1
|
Zhou Y, Wang Y, Liu J, Bai Y, Ma J, Niu MM, Li J, Jiang H. Design, synthesis, and biological evaluation of novel and highly potent peptides targeting syntenin. Eur J Med Chem 2025; 289:117446. [PMID: 40020425 DOI: 10.1016/j.ejmech.2025.117446] [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: 12/08/2024] [Revised: 02/23/2025] [Accepted: 02/24/2025] [Indexed: 03/03/2025]
Abstract
Syntenin, an intracellular scaffold protein, plays a critical role in renal cell carcinoma (RCC) progression, underscoring its potential as a therapeutic target. Herein, we report a novel, highly efficient, and stable peptide inhibitor (PDPP-3) that exhibits excellent inhibitory effects on syntenin. We have constructed a combined virtual screening scheme based on pharmacophore modeling and molecular docking to identify six potential d-amino acid-containing peptide inhibitors targeting syntenin. Among them, PDPP-3 showed the best inhibitory activity against syntenin. Binding affinity experiments and biostability experiments indicated that the interaction between PDPP-3 and syntenin displayed nanomolar-level binding affinity (Kd = 6.15 ± 0.12 nM) and superior biostability in serum. Molecular dynamics simulation results further confirmed that PDPP-3 could stably bind to the active site of syntenin. Additionally, cytotoxicity test results showed that PDPP-3 exhibited potent inhibitory effects on various types of renal cancer cells, with the best inhibitory effect on SK-RC-20 cells. More importantly, PDPP-3 significantly downregulates the expression of matrix metalloenzymes MT1-MMP and MMP2, which are pivotal for tumor invasion, and demonstrates inhibitory effects on tumor growth in SK-RC-20 derived xenografts. These data suggest that PDPP-3 may be a very promising candidate drug for the treatment of RCC.
Collapse
Affiliation(s)
- Yang Zhou
- Department of Pathology, The Second People's Hospital of Changzhou, The Third Affiliated Hospital of Nanjing Medical University, Changzhou, 213100, China.
| | - Yuting Wang
- Department of Pharmaceutical Analysis, China Pharmaceutical University, Nanjing, 211198, China
| | - Juanjuan Liu
- Department of Pharmacy, Taizhou School of Clinical Medicine, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou, 225300, China
| | - Yu Bai
- Department of Urology, The Second People's Hospital of Changzhou, The Third Affiliated Hospital of Nanjing Medical University, Changzhou, 213100, China; Department of Urology, Traditional Chinese Medicine Hospital of Gonghe County, Gonghe County, Hainan Prefecture, 813099, China
| | - Jinliang Ma
- Department of Urology, Traditional Chinese Medicine Hospital of Gonghe County, Gonghe County, Hainan Prefecture, 813099, China
| | - Miao-Miao Niu
- Department of Pharmaceutical Analysis, China Pharmaceutical University, Nanjing, 211198, China
| | - Jindong Li
- Department of Pharmacy, Taizhou School of Clinical Medicine, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou, 225300, China.
| | - Haijing Jiang
- Drug Clinical Trial Facility Office, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221000, China.
| |
Collapse
|
2
|
Wankowicz SA, Fraser JS. Advances in uncovering the mechanisms of macromolecular conformational entropy. Nat Chem Biol 2025; 21:623-634. [PMID: 40275100 DOI: 10.1038/s41589-025-01879-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Accepted: 03/10/2025] [Indexed: 04/26/2025]
Abstract
During protein folding, proteins transition from a disordered polymer into a globular structure, markedly decreasing their conformational degrees of freedom, leading to a substantial reduction in entropy. Nonetheless, folded proteins retain substantial entropy as they fluctuate between the conformations that make up their native state. This residual entropy contributes to crucial functions like binding and catalysis, supported by growing evidence primarily from NMR and simulation studies. Here, we propose three major ways that macromolecules use conformational entropy to perform their functions; first, prepaying entropic cost through ordering of the ground state; second, spatially redistributing entropy, in which a decrease in entropy in one area is reciprocated by an increase in entropy elsewhere; third, populating catalytically competent ensembles, in which conformational entropy within the enzymatic scaffold aids in lowering transition state barriers. We also provide our perspective on how solving the current challenge of structurally defining the ensembles encoding conformational entropy will lead to new possibilities for controlling binding, catalysis and allostery.
Collapse
Affiliation(s)
- Stephanie A Wankowicz
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA, USA.
| | - James S Fraser
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
3
|
Li Q, Zhang BH, Chen Q, Fu Y, Zuo X, Lu P, Zhang W, Wang B. Pathogenic variants in SHROOM3 associated with hemifacial microsomia. J Hum Genet 2025; 70:189-194. [PMID: 39875538 DOI: 10.1038/s10038-025-01317-1] [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: 05/15/2024] [Revised: 12/14/2024] [Accepted: 01/15/2025] [Indexed: 01/30/2025]
Abstract
Hemifacial microsomia (HFM) is a rare congenital disorder that affects facial symmetry, ear development, and other congenital anomalies. However, known causal genes account for only approximately 6% of patients, indicating the need to discover more pathogenic genes. Association tests demonstrated an association between common variants in SHROOM3 and HFM (P = 1.02E-4 for the lead SNP), while gene burden analysis revealed a significant enrichment of rare variants in HFM patients compared to healthy controls (P = 2.78E-5). We then evaluated the expression patterns of SHROOM3 and the consequences of its deleterious variants. Our study identified 7 deleterious variants in SHROOM3 among the 320 Chinese HFM patients and 2 deleterious variants in two HFM trios, respectively, suggesting a model of dominant inheritance with incomplete penetrance. These variants were predicted to significantly impact SHROOM3 function. Furthermore, the gene expression pattern of SHROOM3 in the pharyngeal arches and the presence of facial abnormalities in gene-edited mice suggest that SHROOM3 plays important roles in facial development. Our findings suggest that SHROOM3 is a likely pathogenic gene for HFM.
Collapse
Affiliation(s)
- Qin Li
- Department of Stomatology, Eye&ENT Hospital, Fudan University, Shanghai, 200031, China
| | - Bing-Hua Zhang
- Shanghai Xuhui District Dental Center, Shanghai, 200032, China
| | - Qi Chen
- Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100144, China
| | - Yaoyao Fu
- Department of facial plastic and reconstructive surgery, Eye&ENT Hospital, Fudan University, Shanghai, 200031, China
| | - Xiang Zuo
- Department of Stomatology, Eye&ENT Hospital, Fudan University, Shanghai, 200031, China
| | - Peng Lu
- Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100144, China
| | - Weiwei Zhang
- Department of Stomatology, Eye&ENT Hospital, Fudan University, Shanghai, 200031, China.
| | - Bingqing Wang
- Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100144, China.
| |
Collapse
|
4
|
Bae Y, Lee S, Kim A, Lee S, Kim S, Park S, Noh J, Ryoo K, Yi G, Park J, Hwang EM. Astrocytic NHERF-1 Increases Seizure Susceptibility by Inhibiting Surface Expression of TREK-1. Glia 2025; 73:720-736. [PMID: 39543986 PMCID: PMC11845839 DOI: 10.1002/glia.24644] [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: 06/06/2024] [Revised: 11/01/2024] [Accepted: 11/04/2024] [Indexed: 11/17/2024]
Abstract
Mature hippocampal astrocytes exhibit a linear current-to-voltage (I-V) K+ membrane conductance called passive conductance. It is estimated to enable astrocytes to keep potassium homeostasis in the brain. We previously reported that the TWIK-1/TREK-1 heterodimeric channels are crucial for astrocytic passive conductance. However, the regulatory mechanism of these channels by other binding proteins remains elusive. Here, we identified Na+/H+ exchange regulator-1 (NHERF-1), a protein highly expressed in astrocytes, as a novel interaction partner for these channels. NHERF-1 endogenously bound to TWIK-1/TREK-1 in hippocampal cultured astrocytes. When NHERF-1 is overexpressed or silenced, surface expression and activity of TWIK-1/TREK-1 heterodimeric channels are inhibited or enhanced, respectively. Furthermore, we confirmed that reduced astrocytic passive conductance by NHERF-1 overexpressing in the hippocampus increases kainic acid (KA)-induced seizure sensitivity. Taken together, these results suggest that NHERF-1 is a key regulator of TWIK-1/TREK-1 heterodimeric channels in astrocytes and suppression of TREK-1 surface expression by NHERF-1 increases KA-induced seizure susceptibility via reduction of astrocytic passive conductance.
Collapse
Affiliation(s)
- Yeonju Bae
- Brain Science InstituteKorea Institute of Science and Technology (KIST)SeoulRepublic of Korea
- School of Biosystems and Biomedical Sciences, College of Health SciencesKorea UniversitySeoulRepublic of Korea
| | - Soomin Lee
- Brain Science InstituteKorea Institute of Science and Technology (KIST)SeoulRepublic of Korea
- School of Biosystems and Biomedical Sciences, College of Health SciencesKorea UniversitySeoulRepublic of Korea
| | - Ajung Kim
- Brain Science InstituteKorea Institute of Science and Technology (KIST)SeoulRepublic of Korea
| | - Shinae Lee
- Brain Science InstituteKorea Institute of Science and Technology (KIST)SeoulRepublic of Korea
- Department of bio and Brain Engineering, College of EngineeringKAISTDaejeonRepublic of Korea
| | - Seong‐Seop Kim
- School of Biosystems and Biomedical Sciences, College of Health SciencesKorea UniversitySeoulRepublic of Korea
| | - Sunyoung Park
- School of Biosystems and Biomedical Sciences, College of Health SciencesKorea UniversitySeoulRepublic of Korea
| | - Junyeol Noh
- School of Biosystems and Biomedical Sciences, College of Health SciencesKorea UniversitySeoulRepublic of Korea
| | - Kanghyun Ryoo
- School of Biosystems and Biomedical Sciences, College of Health SciencesKorea UniversitySeoulRepublic of Korea
| | - Gwan‐Su Yi
- Department of bio and Brain Engineering, College of EngineeringKAISTDaejeonRepublic of Korea
| | - Jae‐Yong Park
- School of Biosystems and Biomedical Sciences, College of Health SciencesKorea UniversitySeoulRepublic of Korea
- Astrion, Inc.SeoulRepublic of Korea
| | - Eun Mi Hwang
- Brain Science InstituteKorea Institute of Science and Technology (KIST)SeoulRepublic of Korea
- Division of Bio‐Medical Science & Technology, KIST SchoolKorea University of Science and TechnologySeoulRepublic of Korea
| |
Collapse
|
5
|
Perelli RM, Dewars ER, Cope H, Behura AS, Ponek AQ, Sala AM, Zhang Z, Muralidharan P, Moya-Mendez ME, Berkman A, Monaco GG, Sullivan MC, Ezekian JE, Yang Q, Sun B, Kurzlechner LM, Asokan T, Breglio AM, Jay Campbell M, Spector ZZ, Rehder CW, Tang PC, James CA, Calkins H, Shashi V, Landstrom AP. TAX1BP3 Causes TRPV4-Mediated Autosomal Recessive Arrhythmogenic Cardiomyopathy. Circ Res 2025; 136:667-684. [PMID: 39963794 PMCID: PMC11949706 DOI: 10.1161/circresaha.124.325180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 02/01/2025] [Accepted: 02/05/2025] [Indexed: 03/05/2025]
Abstract
BACKGROUND Arrhythmogenic cardiomyopathy (ACM) is one of the leading causes of sudden cardiac death in children, young adults, and athletes and is characterized by the fibro-fatty replacement of the myocardium, predominantly of the right ventricle. Sixty percent of patients with ACM have a known genetic cause, but for the remainder, the pathogenesis is unknown. This lack of mechanistic understanding has slowed the development of disease-modifying therapies, and children with ACM have a high degree of morbidity and mortality. METHODS Induced pluripotent stem cells (iPSCs) from 3 family members were differentiated into cardiac myocytes (CMs). Calcium imaging was conducted by labeling calcium with CAL-520 and confocal imaging to capture calcium sparks after iPSC-CMs were electrically paced. A cardiac-specific, inducible knockout mouse (Tax1bp3-/-) was made and intracardiac electrophysiology studies conducted to observe arrhythmia inducibility following pacing. RESULTS We identified a kindred with multiple members affected by ACM cosegregating with biallelic variants in the gene TAX1BP3, which encodes the protein TAX1BP3 (Tax1-binding protein 3). iPSC-CMs derived from this kindred demonstrated increased intracellular lipid droplets, induction of TRPV4 (transient receptor potential vanilloid type 4) expression, and inducible TRPV4 current. This was associated with depletion of the intracellular sarcoplasmic reticulum Ca2+ store and increased RyR2 (ryanodine receptor 2)-mediated store Ca2+ leak and delayed afterdepolarizations, a known mechanism of Ca2+-mediated arrhythmogenesis. Similarly, Tax1bp3 cardiac-specific knockout mice had increased Ca2+ leak and were predisposed to ventricular arrhythmias compared with wild-type mice. Ca2+ leak in both the iPSC-CMs and mouse ventricular myocytes was rescued by small molecule TRPV4 inhibition. This strategy also effectively reduced Ca2+ leak in a PKP2 (plakophilin 2) p.His773AlafsX8 iPSC-CM model of ACM. CONCLUSIONS We conclude that TAX1BP3 is associated with rare autosomal recessive ACM through TRPV4-mediated Ca2+ leak from RyR2. Further, TRPV4 current inhibition has the potential to be a new therapeutic target for ACM.
