaminergic neurotoxicity has attracted increased attention. The current paper aims to review the involvement of mGluRs and the contribution of NO to dopaminergic neurotoxicity. More precisely, we focused on studies conducted on the rotenone-induced PD model. This review is also an outline of our own results obtained using the method of electron paramagnetic resonance, which allows quantitation of NO radicals in brain structures.
Collapse
Affiliation(s)
- Valentina Bashkatova
- Laboratory of Physiology Reinforcements, Anokhin Institute of Normal Physiology, Moscow 125315, Russia
| |
Collapse
|
9
|
Klüssendorf M, Song I, Schau L, Morellini F, Dityatev A, Koliwer J, Kreienkamp HJ. The Golgi-Associated PDZ Domain Protein Gopc/PIST Is Required for Synaptic Targeting of mGluR5. Mol Neurobiol 2021; 58:5618-5634. [PMID: 34383253 PMCID: PMC8599212 DOI: 10.1007/s12035-021-02504-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 07/22/2021] [Indexed: 12/21/2022]
Abstract
In neuronal cells, many membrane receptors interact via their intracellular, C-terminal tails with PSD-95/discs large/ZO-1 (PDZ) domain proteins. Some PDZ proteins act as scaffold proteins. In addition, there are a few PDZ proteins such as Gopc which bind to receptors during intracellular transport. Gopc is localized at the trans-Golgi network (TGN) and binds to a variety of receptors, many of which are eventually targeted to postsynaptic sites. We have analyzed the role of Gopc by knockdown in primary cultured neurons and by generating a conditional Gopc knockout (KO) mouse line. In neurons, targeting of neuroligin 1 (Nlgn1) and metabotropic glutamate receptor 5 (mGlu5) to the plasma membrane was impaired upon depletion of Gopc, whereas NMDA receptors were not affected. In the hippocampus and cortex of Gopc KO animals, expression levels of Gopc-associated receptors were not altered, while their subcellular localization was disturbed. The targeting of mGlu5 to the postsynaptic density was reduced, coinciding with alterations in mGluR-dependent synaptic plasticity and deficiencies in a contextual fear conditioning paradigm. Our data imply Gopc in the correct subcellular sorting of its associated mGlu5 receptor in vivo.
Collapse
Affiliation(s)
- Malte Klüssendorf
- Institute for Human Genetics, University Medical Center Hamburg Eppendorf, Hamburg, Germany
| | - Inseon Song
- Molecular Neuroplasticity Group, German Center for Neurodegenerative Diseases (DZNE), 39120, Magdeburg, Germany
| | - Lynn Schau
- Research Group Behavioral Biology, Center for Molecular Neurobiology, University Medical Center Hamburg Eppendorf, Hamburg, Germany
| | - Fabio Morellini
- Research Group Behavioral Biology, Center for Molecular Neurobiology, University Medical Center Hamburg Eppendorf, Hamburg, Germany
| | - Alexander Dityatev
- Molecular Neuroplasticity Group, German Center for Neurodegenerative Diseases (DZNE), 39120, Magdeburg, Germany
- Center for Behavioral Brain Sciences (CBBS), 39106, Magdeburg, Germany
- Medical Faculty, Otto-Von-Guericke University, 39120, Magdeburg, Germany
| | - Judith Koliwer
- Institute for Human Genetics, University Medical Center Hamburg Eppendorf, Hamburg, Germany
| | - Hans-Jürgen Kreienkamp
- Institute for Human Genetics, University Medical Center Hamburg Eppendorf, Hamburg, Germany.