Collapse
Affiliation(s)
- Robin M. Perelli
- Department of Cell Biology, Duke University School of Medicine, Durham, North Carolina, United States
- Department of Pediatrics, Division of Pediatric Cardiology, Duke University School of Medicine, Durham, North Carolina, United States
| | - Enya R. Dewars
- Department of Pediatrics, Division of Pediatric Cardiology, Duke University School of Medicine, Durham, North Carolina, United States
- Department of Cell and Molecular Biology, Duke University School of Medicine, Durham, North Carolina, United States
| | - Heidi Cope
- Department of Pediatrics, Division of Pediatric Cardiology, UT Southwestern Medical Center, Dallas, Texas, United States
| | - Alexander S. Behura
- Department of Pediatrics, Division of Pediatric Cardiology, Duke University School of Medicine, Durham, North Carolina, United States
| | - Anna Q. Ponek
- Department of Pediatrics, Division of Pediatric Cardiology, Duke University School of Medicine, Durham, North Carolina, United States
| | - Angelina M. Sala
- Department of Pediatrics, Division of Pediatric Cardiology, Duke University School of Medicine, Durham, North Carolina, United States
| | - Zhushan Zhang
- Department of Pediatrics, Division of Pediatric Cardiology, Duke University School of Medicine, Durham, North Carolina, United States
| | - Padmapriya Muralidharan
- Department of Pediatrics, Division of Pediatric Cardiology, Duke University School of Medicine, Durham, North Carolina, United States
| | - Mary E. Moya-Mendez
- Department of Pediatrics, Division of Pediatric Cardiology, Duke University School of Medicine, Durham, North Carolina, United States
| | - Amy Berkman
- Department of Pediatrics, Division of Pediatric Cardiology, Duke University School of Medicine, Durham, North Carolina, United States
| | - Gabrielle G. Monaco
- Department of Pediatrics, Division of Pediatric Cardiology, Duke University School of Medicine, Durham, North Carolina, United States
| | - Molly C. Sullivan
- Department of Pediatrics, Division of Pediatric Cardiology, Duke University School of Medicine, Durham, North Carolina, United States
| | - Jordan E. Ezekian
- Department of Pediatrics, Division of Pediatric Cardiology, UT Southwestern Medical Center, Dallas, Texas, United States
| | - Qixin Yang
- Department of Pediatrics, Division of Pediatric Cardiology, Duke University School of Medicine, Durham, North Carolina, United States
| | - Bo Sun
- Department of Pediatrics, Division of Pediatric Cardiology, Duke University School of Medicine, Durham, North Carolina, United States
| | - Leonie M. Kurzlechner
- Department of Pediatrics, Division of Pediatric Cardiology, Duke University School of Medicine, Durham, North Carolina, United States
| | - Tulsi Asokan
- Department of Pediatrics, Division of Pediatric Cardiology, Duke University School of Medicine, Durham, North Carolina, United States
| | - Andrew M. Breglio
- Department of Pediatrics, Division of Pediatric Cardiology, Duke University School of Medicine, Durham, North Carolina, United States
- Department of Anesthesiology, Duke University School of Medicine, Durham, North Carolina, United States
| | - M. Jay Campbell
- Department of Pediatrics, Division of Pediatric Cardiology, Duke University School of Medicine, Durham, North Carolina, United States
| | - Zebulon Z. Spector
- Department of Pediatrics, Division of Pediatric Cardiology, Duke University School of Medicine, Durham, North Carolina, United States
| | - Catherine W. Rehder
- Department of Pathology and Duke University Health Systems Clinical Laboratories, Durham, North Carolina, United States
| | | | - Paul C. Tang
- Department of Cardiac Surgery, University of Michigan, Ann Arbor, Michigan, United States
| | - Cynthia A. James
- Department of Medicine, Johns Hopkins Medical Institutions, Baltimore, Maryland, United States
| | - Hugh Calkins
- Department of Medicine, Johns Hopkins Medical Institutions, Baltimore, Maryland, United States
| | - Vandana Shashi
- Department of Pediatrics, Division of Pediatric Cardiology, UT Southwestern Medical Center, Dallas, Texas, United States
| | - Andrew P. Landstrom
- Department of Cell Biology, Duke University School of Medicine, Durham, North Carolina, United States
- Department of Pediatrics, Division of Pediatric Cardiology, Duke University School of Medicine, Durham, North Carolina, United States
| |
Collapse
|
6
|
Zhou P, Zhang Y, Li K, Ye H, Mei L, Shang S. Computational analysis and protein engineering of artificial PDZ domain/self-binding peptide fusion biomacromolecular system with molecular switch functionality. Int J Biol Macromol 2025; 308:142432. [PMID: 40132705 DOI: 10.1016/j.ijbiomac.2025.142432] [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: 12/29/2024] [Revised: 03/10/2025] [Accepted: 03/21/2025] [Indexed: 03/27/2025]
Abstract
Self-binding peptides (SBPs) are peptide-dependent intramolecular interactions described by our group, which conceptualize the short peptide segment within a monomeric protein as able to perform biological function by its dynamic and reversible binding to a cognate domain partner in the same monomer. Previously, we offered evidences that the SBPs represent a new biomolecular dynamic phenomenon that works across folding and binding, and also proposed the SBPs as potential drug targets for pharmacological intervention. Here, we attempted to model and design artificial protein systems containing SBP with molecular switch functionality by using computational peptidology strategies and protein engineering methods. In the procedure, the free decapeptide ligands were fused to the C-terminal tail of human CAL PDZ domain through a flexible polypeptide linker, thus resulting in a number of PDZ/SBP fusion biomacromolecular systems. The intramolecular binding event of SBP to PDZ was observed in the fusion systems as well as between the free decapeptide and PDZ. We carefully examined linker effects on the intramolecular binding event and systematically optimized the length and composition of linker to improve binding. Computational analysis suggested dynamics, but not thermodynamics, primarily responsible for the binding of SBP to PDZ. The linker plays an important role, as it restrains the SBP within a small local spatial region nearby the binding site of PDZ. The term effective concentration (EC) was defined to characterize the high local concentration of SBP at a small region due to the linker restriction, which improves the binding probability of SBP to PDZ from a dynamics point of view. The CAL PDZ/SBP(iCAL36-K-6) fusion protein system with poly(G)8 linker can work as designed well, where the SBP(iCAL36-K-6) dynamically binds to/unbinds from PDZ in a regulatable manner by externally controlling its C-terminal deamidation/amidation, thus exhibiting a typical molecular switch functionality.
Collapse
Affiliation(s)
- Peng Zhou
- Center for Informational Biology, School of Life Science and Technology, University of Electronic Science and Technology of China (UESTC), Chengdu 611731, China.
| | - Yunyi Zhang
- Center for Informational Biology, School of Life Science and Technology, University of Electronic Science and Technology of China (UESTC), Chengdu 611731, China
| | - Kexin Li
- Center for Informational Biology, School of Life Science and Technology, University of Electronic Science and Technology of China (UESTC), Chengdu 611731, China
| | - Haiyang Ye
- Center for Informational Biology, School of Life Science and Technology, University of Electronic Science and Technology of China (UESTC), Chengdu 611731, China
| | - Li Mei
- College of Culinary and Food Science Engineering, Sichuan Tourism University, Chengdu 610100, China.
| | - Shuyong Shang
- Institute of Ecological Environment Protection, Chengdu Normal University, Chengdu 611130, China
| |
Collapse
|
7
|
Santucci K, Cheng Y, Xu SM, Gao Y, Lindner G, Takenaka K, Janitz M. Discovery of Novel Protein-Coding and Long Non-coding Transcripts in Distinct Regions of the Human Brain. J Mol Neurosci 2025; 75:30. [PMID: 40048072 PMCID: PMC11885362 DOI: 10.1007/s12031-025-02316-9] [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: 11/20/2024] [Accepted: 02/06/2025] [Indexed: 03/09/2025]
Abstract
Recent improvements in the accuracy of long-read sequencing (LRS) technologies have expanded the scope for novel transcriptional isoform discovery. Additionally, these advancements have improved the precision of transcript quantification, enabling a more accurate reconstruction of complex splicing patterns and transcriptomes. Thus, this project aims to take advantage of these analytical developments for the discovery and analysis of RNA isoforms in the human brain. A set of novel transcript isoforms was compiled using three bioinformatic tools, quantifying their expression across eight replicates of the cerebellar hemisphere, five replicates of the frontal cortex, and six replicates of the putamen. By taking a subset of the novel isoforms consistent across all discovery methods, a set of 170 highly confident novel RNA isoforms was curated for downstream analysis. This set consisted of 104 messenger RNAs (mRNAs) and 66 long non-coding RNAs (lncRNAs) isoforms. The detailed structure, expression, and potential encoded proteins of novel mRNA isoform BambuTx321 have been further described as an exemplary representative. Additionally, the tissue-specific expression [mean counts per million (CPM) of 5.979] of novel lncRNA, BambuTx1299, in the cerebellar hemisphere was observed. Overall, this project has identified and annotated several novel RNA isoforms across diverse tissues of the human brain, providing insights into their expression patterns and investigating their potential functional roles. Thus, this project has contributed to a more comprehensive understanding of the brain's transcriptomic landscape for applications in basic research.
Collapse
Affiliation(s)
- Kristina Santucci
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, Australia
| | - Yuning Cheng
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, Australia
| | - Si-Mei Xu
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, Australia
| | - Yulan Gao
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, Australia
| | - Grace Lindner
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, Australia
| | - Konii Takenaka
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, Australia
| | - Michael Janitz
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, Australia.
| |
Collapse
|
8
|
Yang X, Huang YWA, Marshall J. Targeting TrkB-PSD-95 coupling to mitigate neurological disorders. Neural Regen Res 2025; 20:715-724. [PMID: 38886937 PMCID: PMC11433911 DOI: 10.4103/nrr.nrr-d-23-02000] [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: 12/07/2023] [Revised: 03/15/2024] [Accepted: 03/30/2024] [Indexed: 06/20/2024] Open
Abstract
Tropomyosin receptor kinase B (TrkB) signaling plays a pivotal role in dendritic growth and dendritic spine formation to promote learning and memory. The activity-dependent release of brain-derived neurotrophic factor at synapses binds to pre- or postsynaptic TrkB resulting in the strengthening of synapses, reflected by long-term potentiation. Postsynaptically, the association of postsynaptic density protein-95 with TrkB enhances phospholipase Cγ-Ca2+/calmodulin-dependent protein kinase II and phosphatidylinositol 3-kinase-mechanistic target of rapamycin signaling required for long-term potentiation. In this review, we discuss TrkB-postsynaptic density protein-95 coupling as a promising strategy to magnify brain-derived neurotrophic factor signaling towards the development of novel therapeutics for specific neurological disorders. A reduction of TrkB signaling has been observed in neurodegenerative disorders, such as Alzheimer's disease and Huntington's disease, and enhancement of postsynaptic density protein-95 association with TrkB signaling could mitigate the observed deficiency of neuronal connectivity in schizophrenia and depression. Treatment with brain-derived neurotrophic factor is problematic, due to poor pharmacokinetics, low brain penetration, and side effects resulting from activation of the p75 neurotrophin receptor or the truncated TrkB.T1 isoform. Although TrkB agonists and antibodies that activate TrkB are being intensively investigated, they cannot distinguish the multiple human TrkB splicing isoforms or cell type-specific functions. Targeting TrkB-postsynaptic density protein-95 coupling provides an alternative approach to specifically boost TrkB signaling at localized synaptic sites versus global stimulation that risks many adverse side effects.
Collapse
Affiliation(s)
- Xin Yang
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, RI, USA
| | - Yu-Wen Alvin Huang
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, RI, USA
- Department of Neurology, Warren Alpert Medical School of Brown University, Providence, RI, USA
- Center for Translational Neuroscience, Robert J. and Nancy D. Carney Institute for Brain Science and Brown Institute for Translational Science, Brown University, Providence, RI, USA
| | - John Marshall
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, RI, USA
| |
Collapse
|
9
|
Zhang Q, Clinton J, Westerlund K, Mim C. SMCT1 has a low affinity to PDZ domain containing 1 protein. MICROPUBLICATION BIOLOGY 2025; 2025:10.17912/micropub.biology.001502. [PMID: 40052138 PMCID: PMC11883465 DOI: 10.17912/micropub.biology.001502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Figures] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 02/07/2025] [Accepted: 02/11/2025] [Indexed: 03/09/2025]
Abstract
Sodium-coupled monocarboxylate transporter 1 (SMCT1) is a membrane transporter abundantly expressed in colon, kidney, thyroid, brain; and silenced in cancer cells. It transports monocarboxylic acids with little specificity into cells. Based on pulldown experiments, it was proposed that the scaffolding protein PDZ Domain Containing 1 (PDZK1) regulates its surface expression and increases SMCT1's transportation efficiency. Here, we performed pull-down assays, Surface Plasmon Resonance (SPR), and Micro Scale Thermophoresis (MST) to evaluate the affinity between SMCT1 and two PDZ domains in PDZK1. Our results show that SMCT1 binds to these PDZ domains. However, the estimated equilibrium dissociation constants are higher than in canonical PDZ domains and likely physiological not relevant.
Collapse
Affiliation(s)
- Qingyang Zhang
- Department of Protein Science, KTH Royal Institute of Technology, Stockholm, Stockholm, Sweden
- Department of Biomedical Engineering and Health Systems, KTH Royal Institute of Technology, Stockholm, Stockholm, Sweden
| | - Jacob Clinton
- Department of Protein Science, KTH Royal Institute of Technology, Stockholm, Stockholm, Sweden
| | - Kristina Westerlund
- Department of Protein Science, KTH Royal Institute of Technology, Stockholm, Stockholm, Sweden
| | - Carsten Mim
- Department of Protein Science, KTH Royal Institute of Technology, Stockholm, Stockholm, Sweden
| |
Collapse
|
10
|
Gidado KI, Adeshakin FO, Rabiu L, Zhang Z, Zhang G, Wan X. Multifaceted roles of DLG3/SAP102 in neurophysiology, neurological disorders and tumorigenesis. Neuroscience 2025; 565:192-201. [PMID: 39638232 DOI: 10.1016/j.neuroscience.2024.11.081] [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: 09/06/2024] [Revised: 11/15/2024] [Accepted: 11/30/2024] [Indexed: 12/07/2024]
Abstract
DLG3, also known as Synapse-associated protein 102 (SAP102), is essential for the organization and plasticity of excitatory synapses within the central nervous system (CNS). It plays a critical role in clustering and moving key components necessary for learning and memory processes. Mutations in the DLG3 gene, which result in truncated SAP102 proteins, have been associated with a range of neurological disorders, including X-linked intellectual disability (XLID), autism spectrum disorders (ASD), and schizophrenia, all of which can disrupt synaptic structure and cognitive functions. Abnormal SAP102 expression has also been linked to various psychiatric and neurodegenerative conditions, such as bipolar disorder, major depression, and Alzheimer's disease. Recent studies suggest that SAP102 influences cancer development and metastasis by regulating multiple signaling pathways, including the PI3K/AKT axis and the Hippo pathway. Moreover, SAP102 has been demonstrated to regulate tumor-induced bone pain through activating NMDA receptors. These findings highlight SAP102 as a promising therapeutic target for both neurological disorders and cancer. Therefore, further investigation into the regulatory roles of SAP102 in neural development and disease may lead to novel therapeutic approaches for treating synaptic disorders and managing cancer progression.
Collapse
Affiliation(s)
- Khalid Idris Gidado
- Center for Protein and Cell-based Drugs, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, PR China; University of Chinese Academy of Sciences, Beijing 100049, PR China
| | - Funmilayo O Adeshakin
- Center for Protein and Cell-based Drugs, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, PR China; University of Chinese Academy of Sciences, Beijing 100049, PR China
| | - Lawan Rabiu
- Center for Protein and Cell-based Drugs, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, PR China; University of Chinese Academy of Sciences, Beijing 100049, PR China
| | - Ziyang Zhang
- Center for Protein and Cell-based Drugs, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, PR China.
| | - Guizhong Zhang
- Center for Protein and Cell-based Drugs, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, PR China.
| | - Xiaochun Wan
- Center for Protein and Cell-based Drugs, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, PR China.
| |
Collapse
|
11
|
Chartier CA, Woods VA, Xu Y, van Vlimmeren AE, Johns AC, Jovanovic M, McDermott AE, Keedy DA, Shah NH. Allosteric regulation of the tyrosine phosphatase PTP1B by a protein-protein interaction. Protein Sci 2025; 34:e70016. [PMID: 39723820 DOI: 10.1002/pro.70016] [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: 07/16/2024] [Revised: 11/11/2024] [Accepted: 12/13/2024] [Indexed: 12/28/2024]
Abstract
The rapid identification of protein-protein interactions has been significantly enabled by mass spectrometry (MS) proteomics-based methods, including affinity purification-MS, crosslinking-MS, and proximity-labeling proteomics. While these methods can reveal networks of interacting proteins, they cannot reveal how specific protein-protein interactions alter protein function or cell signaling. For instance, when two proteins interact, there can be emergent signaling processes driven purely by the individual activities of those proteins being co-localized. Alternatively, protein-protein interactions can allosterically regulate function, enhancing or suppressing activity in response to binding. In this work, we investigate the interaction between the tyrosine phosphatase PTP1B and the adaptor protein Grb2, which have been annotated as binding partners in a number of proteomics studies. This interaction has been postulated to co-localize PTP1B with its substrate IRS-1 by forming a ternary complex, thereby enhancing the dephosphorylation of IRS-1 to suppress insulin signaling. Here, we report that Grb2 binding to PTP1B also allosterically enhances PTP1B catalytic activity. We show that this interaction is dependent on the proline-rich region of PTP1B, which interacts with the C-terminal SH3 domain of Grb2. Using NMR spectroscopy and hydrogen-deuterium exchange mass spectrometry (HDX-MS) we show that Grb2 binding alters PTP1B structure and/or dynamics. Finally, we use MS proteomics to identify other interactors of the PTP1B proline-rich region that may also regulate PTP1B function similarly to Grb2. This work presents one of the first examples of a protein allosterically regulating the enzymatic activity of PTP1B and lays the foundation for discovering new mechanisms of PTP1B regulation in cell signaling.
Collapse
Affiliation(s)
| | - Virgil A Woods
- Structural Biology Initiative, CUNY Advanced Science Research Center, New York, New York, USA
- PhD Program in Biochemistry, CUNY Graduate Center, New York, New York, USA
| | - Yunyao Xu
- Department of Chemistry, Columbia University, New York, New York, USA
| | - Anne E van Vlimmeren
- Department of Chemistry, Columbia University, New York, New York, USA
- Department of Biological Sciences, Columbia University, New York, New York, USA
| | - Andrew C Johns
- Department of Chemistry, Columbia University, New York, New York, USA
| | - Marko Jovanovic
- Department of Biological Sciences, Columbia University, New York, New York, USA
| | - Ann E McDermott
- Department of Chemistry, Columbia University, New York, New York, USA
| | - Daniel A Keedy
- Structural Biology Initiative, CUNY Advanced Science Research Center, New York, New York, USA
- Department of Chemistry and Biochemistry, City College of New York, New York, New York, USA
- PhD Programs in Biochemistry, Biology, & Chemistry, CUNY Graduate Center, New York, New York, USA
| | - Neel H Shah
- Department of Chemistry, Columbia University, New York, New York, USA
| |
Collapse
|
12
|
Wang B, Solinski AE, Radle MI, Peduzzi OM, Knox HL, Cui J, Maurya RK, Yennawar NH, Booker SJ. Structural Evidence for DUF512 as a Radical S-Adenosylmethionine Cobalamin-Binding Domain. ACS BIO & MED CHEM AU 2024; 4:319-330. [PMID: 39712206 PMCID: PMC11659888 DOI: 10.1021/acsbiomedchemau.4c00067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Revised: 10/04/2024] [Accepted: 10/07/2024] [Indexed: 12/24/2024]
Abstract
Cobalamin (Cbl)-dependent radical S-adenosylmethionine (SAM) enzymes constitute a large subclass of radical SAM (RS) enzymes that use Cbl to catalyze various types of reactions, the most common of which are methylations. Most Cbl-dependent RS enzymes contain an N-terminal Rossmann fold that aids Cbl binding. Recently, it has been demonstrated that the methanogenesis marker protein 10 (Mmp10) requires Cbl to methylate an arginine residue in the α-subunit of methyl coenzyme M reductase. However, Mmp10 contains a Cbl-binding domain in the C-terminal region of its primary structure that does not share significant sequence similarity with canonical RS Cbl-binding domains. Bioinformatic analysis of Mmp10 identified DUF512 (Domain of Unknown Function 512) as a potential Cbl-binding domain in RS enzymes. In this paper, four randomly selected DUF512-containing proteins from various organisms were overexpressed, purified, and shown to bind Cbl. X-ray crystal structures of DUF512-containing proteins from Clostridium sporogenes and Pyrococcus furiosus were determined, confirming their C-terminal Cbl-binding domains. The structure of the DUF512-containing protein from C. sporogenes is the first of an RS enzyme containing a PDZ domain. Its RS domain has an unprecedented β3α4 core, whereas most RS enzymes adopt a (βα)6 core. The DUF512-containing protein from P. furiosus has no PDZ domain, but its RS domain also has an uncommon (βα)5 core.
Collapse
Affiliation(s)
- Bo Wang
- Department
of Chemistry, The Pennsylvania State University, University Park, Pennsylvania 16802, United States
| | - Amy E. Solinski
- Department
of Chemistry, The Pennsylvania State University, University Park, Pennsylvania 16802, United States
| | - Matthew I. Radle
- Department
of Chemistry, The Pennsylvania State University, University Park, Pennsylvania 16802, United States
| | - Olivia M. Peduzzi
- Department
of Chemistry, The Pennsylvania State University, University Park, Pennsylvania 16802, United States
| | - Hayley L. Knox
- Department
of Chemistry, The Pennsylvania State University, University Park, Pennsylvania 16802, United States
| | - Jiayuan Cui
- Department
of Chemistry, The Pennsylvania State University, University Park, Pennsylvania 16802, United States
| | - Ravi K. Maurya
- Department
of Chemistry, The Pennsylvania State University, University Park, Pennsylvania 16802, United States
| | - Neela H. Yennawar
- The
Huck Institutes of the Life Sciences, The
Pennsylvania State University, University Park, Pennsylvania 16802, United States
| | - Squire J. Booker
- Department
of Chemistry, The Pennsylvania State University, University Park, Pennsylvania 16802, United States
- Department
of Biochemistry and Molecular Biology, The
Pennsylvania State University, University Park, Pennsylvania 16802, United States
- Howard
Hughes Medical Institute, The Pennsylvania
State University, University
Park, Pennsylvania 16802, United States
| |
Collapse
|
13
|
Yu Y, Liu J, Zhao Z, Lan X, Li L, Hu J, Zang YA, Zhang X, Chen J. Fish Snx27 promotes viral products by modulating the innate immune response and exosomal machinery. J Virol 2024; 98:e0097424. [PMID: 39494909 DOI: 10.1128/jvi.00974-24] [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: 06/06/2024] [Accepted: 09/29/2024] [Indexed: 11/05/2024] Open
Abstract
Viral nervous necrosis caused by the nervous necrosis virus (NNV) poses a significant threat to the global aquaculture industry. Developing preventive methods to minimize economic losses due to NNV infections is crucial. This study explored the role of the sorting nexin 27 (Snx27) gene, encoded by the orange-spotted grouper (Epinephelus coioides) and referred to as EcSnx27, as an immune regulator affecting red-spotted grouper nervous necrosis virus (RGNNV) infection in vitro. Our findings revealed that EcSnx27 negatively regulates interferon (IFN)-related cytokines and the promoter activities of fish ISRE and NF-κB. Furthermore, we identified the SNX-FERM and SNX-FERM-like domains as responsible for the interaction between EcSnx27 and RGNNV coat protein. Through the detection of viable virions associated with EcSnx27-containing exosomes, we propose that EcSnx27 may contribute to the release process of RGNNV by influencing the apoptosis-linked gene 2-interacting protein X (ALIX)-associated exosomal pathway. Consequently, our study suggests that EcSnx27 promotes RGNNV replication by inhibiting the IFN immune response and facilitating virus production and release through ALIX-mediated exosomal machinery.IMPORTANCERed grouper nervous necrosis virus (RGNNV), a member of the Nodaviridae family, has emerged as a significant cause of fish diseases worldwide, leading to high morbidity and mortality rates. This study investigated the sorting nexin 27 (Snx27) gene encoded by the orange-spotted grouper (Epinephelus coioides) on RGNNV infection in grouper kidney cells. Our findings revealed that EcSnx27 negatively regulated the interferon pathway, resulting in the promotion of RGNNV replication. Additionally, we observed that EcSnx27 could interact with apoptosis-linked gene 2-interacting protein X (ALIX) and the RGNNV coat protein, suggesting its potential involvement in viral release processes through modulation of the exosomal pathway. Our study identified EcSnx27 as a key target that RGNNV exploits to enhance viral production. This finding offers valuable insights into the immune evasion and viral release mechanisms of non-enveloped RNA viruses.