| |
Collapse
|
10
|
Khedr MA, Abu-Zied KM, Zaghary WA, Aly AS, Shouman DN, Haffez H. Novel thienopyrimidine analogues as potential metabotropic glutamate receptors inhibitors and anticancer activity: Synthesis, In-vitro, In-silico, and SAR approaches. Bioorg Chem 2021; 109:104729. [PMID: 33676314 DOI: 10.1016/j.bioorg.2021.104729] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2020] [Revised: 01/12/2021] [Accepted: 02/06/2021] [Indexed: 12/14/2022]
Abstract
There is a continuous need in drug development approach for synthetic anticancer analogues with new therapeutic targets to diminish chemotherapeutic resistance of cancer cells. This study presents new group of synthetic thienopyrimidine analogues (1-9) aims as mGluR-1 inhibitors with anticancer activity. In-vitro antiproliferative assessment was carried out using viability assay against cancer cell lines (MCF-7, A-549 and PC-3) compared to WI-38 normal cell line. Analogues showed variable anticancer activity with IC50 ranging from 6.60 to 121 µg/mL with compound 7b is the most potent analogue against the three cancer cell lines (MCF-7; 6.57 ± 0.200, A-549; 6.31 ± 0.400, PC-3;7.39 ± 0.500 µg/mL) compared to Doxorubicin, 5-Flurouracil and Riluzole controls. Selected compounds were tested as mGluR-1 inhibitors in MCF-7 cell line and results revealed compound 7b induced significant reduction in extracellular glutamate release (IC50; 4.96 ± 0.700 µM) compared to other analogues and next to Riluzole (IC50; 2.80 ± 0.500 µM) of the same suggested mode of action. Furthermore, both cell cycle and apoptosis assays confirmed the potency of compound 7b for early apoptosis of MCF-7 at G2/M phase and apoptotic positive cell shift to (91.4%) compared to untreated control (19.6%) and Raptinal positive control (51.4%). On gene expression level, compound 7b induced over-expression of extrinsic (FasL, TNF-α and Casp-8), intrinsic (Cyt-C, Casp-3, Bax) apoptotic genes with down-regulation of anti-apoptotic Bcl-2 gene with boosted Bax/Bcl-2 ratio to 2.6-fold increase. Molecular docking and dynamic studies confirmed the biological potency through strong binding and stability modes of 7b where it was faster in reaching the equilibrium point and achieving the stability than Riluzole over 20 ns MD. These results suggest compound 7b as a promising mGluR inhibitory scaffold with anticancer activity that deserves further optimization and in-depth In-vivo and clinical investigations.
Collapse
Affiliation(s)
- Mohammed A Khedr
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Helwan University, P.O. Box 11795, Cairo, Egypt.
| | - Khadiga M Abu-Zied
- Photochemistry Department (Heterocyclic Unit), National Research Centre, Dokki, Giza 12622, Egypt
| | - Wafaa A Zaghary
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Helwan University, P.O. Box 11795, Cairo, Egypt.
| | - Ahmed S Aly
- Photochemistry Department (Heterocyclic Unit), National Research Centre, Dokki, Giza 12622, Egypt
| | - Dina N Shouman
- Family Medicine Center, Egyptian Ministry of Health and Population, Dakahlia, Egypt
| | - Hesham Haffez
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy, Helwan University, P.O. Box 11795, Cairo, Egypt; Center of Scientific Excellence "Helwan Structural Biology Research, (HSBR)", Helwan University, Cairo 11795, Egypt
| |
Collapse
|
11
|
Wang Y, Gu L, Yang HM, Zhang H. Cystic fibrosis transmembrane conductance regulator-associated ligand protects dopaminergic neurons by differentially regulating metabotropic glutamate receptor 5 in the progression of neurotoxin 6-hydroxydopamine-induced Parkinson's disease model. Neurotoxicology 2021; 84:14-29. [PMID: 33571554 DOI: 10.1016/j.neuro.2021.02.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2020] [Revised: 12/21/2020] [Accepted: 02/04/2021] [Indexed: 10/22/2022]
Abstract
Due to limitations in early diagnosis and treatments of Parkinson's disease (PD), it is necessary to explore the neuropathological changes that occur early in PD progression and to design neuroprotective therapies to prevent or delay the ongoing degeneration process. Metabotropic glutamate receptor 5 (mGlu5) has shown both diagnostic and therapeutic potential in preclinical studies on PD. Clinical trials using mGlu5 negative allosteric modulators to treat PD have, however, raised limitations about the neuroprotective role of mGlu5. It is likely that mGlu5 has different regulatory roles in different stages of PD. Here, we investigated a protective role of cystic fibrosis transmembrane conductance regulator-associated ligand (CAL) in the progression of PD by differential regulation of mGlu5 expression and activity to protect against 6-hydroxydopamine (6-OHDA)-induced neurotoxicity. Following treatment with 6-OHDA, mGlu5 and CAL expressions were elevated in the early stage and reduced in the late stage, both in vitro and in vivo. Activation of mGlu5 in the early stage by (RS)-2-chloro-5-hydroxyphenylglycine, or blocking mGlu5 in the late stage by 2-methyl-6-(phenylethynyl) pyridine, increased cell survival and inhibited apoptosis, but these effects were significantly weakened by knockdown of CAL. CAL alleviated 6-OHDA-induced neurotoxicity by regulating mGlu5-mediated signaling pathways, thereby maintaining the physiological function of mGlu5 in different disease stages. In PD rat model, CAL deficiency aggravated 6-OHDA toxicity on dopaminergic neurons and increased motor dysfunction because of lack of regulation of mGlu5 activity. These data reveal a potential mechanism by which CAL specifically regulates the opposite activity of mGlu5 in progression of PD to protect against neurotoxicity, suggesting that CAL is a favorable endogenous target for the treatment of PD.