Collapse
Affiliation(s)
- Yepin Yu
- Guangdong Key Laboratory of Animal Conservation and Resource Utilization, Institute of Zoology, Guangdong Academy of Sciences, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, College of Fisheries, Guangdong Ocean University, Zhanjiang, Guangdong, China
| | - Jiaxin Liu
- Biosafety Laboratory, Guangdong Second Provincial General Hospital, Jinan University, Guangzhou, Guangdong, China
| | - Zhiwen Zhao
- Guangdong Key Laboratory of Animal Conservation and Resource Utilization, Institute of Zoology, Guangdong Academy of Sciences, Guangzhou, Guangdong, China
- College of Animal Sciences and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong, China
| | - Xiaoming Lan
- Guangdong Key Laboratory of Animal Conservation and Resource Utilization, Institute of Zoology, Guangdong Academy of Sciences, Guangzhou, Guangdong, China
- School of Life Science, Guangzhou University, Guangzhou, Guangdong, China
| | - Linmiao Li
- Guangdong Key Laboratory of Animal Conservation and Resource Utilization, Institute of Zoology, Guangdong Academy of Sciences, Guangzhou, Guangdong, China
| | - Junjie Hu
- School of Life Science, Guangzhou University, Guangzhou, Guangdong, China
| | - Ying-An Zang
- College of Animal Sciences and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong, China
| | - Xiujuan Zhang
- Guangdong Key Laboratory of Animal Conservation and Resource Utilization, Institute of Zoology, Guangdong Academy of Sciences, Guangzhou, Guangdong, China
| | - Jinping Chen
- Guangdong Key Laboratory of Animal Conservation and Resource Utilization, Institute of Zoology, Guangdong Academy of Sciences, Guangzhou, Guangdong, China
| |
Collapse
|
14
|
Zarzecka U, Skorko-Glonek J. Intricate Structure-Function Relationships: The Case of the HtrA Family Proteins from Gram-Negative Bacteria. Int J Mol Sci 2024; 25:13182. [PMID: 39684892 DOI: 10.3390/ijms252313182] [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: 11/14/2024] [Revised: 12/04/2024] [Accepted: 12/05/2024] [Indexed: 12/18/2024] Open
Abstract
Proteolytic enzymes play key roles in living organisms. Because of their potentially destructive action of degrading other proteins, their activity must be very tightly controlled. The evolutionarily conserved proteins of the HtrA family are an excellent example illustrating strategies for regulating enzymatic activity, enabling protease activation in response to an appropriate signal, and protecting against uncontrolled proteolysis. Because HtrA homologs play key roles in the virulence of many Gram-negative bacterial pathogens, they are subject to intense investigation as potential therapeutic targets. Model HtrA proteins from bacterium Escherichia coli are allosteric proteins with reasonably well-studied properties. Binding of appropriate ligands induces very large structural changes in these enzymes, including changes in the organization of the oligomer, which leads to the acquisition of the active conformation. Properly coordinated events occurring during the process of HtrA activation ensure proper functioning of HtrA and, consequently, ensure fitness of bacteria. The aim of this review is to present the current state of knowledge on the structure and function of the exemplary HtrA family proteins from Gram-negative bacteria, including human pathogens. Special emphasis is paid to strategies for regulating the activity of these enzymes.
Collapse
Affiliation(s)
- Urszula Zarzecka
- Department of General and Medical Biochemistry, Faculty of Biology, University of Gdansk, Wita Stwosza 59, 80-308 Gdansk, Poland
| | - Joanna Skorko-Glonek
- Department of General and Medical Biochemistry, Faculty of Biology, University of Gdansk, Wita Stwosza 59, 80-308 Gdansk, Poland
| |
Collapse
|
15
|
Patel NM, Ripoll L, Peach CJ, Ma N, Blythe EE, Vaidehi N, Bunnett NW, von Zastrow M, Sivaramakrishnan S. Myosin VI drives arrestin-independent internalization and signaling of GPCRs. Nat Commun 2024; 15:10636. [PMID: 39638791 PMCID: PMC11621365 DOI: 10.1038/s41467-024-55053-9] [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: 05/30/2024] [Accepted: 11/26/2024] [Indexed: 12/07/2024] Open
Abstract
G protein-coupled receptor (GPCR) endocytosis is canonically associated with β-arrestins. Here, we delineate a β-arrestin-independent endocytic pathway driven by the cytoskeletal motor, myosin VI. Myosin VI engages GIPC, an adaptor protein that binds a PDZ sequence motif present at the C-terminus of several GPCRs. Using the D2 dopamine receptor (D2R) as a prototype, we find that myosin VI regulates receptor endocytosis, spatiotemporal localization, and signaling. We find that access to the D2R C-tail for myosin VI-driven internalization is controlled by an interaction between the C-tail and the third intracellular loop of the receptor. Agonist efficacy, co-factors, and GIPC expression modulate this interaction to tune agonist trafficking. Myosin VI is differentially regulated by distinct GPCR C-tails, suggesting a mechanism to shape spatiotemporal signaling profiles in different ligand and physiological contexts. Our biophysical and structural insights may advance orthogonal therapeutic strategies for targeting GPCRs through cytoskeletal motor proteins.
Collapse
Affiliation(s)
- Nishaben M Patel
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, MN, USA
| | - Léa Ripoll
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, San Francisco, CA, USA
| | - Chloe J Peach
- Department of Molecular Pathobiology, New York University, New York, NY, USA
- School of Life Sciences, Centre of Membrane Proteins and Receptors (COMPARE), University of Nottingham, Nottingham, UK
| | - Ning Ma
- Irell and Manella Graduate School of Biological Sciences, Beckman Research Institute of the City of Hope, Duarte, CA, USA
- Department of Computational and Quantitative Medicine, Beckman Research Institute of the City of Hope, Duarte, CA, USA
| | - Emily E Blythe
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, San Francisco, CA, USA
| | - Nagarajan Vaidehi
- Irell and Manella Graduate School of Biological Sciences, Beckman Research Institute of the City of Hope, Duarte, CA, USA
- Department of Computational and Quantitative Medicine, Beckman Research Institute of the City of Hope, Duarte, CA, USA
| | - Nigel W Bunnett
- Department of Molecular Pathobiology, New York University, New York, NY, USA
| | - Mark von Zastrow
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, San Francisco, CA, USA
| | - Sivaraj Sivaramakrishnan
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, MN, USA.
| |
Collapse
|
16
|
Gerken OJ, Catone N, Legler DF. Identification of critical residues at the C-terminal tip of ACKR4 regulating chemokine internalization and βarrestin involvement. Cell Commun Signal 2024; 22:576. [PMID: 39623381 PMCID: PMC11610291 DOI: 10.1186/s12964-024-01961-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 11/27/2024] [Indexed: 12/06/2024] Open
Abstract
BACKGROUND Atypical chemokine receptors (ACKRs) play an important role in regulating the availability of chemokines and are responsible for the formation of chemokine gradients required for the directed migration of immune cells in health and disease. ACKR4 shapes gradients of the chemokines CCL19 and CCL21, which are essential for guiding leukocyte homing to lymphoid organs where they initiate an adaptive immune response against invading pathogens. How ACKRs internalize and scavenge chemokines on the molecular level remains poorly understood. Current state-of the art methods to study βarrestin recruitment, signaling and trafficking of ACKRs - and G-protein-coupled receptors in general - rely heavily on C-terminally tagged receptors with unknown consequences for receptor functions. METHODS Fluorescently labelled CCL19 was used to quantify chemokine internalization by native and tagged receptors as assessed by flow cytometry and live cell confocal microscopy. Steady-state interaction and chemokine-driven recruitment of βarrestins was determined by NanoBiT bystander assays. βarrestin-dependency for CCL19 internalization was determined in wild-type versus βarrestin1/2-double deficient cell lines. Statistical significance was determined by unpaired t-test or one-way ANOVA with Dunnett's or Tukey's multiple comparison tests. RESULTS Addition of a C-terminal tag selectively affected the function of ACKR4, but not other ACKRs. Fusing a short peptide tag or a fluorescent protein to ACKR4 significantly augmented its ability to internalize its cognate ligand CCL19. In comparison to native ACKR4, its C-terminal tagging provoked an elevated pre-association of βarrestins with the plasma membrane, yet a reduction in chemokine-driven βarrestin recruitment. Furthermore, the addition of a C-terminal tag led to a shift from a βarrestin-dependent towards a βarrestin-independent endocytosis pathway. Similar results on chemokine uptake and on βarrestin-dependency were obtained with ACKR4 variants, in which a putative class II PDZ-binding domain located at the C-terminal tip of the receptor was mutated. CONCLUSION This study identifies that the integrity of the C-terminus of ACKR4 is critical for receptor function. The addition of a C-terminal tag to ACKR4 enhances chemokine uptake and alters the involvement of βarrestins in receptor trafficking.
Collapse
Affiliation(s)
- Oliver J Gerken
- Institute of Cell Biology and Immunology Thurgau (BITG) at the University of Konstanz, Kreuzlingen, CH-8280, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, CH-3012, Switzerland
| | - Nicola Catone
- Institute of Cell Biology and Immunology Thurgau (BITG) at the University of Konstanz, Kreuzlingen, CH-8280, Switzerland
| | - Daniel F Legler
- Institute of Cell Biology and Immunology Thurgau (BITG) at the University of Konstanz, Kreuzlingen, CH-8280, Switzerland.
- Theodor Kocher Institute, University of Bern, Bern, CH-3012, Switzerland.
- Faculty of Biology, University of Konstanz, D-78464, Konstanz, Germany.
| |
Collapse
|
17
|
He Y, Li R, Yu Y, Xu Z, Gao J, Wang C, Huang C, Qi Z. HucMSC extracellular vesicles increasing SATB 1 to activate the Wnt/β-catenin pathway in 6-OHDA-induced Parkinson's disease model. IUBMB Life 2024; 76:1154-1174. [PMID: 39082886 DOI: 10.1002/iub.2893] [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: 02/12/2024] [Accepted: 05/20/2024] [Indexed: 11/22/2024]
Abstract
Parkinson's disease (PD) is a degenerative disorder of the nervous system characterized by the loss of dopaminergic neurons and damage of neurons in the substantia nigra (SN) and striatum, resulting in impaired motor functions. This study aims to investigate how extracellular vesicles (EVs) derived from human umbilical cord mesenchymal stem cells (HucMSC) regulate Special AT-rich sequence-binding protein-1 (SATB 1) and influence Wnt/β-catenin pathway and autophagy in PD model. The PD model was induced by damaging SH-SY5Y cells and mice using 6-OHDA. According to the study, administering EVs every other day for 14 days improved the motor behavior of 6-OHDA-induced PD mice and reduced neuronal damage, including dopaminergic neurons. Treatment with EVs for 12 hours increased the viability of 6-OHDA-induced SH-SY5Y cells. The upregulation of SATB 1 expression with EV treatment resulted in the activation of the Wnt/β-catenin pathway in PD model and led to overexpression of β-catenin. Meanwhile, the expression of LC3 II was decreased, indicating alterations in autophagy. In conclusion, EVs could mitigate neuronal damage in the 6-OHDA-induced PD model by upregulating SATB 1 and activating Wnt/β-catenin pathway while also regulating autophagy. Further studies on the potential therapeutic applications of EVs for PD could offer new insights and strategies.
Collapse
Affiliation(s)
- Ying He
- Medical College, Guangxi University, Nanning, China
- Department of Pharmacy, The Fourth Affiliated Hospital of Guangxi Medical University, Liuzhou, China
| | - Ruicheng Li
- Medical College, Guangxi University, Nanning, China
| | - Yuxi Yu
- Medical College, Guangxi University, Nanning, China
| | - Zhiran Xu
- Translational Medicine Research Center, Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, Nanning, China
| | - Jiaxin Gao
- Medical College, Guangxi University, Nanning, China
| | - Cancan Wang
- Medical College, Guangxi University, Nanning, China
| | - Chusheng Huang
- Department of Thoracic and Cardiovascular Surgery, The Second Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Zhongquan Qi
- Medical College, Guangxi University, Nanning, China
| |
Collapse
|
18
|
Lyu J, MacDonald ML, Ruiz S, Chou S, Gilardi J, Buchwald SC, Grubisha MJ, Sweet RA. Deciphering the alteration of MAP2 interactome caused by a schizophrenia-associated phosphorylation. Neurobiol Dis 2024; 203:106731. [PMID: 39532265 DOI: 10.1016/j.nbd.2024.106731] [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: 03/11/2024] [Revised: 11/06/2024] [Accepted: 11/06/2024] [Indexed: 11/16/2024] Open
Abstract
Microtubule-associated protein 2 (MAP2) is a crucial regulator of dendritic structure and neuronal function, orchestrating diverse protein interactions within the microtubule network. We have shown MAP2 is hyperphosphorylated at serine 1782 (S1782) in schizophrenia and phosphomimetic mutation of S1782 in mice (MAP2S1782E) is sufficient to impair dendritic architecture. We sought to determine how this hyperphosphorylation affects the MAP2 interactome to provide insights into the disorder's mechanisms. We investigated the MAP2 interactome using co-immunoprecipitation and mass spectrometry in MAP2S1782E and MAP2WT mice. We found that S1782E MAP2 led to a substantial disruption of protein-protein interactions relative to WT MAP2. Reduced interactions with PDZ domain-containing proteins, calmodulin-binding proteins, ribosome proteins, and kinesin proteins may all contribute to dendritic impairments induced by S1782E, and may be linked to schizophrenia pathogenesis. Interestingly, novel gain-of-function interactions with PPM1L and KLHL8 nominated these as regulators of phosphoS1782 MAP2 abundance and potential therapeutic targets in schizophrenia.
Collapse
Affiliation(s)
- Jiali Lyu
- School of Medicine, Tsinghua University, China; Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh, United States of America
| | - Matthew L MacDonald
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh, United States of America; Health Sciences Mass Spectrometry Core, University of Pittsburgh, United States of America
| | - Shelby Ruiz
- Department of Neurobiology, University of Pittsburgh, United States of America
| | - Shinnyi Chou
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh, United States of America
| | - Jordan Gilardi
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh, United States of America
| | - Serena C Buchwald
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh, United States of America
| | - Melanie J Grubisha
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh, United States of America
| | - Robert A Sweet
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh, United States of America; Department of Neurology, University of Pittsburgh, United States of America.
| |
Collapse
|
19
|
Felício D, Martins S, Alves GP, Amorim A, Macedo‐Ribeiro S, Merski M. Evolutionary model of repeat insertions in Ataxin-3 traces the origin of the polyglutamine stretch to an ancestral ubiquitin binding module. Protein Sci 2024; 33:e5236. [PMID: 39589068 PMCID: PMC11590126 DOI: 10.1002/pro.5236] [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: 08/05/2024] [Revised: 11/08/2024] [Accepted: 11/09/2024] [Indexed: 11/27/2024]
Abstract
The human ataxin-3 protein contains an N-terminal Josephin domain, composed of a papain-like cysteine protease with a helical hairpin insertion, and a C-terminal region with two or three ubiquitin interacting motifs and a polyglutamine tract. Expansion of the polyglutamine tract leading to protein aggregation and neuronal degradation has been linked to Machado-Joseph disease/spinocerebellar ataxia type 3, the most common form of dominantly inherited ataxia. In this study, we performed sequence self-homology dot plot analysis and compared orthologous proteins to analyze the architecture of ataxin-3 during the evolution of Filozoa. This analysis uncovered up to three additional repetitions of the ubiquitin binding motif in ataxin-3, including the helical hairpin insertion in the Josephin domain, and revealed a highly conserved multimodular architecture that is broadly preserved throughout the Filozoa. Overall, a set of 78 putative ubiquitin binding repeats from 18 exemplar proteins were identified. Apparent neofunctionalization events could also be recognized, including modification of repeat 5 which gave rise to the disease-linked polyglutamine tract, just before the Sarcopterygian divergence. This model provides a unifying principle for the ataxin-3 protein architecture and can potentially provide new insights into the role of molecular interactions in ataxin-3 function and Machado-Joseph disease/spinocerebellar ataxia type 3 disease mechanisms.
Collapse
Affiliation(s)
- Daniela Felício
- i3S‐Instituto de Investigação e Inovação em SaúdeUniversidade do PortoPortoPortugal
- IPATIMUP‐Institute of Molecular Pathology and Immunology of the University of PortoPortoPortugal
- ICBAS‐Instituto Ciências Biomédicas Abel SalazarUniversity of PortoPortoPortugal
| | - Sandra Martins
- i3S‐Instituto de Investigação e Inovação em SaúdeUniversidade do PortoPortoPortugal
- IPATIMUP‐Institute of Molecular Pathology and Immunology of the University of PortoPortoPortugal
| | | | - António Amorim
- i3S‐Instituto de Investigação e Inovação em SaúdeUniversidade do PortoPortoPortugal
- IPATIMUP‐Institute of Molecular Pathology and Immunology of the University of PortoPortoPortugal
- Faculty of SciencesUniversity of PortoPortoPortugal
| | - Sandra Macedo‐Ribeiro
- i3S‐Instituto de Investigação e Inovação em SaúdeUniversidade do PortoPortoPortugal
- IBMC‐Instituto de Biologia Molecular e CelularUniversidade do PortoPortoPortugal
| | - Matthew Merski
- i3S‐Instituto de Investigação e Inovação em SaúdeUniversidade do PortoPortoPortugal
- IBMC‐Instituto de Biologia Molecular e CelularUniversidade do PortoPortoPortugal
| |
Collapse
|
20
|
Chinchilla P, Wang B, Lubin JH, Yang X, Roth J, Khare SD, Baum J. Synergistic Multi-Pronged Interactions Mediate the Effective Inhibition of Alpha-Synuclein Aggregation by the Chaperone HtrA1. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.25.624572. [PMID: 39651184 PMCID: PMC11623516 DOI: 10.1101/2024.11.25.624572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2024]
Abstract
The misfolding, aggregation, and the seeded spread of alpha synuclein (α-Syn) aggregates are linked to the pathogenesis of various neurodegenerative diseases, including Parkinson's disease (PD). Understanding the mechanisms by which chaperone proteins prevent the production and seeding of α-Syn aggregates is crucial for developing effective therapeutic leads for tackling neurodegenerative diseases. We show that a catalytically inactive variant of the chaperone HtrA1 (HtrA1*) effectively inhibits both α-Syn monomer aggregation and templated fibril seeding, and demonstrate that this inhibition is mediated by synergistic interactions between its PDZ and Protease domains and α-Syn. Using biomolecular NMR, AFM and Rosetta-based computational analyses, we propose that the PDZ domain interacts with the C-terminal end of the monomer and the intrinsically disordered C-terminal domain of the α-Syn fibril. Furthermore, in agreement with sequence specificity calculations, the Protease domain cleaves in the aggregation-prone NAC domain at site T92/A93 in the monomer. Thus, through multi-pronged interactions and multi-site recognition of α-Syn, HtrA1* can effectively intervene at different stages along the α-Syn aggregation pathway, making it a robust inhibitor of α-Syn aggregation and templated seeding. Our studies illustrate, at high resolution, the crucial role of HtrA1 interactions with both the intrinsically disordered α-Syn monomers and with the dynamic flanking regions around the fibril core for inhibition of aggregation. This inhibition mechanism of the HtrA1 chaperone may provide a natural mechanistic blueprint for highly effective therapeutic agents against protein aggregation. Significance Statement PD and other synucleinopathies are marked by misfolding and aggregation of α-Syn, forming higher-order species that propagate aggregation in a prion-like manner. Understanding how chaperone proteins inhibit α-Syn aggregation and spread is essential for therapeutic development against neurodegeneration. Through an integrative approach of solution-based NMR, AFM, aggregation kinetics, and computational analysis, we reveal how a catalytically inactive variant of the chaperone HtrA1 effectively disrupts aggregation pathways. We find that the inactive Protease and PDZ domains of HtrA1 synergistically bind to key intrinsically disordered sites on both α-Syn monomers and fibrils, thereby effectively inhibiting both aggregation and templated seeding. Our work provides a natural and unique blueprint for designing inhibitors to prevent the formation and seeding of aggregates in neurodegenerative diseases.
Collapse
|
21
|
Chartier CA, Woods VA, Xu Y, van Vlimmeren AE, Johns AC, Jovanovic M, McDermott AE, Keedy DA, Shah NH. Allosteric regulation of the tyrosine phosphatase PTP1B by a protein-protein interaction. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.16.603632. [PMID: 39071364 PMCID: PMC11275736 DOI: 10.1101/2024.07.16.603632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
The rapid identification of protein-protein interactions has been significantly enabled by mass spectrometry (MS) proteomics-based methods, including affinity purification-MS, crosslinking-MS, and proximity-labeling proteomics. While these methods can reveal networks of interacting proteins, they cannot reveal how specific protein-protein interactions alter protein function or cell signaling. For instance, when two proteins interact, there can be emergent signaling processes driven purely by the individual activities of those proteins being co-localized. Alternatively, protein-protein interactions can allosterically regulate function, enhancing or suppressing activity in response to binding. In this work, we investigate the interaction between the tyrosine phosphatase PTP1B and the adaptor protein Grb2, which have been annotated as binding partners in a number of proteomics studies. This interaction has been postulated to co-localize PTP1B with its substrate IRS-1 by forming a ternary complex, thereby enhancing the dephosphorylation of IRS-1 to suppress insulin signaling. Here, we report that Grb2 binding to PTP1B also allosterically enhances PTP1B catalytic activity. We show that this interaction is dependent on the proline-rich region of PTP1B, which interacts with the C-terminal SH3 domain of Grb2. Using NMR spectroscopy and hydrogen-deuterium exchange mass spectrometry (HDX-MS) we show that Grb2 binding alters PTP1B structure and/or dynamics. Finally, we use MS proteomics to identify other interactors of the PTP1B proline-rich region that may also regulate PTP1B function similarly to Grb2. This work presents one of the first examples of a protein allosterically regulating the enzymatic activity of PTP1B and lays the foundation for discovering new mechanisms of PTP1B regulation in cell signaling.
Collapse
Affiliation(s)
| | - Virgil A. Woods
- Structural Biology Initiative, CUNY Advanced Science Research Center, New York, NY 10031
- PhD Program in Biochemistry, CUNY Graduate Center, New York, NY 10016
| | - Yunyao Xu
- Department of Chemistry, Columbia University, New York, NY 10027
| | - Anne E. van Vlimmeren
- Department of Chemistry, Columbia University, New York, NY 10027
- Department of Biological Sciences, Columbia University, New York, NY 10027
| | - Andrew C. Johns
- Department of Chemistry, Columbia University, New York, NY 10027
| | - Marko Jovanovic
- Department of Biological Sciences, Columbia University, New York, NY 10027
| | - Ann E. McDermott
- Department of Chemistry, Columbia University, New York, NY 10027
| | - Daniel A. Keedy
- Structural Biology Initiative, CUNY Advanced Science Research Center, New York, NY 10031
- Department of Chemistry and Biochemistry, City College of New York, New York, NY 10031
- PhD Programs in Biochemistry, Biology, & Chemistry, CUNY Graduate Center, New York, NY 10016
| | - Neel H. Shah
- Department of Chemistry, Columbia University, New York, NY 10027
| |
Collapse
|
22
|
Wang L, Zhu R, Wen Z, Fan HJS, Norwood-Jackson T, Jathan D, Lee HJ. Structural and Functional Insights into Dishevelled-Mediated Wnt Signaling. Cells 2024; 13:1870. [PMID: 39594618 PMCID: PMC11592973 DOI: 10.3390/cells13221870] [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: 10/14/2024] [Revised: 11/01/2024] [Accepted: 11/06/2024] [Indexed: 11/28/2024] Open
Abstract
Dishevelled (DVL) proteins precisely control Wnt signaling pathways with many effectors. While substantial research has advanced our understanding of DVL's role in Wnt pathways, key questions regarding its regulatory mechanisms and interactions remain unresolved. Herein, we present the recent advances and perspectives on how DVL regulates signaling. The experimentally determined conserved domain structures of DVL in conjunction with AlphaFold-predicted structures are used to understand the DVL's role in Wnt signaling regulation. We also summarize the role of DVL in various diseases and provide insights into further directions for research on the DVL-mediated signaling mechanisms. These findings underscore the importance of DVL as a pharmaceutical target or biological marker in diseases, offering exciting potential for future biomedical applications.
Collapse
Affiliation(s)
- Lei Wang
- College of Chemical Engineering, Sichuan University of Science and Engineering, Zigong 643000, China; (L.W.); (R.Z.); (Z.W.); (H.-J.S.F.)
| | - Rui Zhu
- College of Chemical Engineering, Sichuan University of Science and Engineering, Zigong 643000, China; (L.W.); (R.Z.); (Z.W.); (H.-J.S.F.)
| | - Zehua Wen
- College of Chemical Engineering, Sichuan University of Science and Engineering, Zigong 643000, China; (L.W.); (R.Z.); (Z.W.); (H.-J.S.F.)
| | - Hua-Jun Shawn Fan
- College of Chemical Engineering, Sichuan University of Science and Engineering, Zigong 643000, China; (L.W.); (R.Z.); (Z.W.); (H.-J.S.F.)
| | - Teresa Norwood-Jackson
- Division of Natural & Mathematical Sciences, LeMoyne-Owen College, Memphis, TN 38126, USA; (T.N.-J.); (D.J.)
| | - Danielle Jathan
- Division of Natural & Mathematical Sciences, LeMoyne-Owen College, Memphis, TN 38126, USA; (T.N.-J.); (D.J.)
| | - Ho-Jin Lee
- Division of Natural & Mathematical Sciences, LeMoyne-Owen College, Memphis, TN 38126, USA; (T.N.-J.); (D.J.)
| |
Collapse
|
23
|
Agogo-Mawuli PS, Mendez J, Oestreich EA, Bosch DE, Siderovski DP. Molecular Modeling and In Vitro Functional Analysis of the RGS12 PDZ Domain Variant Associated with High-Penetrance Familial Bipolar Disorder. Int J Mol Sci 2024; 25:11431. [PMID: 39518985 PMCID: PMC11546610 DOI: 10.3390/ijms252111431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 10/17/2024] [Accepted: 10/21/2024] [Indexed: 11/16/2024] Open
Abstract
Bipolar disorder's etiology involves genetics, environmental factors, and gene-environment interactions, underlying its heterogeneous nature and treatment complexity. In 2020, Forstner and colleagues catalogued 378 sequence variants co-segregating with familial bipolar disorder. A notable candidate was an R59Q missense mutation in the PDZ (PSD-95/Dlg1/ZO-1) domain of RGS12. We previously demonstrated that RGS12 loss removes negative regulation on the kappa opioid receptor, disrupting basal ganglia dopamine homeostasis and dampening responses to dopamine-eliciting psychostimulants. Here, we investigated the R59Q variation in the context of potential PDZ domain functional alterations. We first validated a new target for the wildtype RGS12 PDZ domain-the SAPAP3 C-terminus-by molecular docking, surface plasmon resonance (SPR), and co-immunoprecipitation. While initial molecular dynamics (MD) studies predicted negligible effects of the R59Q variation on ligand binding, SPR showed a significant reduction in binding affinity for the three peptide targets tested. AlphaFold2-generated models predicted a modest reduction in protein-peptide interactions, which is consistent with the reduced binding affinity observed by SPR, suggesting that the substituted glutamine side chain may weaken the affinity of RGS12 for its in vivo binding targets, likely through allosteric changes. This difference may adversely affect the CNS signaling related to dynorphin and dopamine in individuals with this R59Q variation, potentially impacting bipolar disorder pathophysiology.