Collapse
Affiliation(s)
- Yuan Wang
- Department of Neurobiology, School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Beijing Institute for Brain Disorders, Capital Medical University, No. 10 Xitoutiao, You An Men, Beijing, 100069, China
| | - Li Gu
- Department of Neurobiology, School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Beijing Institute for Brain Disorders, Capital Medical University, No. 10 Xitoutiao, You An Men, Beijing, 100069, China
| | - Hui Min Yang
- Department of Neurobiology, School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Beijing Institute for Brain Disorders, Capital Medical University, No. 10 Xitoutiao, You An Men, Beijing, 100069, China
| | - Hong Zhang
- Department of Neurobiology, School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Beijing Institute for Brain Disorders, Capital Medical University, No. 10 Xitoutiao, You An Men, Beijing, 100069, China.
| |
Collapse
|
12
|
Abdelaal MR, Soror SH, Elnagar MR, Haffez H. Revealing the Potential Application of EC-Synthetic Retinoid Analogues in Anticancer Therapy. Molecules 2021; 26:506. [PMID: 33477997 PMCID: PMC7835894 DOI: 10.3390/molecules26020506] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 12/31/2020] [Accepted: 01/08/2021] [Indexed: 12/12/2022] Open
Abstract
(1) Background and Aim: All-trans retinoic acid (ATRA) induces differentiation and inhibits growth of many cancer cells. However, resistance develops rapidly prompting the urgent need for new synthetic and potent derivatives. EC19 and EC23 are two synthetic retinoids with potent stem cell neuro-differentiation activity. Here, these compounds were screened for their in vitro antiproliferative and cytotoxic activity using an array of different cancer cell lines. (2) Methods: MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) assay, AV/PI (annexin V-fluorescein isothiocyanate (FITC)/propidium iodide (PI)), cell cycle analysis, immunocytochemistry, gene expression analysis, Western blotting, measurement of glutamate and total antioxidant concentrations were recruited. (3) Results: HepG2, Caco-2, and MCF-7 were the most sensitive cell lines; HepG2 (ATRA; 36.2, EC19; 42.2 and EC23; 0.74 µM), Caco-2 (ATRA; 58.0, EC19; 10.8 and EC23; 14.7 µM) and MCF-7 (ATRA; 99.0, EC19; 9.4 and EC23; 5.56 µM). Caco-2 cells were selected for further biochemical investigations. Isobologram analysis revealed the combined synergistic effects with 5-fluorouracil with substantial reduction in IC50. All retinoids induced apoptosis but EC19 had higher potency, with significant cell cycle arrest at subG0-G1, -S and G2/M phases, than ATRA and EC23. Moreover, EC19 reduced cellular metastasis in a transwell invasion assay due to overexpression of E-cadherin, retinoic acid-induced 2 (RAI2) and Werner (WRN) genes. (4) Conclusion: The present study suggests that EC-synthetic retinoids, particularly EC19, can be effective, alone or in combinations, for potential anticancer activity to colorectal cancer. Further in vivo studies are recommended to pave the way for clinical applications.
Collapse
Affiliation(s)
- Mohamed R. Abdelaal
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy, Helwan University, Cairo 11795, Egypt; (M.R.A.); (S.H.S.)
- Center of Scientific Excellence “Helwan Structural Biology Research, (HSBR)”, Helwan University, Cairo 11795, Egypt
| | - Sameh H. Soror
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy, Helwan University, Cairo 11795, Egypt; (M.R.A.); (S.H.S.)
- Center of Scientific Excellence “Helwan Structural Biology Research, (HSBR)”, Helwan University, Cairo 11795, Egypt
| | - Mohamed R. Elnagar
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Al-Azhar University, Cairo 11823, Egypt;
| | - Hesham Haffez
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy, Helwan University, Cairo 11795, Egypt; (M.R.A.); (S.H.S.)