Collapse
Affiliation(s)
- Percy S. Agogo-Mawuli
- Department of Pharmacology & Neuroscience, University of North Texas Health Science Center, Fort Worth, TX 76107, USA; (P.S.A.-M.)
| | - Joseph Mendez
- Department of Pharmacology & Neuroscience, University of North Texas Health Science Center, Fort Worth, TX 76107, USA; (P.S.A.-M.)
| | - Emily A. Oestreich
- Department of Biomedical Sciences, Pacific Northwest University of Health Sciences, Yakima, WA 98901, USA
| | - Dustin E. Bosch
- Department of Pathology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - David P. Siderovski
- Department of Pharmacology & Neuroscience, University of North Texas Health Science Center, Fort Worth, TX 76107, USA; (P.S.A.-M.)
| |
Collapse
|
24
|
Drobnič T, Cohen EJ, Calcraft T, Alzheimer M, Froschauer K, Svensson S, Hoffmann WH, Singh N, Garg SG, Henderson L, Umrekar TR, Nans A, Ribardo D, Pedaci F, Nord AL, Hochberg GKA, Hendrixson DR, Sharma CM, Rosenthal PB, Beeby M. Molecular model of a bacterial flagellar motor in situ reveals a "parts-list" of protein adaptations to increase torque. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.09.08.556779. [PMID: 39416179 PMCID: PMC11482838 DOI: 10.1101/2023.09.08.556779] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
One hurdle to understanding how molecular machines work, and how they evolve, is our inability to see their structures in situ. Here we describe a minicell system that enables in situ cryogenic electron microscopy imaging and single particle analysis to investigate the structure of an iconic molecular machine, the bacterial flagellar motor, which spins a helical propeller for propulsion. We determine the structure of the high-torque Campylobacter jejuni motor in situ, including the subnanometre-resolution structure of the periplasmic scaffold, an adaptation essential to high torque. Our structure enables identification of new proteins, and interpretation with molecular models highlights origins of new components, reveals modifications of the conserved motor core, and explain how these structures both template a wider ring of motor proteins, and buttress the motor during swimming reversals. We also acquire insights into universal principles of flagellar torque generation. This approach is broadly applicable to other membrane-residing bacterial molecular machines complexes.
Collapse
Affiliation(s)
- Tina Drobnič
- Department of Life Sciences, Imperial College London, London, SW7 2AZ, UK
- Tina Drobnič current affiliation: MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge CB2 0QH, UK
| | - Eli J. Cohen
- Department of Life Sciences, Imperial College London, London, SW7 2AZ, UK
| | - Tom Calcraft
- Structural Biology of Cells and Viruses Laboratory, The Francis Crick Institute, London NW1 1AT, UK
| | - Mona Alzheimer
- University of Würzburg, Institute of Molecular Infection Biology, Department of Molecular Infection Biology II, Josef-Schneider-Straße 2/D15, 97080 Würzburg, Germany
| | - Kathrin Froschauer
- University of Würzburg, Institute of Molecular Infection Biology, Department of Molecular Infection Biology II, Josef-Schneider-Straße 2/D15, 97080 Würzburg, Germany
| | - Sarah Svensson
- University of Würzburg, Institute of Molecular Infection Biology, Department of Molecular Infection Biology II, Josef-Schneider-Straße 2/D15, 97080 Würzburg, Germany
- Current affiliation: The Center for Microbes, Development and Health, CAS Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China 200031.
| | - William H. Hoffmann
- Centre de Biologie Structurale, Universite de Montpellier, CNRS, INSERM. Montpellier, France
| | - Nanki Singh
- Department of Life Sciences, Imperial College London, London, SW7 2AZ, UK
| | - Sriram G. Garg
- Max Planck Institute for Terrestrial Microbiology, Marburg, Germany
| | - Louie Henderson
- Department of Life Sciences, Imperial College London, London, SW7 2AZ, UK
- Current affiliation: Peptone Ltd, 370 Grays Inn Road, London WC1X 8BB, UK
| | | | - Andrea Nans
- Structural Biology Science Technology Platform, The Francis Crick Institute, London NW1 1AT, UK
| | - Deborah Ribardo
- Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Francesco Pedaci
- Centre de Biologie Structurale, Universite de Montpellier, CNRS, INSERM. Montpellier, France
| | - Ashley L. Nord
- Centre de Biologie Structurale, Universite de Montpellier, CNRS, INSERM. Montpellier, France
| | | | - David R. Hendrixson
- Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Cynthia M. Sharma
- University of Würzburg, Institute of Molecular Infection Biology, Department of Molecular Infection Biology II, Josef-Schneider-Straße 2/D15, 97080 Würzburg, Germany
| | - Peter B. Rosenthal
- Structural Biology of Cells and Viruses Laboratory, The Francis Crick Institute, London NW1 1AT, UK
| | - Morgan Beeby
- Department of Life Sciences, Imperial College London, London, SW7 2AZ, UK
| |
Collapse
|
25
|
Lee HC, Park SH, Jeong HM, Shin G, Lim SI, Kim J, Shim J, Park YM, Song KS. LPS-induced systemic inflammation is suppressed by the PDZ motif peptide of ZO-1 via regulation of macrophage M1/M2 polarization. eLife 2024; 13:RP95285. [PMID: 39377568 PMCID: PMC11460976 DOI: 10.7554/elife.95285] [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] [Indexed: 10/09/2024] Open
Abstract
The gram-negative bacterium lipopolysaccharide (LPS) is frequently administered to generate models of systemic inflammation. However, there are several side effects and no effective treatment for LPS-induced systemic inflammation. PEGylated PDZ peptide based on zonula occludens-1 (ZO-1) was analyzed for its effects on systemic inflammation induced by LPS. PDZ peptide administration led to the restoration of tissue injuries (kidney, liver, and lung) and prevented alterations in biochemical plasma markers. The production of pro-inflammatory cytokines was significantly decreased in the plasma and lung BALF in the PDZ-administered mice. Flow cytometry analysis revealed the PDZ peptide significantly inhibited inflammation, mainly by decreasing the population of M1 macrophages, and neutrophils (immature and mature), and increasing M2 macrophages. Using RNA sequencing analysis, the expression levels of the NF-κB-related proteins were lower in PDZ-treated cells than in LPS-treated cells. In addition, wild-type PDZ peptide significantly increased mitochondrial membrane integrity and decreased LPS-induced mitochondria fission. Interestingly, PDZ peptide dramatically could reduce LPS-induced NF-κB signaling, ROS production, and the expression of M1 macrophage marker proteins, but increased the expression of M2 macrophage marker proteins. These results indicated that PEGylated PDZ peptide inhibits LPS-induced systemic inflammation, reducing tissue injuries and reestablishing homeostasis, and may be a therapeutic candidate against systemic inflammation.
Collapse
Affiliation(s)
- Hyun-Chae Lee
- Department of Medical Science, Kosin University College of MedicineBusanRepublic of Korea
| | - Sun-Hee Park
- Department of Medical Science, Kosin University College of MedicineBusanRepublic of Korea
| | - Hye Min Jeong
- Department of Medical Science, Kosin University College of MedicineBusanRepublic of Korea
| | - Goeun Shin
- Department of Chemical Engineering, Pukyong National UniversityBusanRepublic of Korea
| | - Sung In Lim
- Department of Chemical Engineering, Pukyong National UniversityBusanRepublic of Korea
| | - Jeongtae Kim
- Department of Anatomy, Kosin University College of MedicineBusanRepublic of Korea
| | - Jaewon Shim
- Department of Biochemistry, Kosin University College of MedicineBusanRepublic of Korea
| | - Yeong-Min Park
- Department of Integrative Biological Sciences and Industry, College of Life Sciences, Sejong UniversitySeoulRepublic of Korea
| | - Kyoung Seob Song
- Department of Medical Science, Kosin University College of MedicineBusanRepublic of Korea
| |
Collapse
|
26
|
do Nascimento AM, Marques RB, Roldão AP, Rodrigues AM, Eslava RM, Dale CS, Reis EM, Schechtman D. Exploring protein-protein interactions for the development of new analgesics. Sci Signal 2024; 17:eadn4694. [PMID: 39378285 DOI: 10.1126/scisignal.adn4694] [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: 12/12/2023] [Revised: 06/13/2024] [Accepted: 09/16/2024] [Indexed: 10/10/2024]
Abstract
The development of new analgesics has been challenging. Candidate drugs often have limited clinical utility due to side effects that arise because many drug targets are involved in signaling pathways other than pain transduction. Here, we explored the potential of targeting protein-protein interactions (PPIs) that mediate pain signaling as an approach to developing drugs to treat chronic pain. We reviewed the approaches used to identify small molecules and peptide modulators of PPIs and their ability to decrease pain-like behaviors in rodent animal models. We analyzed data from rodent and human sensory nerve tissues to build associated signaling networks and assessed both validated and potential interactions and the structures of the interacting domains that could inform the design of synthetic peptides and small molecules. This resource identifies PPIs that could be explored for the development of new analgesics, particularly between scaffolding proteins and receptors for various growth factors and neurotransmitters, as well as ion channels and other enzymes. Targeting the adaptor function of CBL by blocking interactions between its proline-rich carboxyl-terminal domain and its SH3-domain-containing protein partners, such as GRB2, could disrupt endosomal signaling induced by pain-associated growth factors. This approach would leave intact its E3-ligase functions, which are mediated by other domains and are critical for other cellular functions. This potential of PPI modulators to be more selective may mitigate side effects and improve the clinical management of pain.
Collapse
Affiliation(s)
- Alexandre Martins do Nascimento
- Department of Biochemistry, Chemistry Institute, University of São Paulo, SP 05508-000, Brazil
- Laboratory of Neuromodulation of Experimental Pain (LaNed), Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo, SP 05508-000, Brazil
| | - Rauni Borges Marques
- Department of Biochemistry, Chemistry Institute, University of São Paulo, SP 05508-000, Brazil
- Interunit Graduate Program in Bioinformatics, University of São Paulo, SP 05508-000, Brazil
| | - Allan Pradelli Roldão
- Department of Biochemistry, Chemistry Institute, University of São Paulo, SP 05508-000, Brazil
| | - Ana Maria Rodrigues
- Department of Biochemistry, Chemistry Institute, University of São Paulo, SP 05508-000, Brazil
| | - Rodrigo Mendes Eslava
- Department of Biochemistry, Chemistry Institute, University of São Paulo, SP 05508-000, Brazil
| | - Camila Squarzoni Dale
- Laboratory of Neuromodulation of Experimental Pain (LaNed), Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo, SP 05508-000, Brazil
| | - Eduardo Moraes Reis
- Department of Biochemistry, Chemistry Institute, University of São Paulo, SP 05508-000, Brazil
| | - Deborah Schechtman
- Department of Biochemistry, Chemistry Institute, University of São Paulo, SP 05508-000, Brazil
| |
Collapse
|
27
|
Soto DC, Uribe-Salazar JM, Kaya G, Valdarrago R, Sekar A, Haghani NK, Hino K, La GN, Mariano NAF, Ingamells C, Baraban AE, Turner TN, Green ED, Simó S, Quon G, Andrés AM, Dennis MY. Gene expansions contributing to human brain evolution. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.26.615256. [PMID: 39386494 PMCID: PMC11463660 DOI: 10.1101/2024.09.26.615256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Genomic drivers of human-specific neurological traits remain largely undiscovered. Duplicated genes expanded uniquely in the human lineage likely contributed to brain evolution, including the increased complexity of synaptic connections between neurons and the dramatic expansion of the neocortex. Discovering duplicate genes is challenging because the similarity of paralogs makes them prone to sequence-assembly errors. To mitigate this issue, we analyzed a complete telomere-to-telomere human genome sequence (T2T-CHM13) and identified 213 duplicated gene families likely containing human-specific paralogs (>98% identity). Positing that genes important in universal human brain features should exist with at least one copy in all modern humans and exhibit expression in the brain, we narrowed in on 362 paralogs with at least one copy across thousands of ancestrally diverse genomes and present in human brain transcriptomes. Of these, 38 paralogs co-express in gene modules enriched for autism-associated genes and potentially contribute to human language and cognition. We narrowed in on 13 duplicate gene families with human-specific paralogs that are fixed among modern humans and show convincing brain expression patterns. Using long-read DNA sequencing revealed hidden variation across 200 modern humans of diverse ancestries, uncovering signatures of selection not previously identified, including possible balancing selection of CD8B. To understand the roles of duplicated genes in brain development, we generated zebrafish CRISPR "knockout" models of nine orthologs and transiently introduced mRNA-encoding paralogs, effectively "humanizing" the larvae. Morphometric, behavioral, and single-cell RNA-seq screening highlighted, for the first time, a possible role for GPR89B in dosage-mediated brain expansion and FRMPD2B function in altered synaptic signaling, both hallmark features of the human brain. Our holistic approach provides important insights into human brain evolution as well as a resource to the community for studying additional gene expansion drivers of human brain evolution.
Collapse
Affiliation(s)
- Daniela C. Soto
- Department of Biochemistry & Molecular Medicine, MIND Institute, University of California,Davis, CA 95616, USA
- Genome Center, University of California, Davis, CA 95616, USA
| | - José M. Uribe-Salazar
- Department of Biochemistry & Molecular Medicine, MIND Institute, University of California,Davis, CA 95616, USA
- Genome Center, University of California, Davis, CA 95616, USA
| | - Gulhan Kaya
- Department of Biochemistry & Molecular Medicine, MIND Institute, University of California,Davis, CA 95616, USA
- Genome Center, University of California, Davis, CA 95616, USA
| | - Ricardo Valdarrago
- Department of Molecular and Cellular Biology, University of California, Davis, CA 95616, USA
| | - Aarthi Sekar
- Department of Biochemistry & Molecular Medicine, MIND Institute, University of California,Davis, CA 95616, USA
- Genome Center, University of California, Davis, CA 95616, USA
| | - Nicholas K. Haghani
- Department of Biochemistry & Molecular Medicine, MIND Institute, University of California,Davis, CA 95616, USA
- Genome Center, University of California, Davis, CA 95616, USA
| | - Keiko Hino
- Department of Cell Biology & Human Anatomy, University of California, Davis, CA 95616, USA
| | - Gabriana N. La
- Department of Biochemistry & Molecular Medicine, MIND Institute, University of California,Davis, CA 95616, USA
- Genome Center, University of California, Davis, CA 95616, USA
| | - Natasha Ann F. Mariano
- Department of Biochemistry & Molecular Medicine, MIND Institute, University of California,Davis, CA 95616, USA
- Genome Center, University of California, Davis, CA 95616, USA
- Postbaccalaureate Research Education Program, University of California, Davis, CA 95616, USA
| | - Cole Ingamells
- Department of Biochemistry & Molecular Medicine, MIND Institute, University of California,Davis, CA 95616, USA
- Genome Center, University of California, Davis, CA 95616, USA
| | - Aidan E. Baraban
- Department of Biochemistry & Molecular Medicine, MIND Institute, University of California,Davis, CA 95616, USA
- Genome Center, University of California, Davis, CA 95616, USA
| | - Tychele N. Turner
- Department of Genetics, Washington University School of Medicine, St Louis, MS, 63110, USA
| | - Eric D. Green
- National Human Genome Research Institute, National Institutes of Health, Bethesda, MD,20892, USA
| | - Sergi Simó
- Department of Cell Biology & Human Anatomy, University of California, Davis, CA 95616, USA
| | - Gerald Quon
- Genome Center, University of California, Davis, CA 95616, USA
- Department of Molecular and Cellular Biology, University of California, Davis, CA 95616, USA
| | - Aida M. Andrés
- UCL Genetics Institute, Department of Genetics, Evolution and Environment, University College, London, WC1E 6BT, UK
| | - Megan Y. Dennis
- Department of Biochemistry & Molecular Medicine, MIND Institute, University of California,Davis, CA 95616, USA
- Genome Center, University of California, Davis, CA 95616, USA
| |
Collapse
|
28
|
Sarkar S, Dhibar S, Jana B. Modulation of the conformational landscape of the PDZ3 domain by perturbation on a distal non-canonical α3 helix: decoding the microscopic mechanism of allostery in the PDZ3 domain. Phys Chem Chem Phys 2024; 26:21249-21259. [PMID: 39076021 DOI: 10.1039/d4cp01806k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/31/2024]
Abstract
While allosteric signal transduction is crucial for protein signaling and regulation, the dynamic process of allosteric communication remains poorly understood. The third PDZ domain (PDZ stands for the common structural domain shared by the postsynaptic density protein (PSD95), Drosophila disc large tumor suppressor (DlgA), and zonula occludens-1 protein (ZO-1)) serves as a classic example of a single-domain allosteric protein, demonstrating a long-range coupling between the C-terminal α helix (known as the α3 helix) and ligand binding. A molecular level understanding of how the α3 helix modulates the ligand binding affinity of the PDZ3 domain is still lacking. In this study, extensive molecular dynamics simulations corroborated with principal component analysis (PCA), ligand binding free energy calculations, energetic frustration analysis and Markov state model analysis are employed to uncover such molecular details. We demonstrate the definite presence of a binding competent closed-like state in the conformational landscape of wild-type PDZ3. The population modulations of this closed state and other binding incompetent states in the landscape due to α3-truncation/mutation of PDZ3 are explored. A correlation between the closed state population and calculated binding free energy is established, which supports the conformation selection mechanism. Covariance analysis identified the presence of correlated motion between two distant loops (β1-β2 and β2-β3) in the wild-type PDZ3 system, which weakened due to truncation/mutation in the distant α3 helix. It has also been observed that whenever the α3 helix was perturbed, the β2-β3 loop got further away from the binding groove and it is found to be correlated with the binding free energy values. Energetic frustration analysis of the PDZ3 domain also showed that the β2-β3 loop is highly frustrated. Finally, MSM analysis revealed a relevant timescale (closed to open state transition), which is similar to the observed experimental signal transduction timescale for the system. These observations led to the conclusion that the distantly located α3 helix plays a pivotal role in regulating the conformational landscape of the PDZ3 domain, determining the ligand binding affinity and resulting in allosteric behavior of the domain.
Collapse
Affiliation(s)
- Subhajit Sarkar
- School of Chemical Sciences, Indian Association for the Cultivation of Science, Kolkata-700032, India.
| | - Saikat Dhibar
- School of Chemical Sciences, Indian Association for the Cultivation of Science, Kolkata-700032, India.
| | - Biman Jana
- School of Chemical Sciences, Indian Association for the Cultivation of Science, Kolkata-700032, India.
| |
Collapse
|
29
|
Honrubia JM, Valverde JR, Muñoz-Santos D, Ripoll-Gómez J, de la Blanca N, Izquierdo J, Villarejo-Torres M, Marchena-Pasero A, Rueda-Huélamo M, Nombela I, Ruiz-Yuste M, Zuñiga S, Sola I, Enjuanes L. Interaction between SARS-CoV PBM and Cellular PDZ Domains Leading to Virus Virulence. Viruses 2024; 16:1214. [PMID: 39205188 PMCID: PMC11359647 DOI: 10.3390/v16081214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 07/19/2024] [Accepted: 07/26/2024] [Indexed: 09/04/2024] Open
Abstract
The interaction between SARS-CoV PDZ-binding motifs (PBMs) and cellular PDZs is responsible for virus virulence. The PBM sequence present in the 3a and envelope (E) proteins of SARS-CoV can potentially bind to over 400 cellular proteins containing PDZ domains. The role of SARS-CoV 3a and E proteins was studied. SARS-CoVs, in which 3a-PBM and E-PMB have been deleted (3a-PBM-/E-PBM-), reduced their titer around one logarithmic unit but still were viable. In addition, the absence of the E-PBM and the replacement of 3a-PBM with that of E did not allow the rescue of SARS-CoV. E protein PBM was necessary for virulence, activating p38-MAPK through the interaction with Syntenin-1 PDZ domain. However, the presence or absence of the homologous motif in the 3a protein, which does not bind to Syntenin-1, did not affect virus pathogenicity. Mutagenesis analysis and in silico modeling were performed to study the extension of the PBM of the SARS-CoV E protein. Alanine and glycine scanning was performed revealing a pair of amino acids necessary for optimum virus replication. The binding of E protein with the PDZ2 domain of the Syntenin-1 homodimer induced conformational changes in both PDZ domains 1 and 2 of the dimer.
Collapse
Affiliation(s)
- Jose M. Honrubia
- Department of Molecular and Cell Biology, Centro Nacional de Biotecnología (CNB-CSIC), Darwin 3, Campus Universidad Autónoma de Madrid, 28049 Madrid, Spain
| | - Jose R. Valverde
- Scientific Computing Service, Centro Nacional de Biotecnología (CNB-CSIC), Darwin 3, Campus Universidad Autónoma de Madrid, 28049 Madrid, Spain
| | - Diego Muñoz-Santos
- Department of Molecular and Cell Biology, Centro Nacional de Biotecnología (CNB-CSIC), Darwin 3, Campus Universidad Autónoma de Madrid, 28049 Madrid, Spain
| | - Jorge Ripoll-Gómez
- Department of Molecular and Cell Biology, Centro Nacional de Biotecnología (CNB-CSIC), Darwin 3, Campus Universidad Autónoma de Madrid, 28049 Madrid, Spain
| | - Nuria de la Blanca
- Scientific Computing Service, Centro Nacional de Biotecnología (CNB-CSIC), Darwin 3, Campus Universidad Autónoma de Madrid, 28049 Madrid, Spain
| | - Jorge Izquierdo
- Scientific Computing Service, Centro Nacional de Biotecnología (CNB-CSIC), Darwin 3, Campus Universidad Autónoma de Madrid, 28049 Madrid, Spain
| | - Marta Villarejo-Torres
- Department of Molecular and Cell Biology, Centro Nacional de Biotecnología (CNB-CSIC), Darwin 3, Campus Universidad Autónoma de Madrid, 28049 Madrid, Spain
| | - Ana Marchena-Pasero
- Department of Molecular and Cell Biology, Centro Nacional de Biotecnología (CNB-CSIC), Darwin 3, Campus Universidad Autónoma de Madrid, 28049 Madrid, Spain
| | - María Rueda-Huélamo
- Department of Molecular and Cell Biology, Centro Nacional de Biotecnología (CNB-CSIC), Darwin 3, Campus Universidad Autónoma de Madrid, 28049 Madrid, Spain
| | - Ivan Nombela
- Department of Molecular and Cell Biology, Centro Nacional de Biotecnología (CNB-CSIC), Darwin 3, Campus Universidad Autónoma de Madrid, 28049 Madrid, Spain
| | - Mercedes Ruiz-Yuste
- Department of Molecular and Cell Biology, Centro Nacional de Biotecnología (CNB-CSIC), Darwin 3, Campus Universidad Autónoma de Madrid, 28049 Madrid, Spain
| | - Sonia Zuñiga
- Department of Molecular and Cell Biology, Centro Nacional de Biotecnología (CNB-CSIC), Darwin 3, Campus Universidad Autónoma de Madrid, 28049 Madrid, Spain
| | - Isabel Sola
- Department of Molecular and Cell Biology, Centro Nacional de Biotecnología (CNB-CSIC), Darwin 3, Campus Universidad Autónoma de Madrid, 28049 Madrid, Spain
| | - Luis Enjuanes
- Department of Molecular and Cell Biology, Centro Nacional de Biotecnología (CNB-CSIC), Darwin 3, Campus Universidad Autónoma de Madrid, 28049 Madrid, Spain
| |
Collapse
|
30
|
Li Y, Trinh CH, Acevedo-Jake A, Gimenez D, Warriner SL, Wilson AJ. Biophysical and structural analyses of the interaction between the SHANK1 PDZ domain and an internal SLiM. Biochem J 2024; 481:945-955. [PMID: 38899489 PMCID: PMC11346428 DOI: 10.1042/bcj20240126] [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: 03/21/2024] [Revised: 06/12/2024] [Accepted: 06/19/2024] [Indexed: 06/21/2024]
Abstract
The PDZ (Postsynaptic density protein-95[PSD-95]/Discs-large) domain, prevalent as a recognition module, has attracted significant attention given its ability to specifically recognize ligands with consensus motifs (also termed PDZ binding motifs [PBMs]). PBMs typically bear a C-terminal carboxylate as a recognition handle and have been extensively characterized, whilst internal ligands are less well known. Here we characterize a short linear motif (SLiM) - EESTSFQGP - as an internal PBM based on its strong binding affinity towards the SHANK1 PDZ domain (SHANK1656-762 hereafter referred to as SHANK1). Using the acetylated analogue Ac-EESTSFQGP-CONH2 as a competitor for the interaction of SHANK1 with FAM-Ahx-EESTSFQGP-CONH2 or a typical fluorophore-labelled C-terminal PBM - GKAP - FITC-Ahx-EAQTRL-COOH - the internal SLiM was demonstrated to show comparable low-micromolar IC50 by competition fluorescent anisotropy. To gain further insight into the internal ligand interaction at the molecular level, we obtained the X-ray co-crystal structure of the Ac-EESTSFQGP-CONH2/SHANK1 complex and compared this to the Ac-EAQTRL-COOH/SHANK1 complex. The crystallographic studies reveal that the SHANK1 backbones for the two interactions overlap significantly. The main structural differences were shown to result from the flexible loops which reorganize to accommodate the two PBMs with distinct lengths and terminal groups. In addition, the two C-terminal residues Gly and Pro in Ac-EESTSFQGP-CONH2 were shown not to participate in interaction with the target protein, implying further truncation and structural modification using peptidomimetic approaches on this sequence may be feasible. Taken together, the SLiM Ac-EESTSFQGP-CONH2 holds potential as an internal ligand for targeting SHANK1.