- Center of Scientific Excellence “Helwan Structural Biology Research, (HSBR)”, Helwan University, Cairo 11795, Egypt
| |
Collapse
|
13
|
Shi ZL, Zhang H, Fan ZY, Ma W, Song YZ, Li M, Li TQ, Cao SX, Feng GJ. Long noncoding RNA LINC00314 facilitates osteogenic differentiation of adipose-derived stem cells through the hsa-miR-129-5p/GRM5 axis via the Wnt signaling pathway. Stem Cell Res Ther 2020; 11:240. [PMID: 32552820 PMCID: PMC7302136 DOI: 10.1186/s13287-020-01754-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2020] [Revised: 05/28/2020] [Accepted: 06/01/2020] [Indexed: 12/11/2022] Open
Abstract
Background Many studies have shown that long noncoding RNAs (lncRNAs) are closely related to the stimulation of osteogenic differentiation of adipose-derived stem cells (ADSCs) and the prevention of osteoporosis. Current research aimed to investigate the novel lncRNA and explored the function and molecular mechanism of the LINC00314/miR-129-5p/GRM5 axis in regulating osteogenic differentiation of ADSCs. Methods LncRNA and miRNA sequencing was performed in normal and osteogenic differentiation-induced ADSCs (osteogenic group). Abnormally expressed lncRNAs and miRNAs were obtained by the R software and the relative expression of LINC00314, miR-129-5p, and GRM5 during osteogenic induction was measured by RT-PCR. ADSCs were then transfected with pcDNA3.1-sh-LINC00314 and agomiR-129-5p. Alizarin red staining (ARS) and alkaline phosphatase (ALP) staining were performed to identify the mechanism of the LINC00314/miR-129-5p/GRM5 axis in regulating osteogenic differentiation of ADSCs. Results LINC00314 was significantly upregulated in the group of osteogenic-induced ADSCs. LINC00314 and GRM5 mimics increased the early and late markers of osteogenic differentiation, which manifest in not only the markedly increased ALP activity but also higher calcium deposition, while miR-129-5p mimic had the opposite effects. LINC00314 directly targeted miR-129-5p through luciferase reporter assay, and miR-129-5p suppressed GRM5 expression. Moreover, the LINC00314/miR-129-5p/GRM5 regulatory axis activated the Wnt/β-catenin signaling pathway. Conclusions LINC00314 confers contributory function in the osteogenic differentiation of ADSCs and thus the LINC00314/miR-129-5p/GRM5 axis may be a novel mechanism for osteogenic-related disease.
Collapse
Affiliation(s)
- Zheng-Liang Shi
- Department of Orthopedics, The Second Hospital of Hebei Medical University, No. 215, Hepingxi Road, Shijiazhuang, 050000, Hebei Province, China
| | - Hua Zhang
- Department of Orthopedics, The Second Hospital of Hebei Medical University, No. 215, Hepingxi Road, Shijiazhuang, 050000, Hebei Province, China.
| | - Zhi-Yong Fan
- Department of Orthopedics, The Second Hospital of Hebei Medical University, No. 215, Hepingxi Road, Shijiazhuang, 050000, Hebei Province, China
| | - Wei Ma
- Department of Orthopedics, The Second Hospital of Hebei Medical University, No. 215, Hepingxi Road, Shijiazhuang, 050000, Hebei Province, China
| | - Yong-Zhou Song
- Department of Orthopedics, The Second Hospital of Hebei Medical University, No. 215, Hepingxi Road, Shijiazhuang, 050000, Hebei Province, China
| | - Ming Li
- Department of Orthopedics, The Second Hospital of Hebei Medical University, No. 215, Hepingxi Road, Shijiazhuang, 050000, Hebei Province, China
| | - Tong-Qiu Li
- Department of Orthopedics, The Second Hospital of Hebei Medical University, No. 215, Hepingxi Road, Shijiazhuang, 050000, Hebei Province, China
| | - Shu-Xing Cao
- Department of Orthopedics, The Second Hospital of Hebei Medical University, No. 215, Hepingxi Road, Shijiazhuang, 050000, Hebei Province, China
| | - Guo-Jun Feng
- Department of Orthopedics, The Second Hospital of Hebei Medical University, No. 215, Hepingxi Road, Shijiazhuang, 050000, Hebei Province, China
| |
Collapse
|