Collapse
Affiliation(s)
- Yue Li
- School of Chemistry, University of Leeds, Woodhouse Lane, Leeds LS2 9JT, U.K
- Astbury Centre for Structural Molecular Biology, University of Leeds, Woodhouse Lane, Leeds LS2 9JT, U.K
| | - Chi H. Trinh
- Astbury Centre for Structural Molecular Biology, University of Leeds, Woodhouse Lane, Leeds LS2 9JT, U.K
- School of Molecular and Cellular Biology, University of Leeds, Woodhouse Lane, Leeds LS2 9JT, U.K
| | - Amanda Acevedo-Jake
- School of Chemistry, University of Birmingham, Edgbaston, Birmingham B15 2TT, U.K
| | - Diana Gimenez
- School of Chemistry, University of Birmingham, Edgbaston, Birmingham B15 2TT, U.K
| | - Stuart L. Warriner
- School of Chemistry, University of Leeds, Woodhouse Lane, Leeds LS2 9JT, U.K
- Astbury Centre for Structural Molecular Biology, University of Leeds, Woodhouse Lane, Leeds LS2 9JT, U.K
| | - Andrew J. Wilson
- School of Chemistry, University of Leeds, Woodhouse Lane, Leeds LS2 9JT, U.K
- Astbury Centre for Structural Molecular Biology, University of Leeds, Woodhouse Lane, Leeds LS2 9JT, U.K
- School of Chemistry, University of Birmingham, Edgbaston, Birmingham B15 2TT, U.K
| |
Collapse
|
31
|
Rossi M, Banskota N, Shin CH, Anerillas C, Tsitsipatis D, Yang JH, Munk R, Martindale J, Yang X, Piao Y, Mazan-Mamczarz K, Fan J, Lehrmann E, Lam KW, De S, Abdelmohsen K, Gorospe M. Increased PTCHD4 expression via m6A modification of PTCHD4 mRNA promotes senescent cell survival. Nucleic Acids Res 2024; 52:7261-7278. [PMID: 38721764 PMCID: PMC11229380 DOI: 10.1093/nar/gkae322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Revised: 03/27/2024] [Accepted: 04/12/2024] [Indexed: 07/09/2024] Open
Abstract
RNA modifications, including N6-methyladenosine (m6A), critically modulate protein expression programs in a range of cellular processes. Although the transcriptomes of cells undergoing senescence are strongly regulated, the landscape and impact of m6A modifications during senescence are poorly understood. Here, we report a robust m6A modification of PTCHD4 mRNA, encoding Patched Domain-Containing Protein 4, in senescent cells. The METTL3/METTL14 complex was found to incorporate the m6A modification on PTCHD4 mRNA; addition of m6A rendered PTCHD4 mRNA more stable and increased PTCHD4 production. MeRIP RT-qPCR and eCLIP analyses were used to map this m6A modification to the last exon of PTCHD4 mRNA. Further investigation identified IGF2BP1, but not other m6A readers, as responsible for the stabilization and increased abundance of m6A-modified PTCHD4 mRNA. Silencing PTCHD4, a transmembrane protein, enhanced growth arrest and DNA damage in pre-senescent cells and sensitized them to senolysis and apoptosis. Our results indicate that m6A modification of PTCHD4 mRNA increases the production of PTCHD4, a protein associated with senescent cell survival, supporting the notion that regulating m6A modification on specific mRNAs could be exploited to eliminate senescent cells for therapeutic benefit.
Collapse
Affiliation(s)
- Martina Rossi
- Laboratory of Genetics and Genomics, National Institute on Aging (NIA) Intramural Research Program (IRP), National Institutes of Health (NIH), Baltimore, MD, USA
| | - Nirad Banskota
- Laboratory of Genetics and Genomics, National Institute on Aging (NIA) Intramural Research Program (IRP), National Institutes of Health (NIH), Baltimore, MD, USA
| | - Chang Hoon Shin
- Laboratory of Genetics and Genomics, National Institute on Aging (NIA) Intramural Research Program (IRP), National Institutes of Health (NIH), Baltimore, MD, USA
| | - Carlos Anerillas
- Laboratory of Genetics and Genomics, National Institute on Aging (NIA) Intramural Research Program (IRP), National Institutes of Health (NIH), Baltimore, MD, USA
| | - Dimitrios Tsitsipatis
- Laboratory of Genetics and Genomics, National Institute on Aging (NIA) Intramural Research Program (IRP), National Institutes of Health (NIH), Baltimore, MD, USA
| | - Jen-Hao Yang
- Laboratory of Genetics and Genomics, National Institute on Aging (NIA) Intramural Research Program (IRP), National Institutes of Health (NIH), Baltimore, MD, USA
- Institute of Biomedical Sciences, National Sun Yat-sen University, Kaohsiung, Taiwan
| | - Rachel Munk
- Laboratory of Genetics and Genomics, National Institute on Aging (NIA) Intramural Research Program (IRP), National Institutes of Health (NIH), Baltimore, MD, USA
| | - Jennifer L Martindale
- Laboratory of Genetics and Genomics, National Institute on Aging (NIA) Intramural Research Program (IRP), National Institutes of Health (NIH), Baltimore, MD, USA
| | - Xiaoling Yang
- Laboratory of Genetics and Genomics, National Institute on Aging (NIA) Intramural Research Program (IRP), National Institutes of Health (NIH), Baltimore, MD, USA
| | - Yulan Piao
- Laboratory of Genetics and Genomics, National Institute on Aging (NIA) Intramural Research Program (IRP), National Institutes of Health (NIH), Baltimore, MD, USA
| | - Krystyna Mazan-Mamczarz
- Laboratory of Genetics and Genomics, National Institute on Aging (NIA) Intramural Research Program (IRP), National Institutes of Health (NIH), Baltimore, MD, USA
| | - Jinshui Fan
- Laboratory of Genetics and Genomics, National Institute on Aging (NIA) Intramural Research Program (IRP), National Institutes of Health (NIH), Baltimore, MD, USA
| | - Elin Lehrmann
- Laboratory of Genetics and Genomics, National Institute on Aging (NIA) Intramural Research Program (IRP), National Institutes of Health (NIH), Baltimore, MD, USA
| | - Kwan-Wood Gabriel Lam
- Laboratory of Genetics and Genomics, National Institute on Aging (NIA) Intramural Research Program (IRP), National Institutes of Health (NIH), Baltimore, MD, USA
| | - Supriyo De
- Laboratory of Genetics and Genomics, National Institute on Aging (NIA) Intramural Research Program (IRP), National Institutes of Health (NIH), Baltimore, MD, USA
| | - Kotb Abdelmohsen
- Laboratory of Genetics and Genomics, National Institute on Aging (NIA) Intramural Research Program (IRP), National Institutes of Health (NIH), Baltimore, MD, USA
| | - Myriam Gorospe
- Laboratory of Genetics and Genomics, National Institute on Aging (NIA) Intramural Research Program (IRP), National Institutes of Health (NIH), Baltimore, MD, USA
| |
Collapse
|
32
|
Maisonneuve P, Sahmi M, Bergeron-Labrecque F, Ma XI, Queguiner J, Arseneault G, Lefrançois M, Kurinov I, Fronzes R, Sicheri F, Therrien M. The CNK-HYP scaffolding complex promotes RAF activation by enhancing KSR-MEK interaction. Nat Struct Mol Biol 2024; 31:1028-1038. [PMID: 38388830 PMCID: PMC11257983 DOI: 10.1038/s41594-024-01233-6] [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: 01/23/2023] [Accepted: 01/29/2024] [Indexed: 02/24/2024]
Abstract
The RAS-MAPK pathway regulates cell proliferation, differentiation and survival, and its dysregulation is associated with cancer development. The pathway minimally comprises the small GTPase RAS and the kinases RAF, MEK and ERK. Activation of RAF by RAS is notoriously intricate and remains only partially understood. There are three RAF isoforms in mammals (ARAF, BRAF and CRAF) and two related pseudokinases (KSR1 and KSR2). RAS-mediated activation of RAF depends on an allosteric mechanism driven by the dimerization of its kinase domain. Recent work on human RAFs showed that MEK binding to KSR1 promotes KSR1-BRAF heterodimerization, which leads to the phosphorylation of free MEK molecules by BRAF. Similar findings were made with the single Drosophila RAF homolog. Here we show that the fly scaffold proteins CNK and HYP stabilize the KSR-MEK interaction, which in turn enhances RAF-KSR heterodimerization and RAF activation. The cryogenic electron microscopy structure of the minimal KSR-MEK-CNK-HYP complex reveals a ring-like arrangement of the CNK-HYP complex allowing CNK to simultaneously engage KSR and MEK, thus stabilizing the binary interaction. Together, these results illuminate how CNK contributes to RAF activation by stimulating the allosteric function of KSR and highlight the diversity of mechanisms impacting RAF dimerization as well as the regulatory potential of the KSR-MEK interaction.
Collapse
Affiliation(s)
- Pierre Maisonneuve
- Université de Bordeaux, CNRS, Bordeaux INP, CBMN, UMR 5248, IECB, F-33600, Pessac, France.
- Center for Molecular, Cell and Systems Biology, Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, Ontario, Canada.
| | - Malha Sahmi
- Institute for Research in Immunology and Cancer, Laboratory of Intracellular Signaling, Université de Montréal, Montréal, Quebec, Canada
| | - Fanny Bergeron-Labrecque
- Institute for Research in Immunology and Cancer, Laboratory of Intracellular Signaling, Université de Montréal, Montréal, Quebec, Canada
| | - Xianjie Iris Ma
- Center for Molecular, Cell and Systems Biology, Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, Ontario, Canada
| | - Juliette Queguiner
- Institute for Research in Immunology and Cancer, Laboratory of Intracellular Signaling, Université de Montréal, Montréal, Quebec, Canada
| | - Geneviève Arseneault
- Institute for Research in Immunology and Cancer, Laboratory of Intracellular Signaling, Université de Montréal, Montréal, Quebec, Canada
| | - Martin Lefrançois
- Institute for Research in Immunology and Cancer, Laboratory of Intracellular Signaling, Université de Montréal, Montréal, Quebec, Canada
| | - Igor Kurinov
- Department of Chemistry and Chemical Biology, Cornell University, NE-CAT, Argonne, IL, USA
| | - Rémi Fronzes
- Institut Européen de Chimie et Biologie, Université de Bordeaux-CNRS (UMR 5234), Pessac, France
| | - Frank Sicheri
- Center for Molecular, Cell and Systems Biology, Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, Ontario, Canada.
- Departments of Molecular Genetics and Biochemistry, University of Toronto, Toronto, Ontario, Canada.
| | - Marc Therrien
- Institute for Research in Immunology and Cancer, Laboratory of Intracellular Signaling, Université de Montréal, Montréal, Quebec, Canada.
- Département de pathologie et biologie cellulaire, Université de Montréal, Montréal, Quebec, Canada.
| |
Collapse
|
33
|
Giacon N, Lo Cascio E, Pennacchietti V, De Maio F, Santarelli G, Sibilia D, Tiberio F, Sanguinetti M, Lattanzi W, Toto A, Arcovito A. PDZ2-conjugated-PLGA nanoparticles are tiny heroes in the battle against SARS-CoV-2. Sci Rep 2024; 14:13059. [PMID: 38844490 PMCID: PMC11156922 DOI: 10.1038/s41598-024-63239-w] [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: 03/22/2024] [Accepted: 05/27/2024] [Indexed: 06/09/2024] Open
Abstract
The COVID-19 pandemic caused by SARS-CoV-2 has highlighted the urgent need for innovative antiviral strategies to fight viral infections. Although a substantial part of the overall effort has been directed at the Spike protein to create an effective global vaccination strategy, other proteins have also been examined and identified as possible therapeutic targets. Among them, although initially underestimated, there is the SARS-CoV-2 E-protein, which turned out to be a key factor in viral pathogenesis due to its role in virus budding, assembly and spreading. The C-terminus of E-protein contains a PDZ-binding motif (PBM) that plays a key role in SARS-CoV-2 virulence as it is recognized and bound by the PDZ2 domain of the human tight junction protein ZO-1. The binding between the PDZ2 domain of ZO-1 and the C-terminal portion of SARS-CoV-2 E-protein has been extensively characterized. Our results prompted us to develop a possible adjuvant therapeutic strategy aimed at slowing down or inhibiting virus-mediated pathogenesis. Such innovation consists in the design and synthesis of externally PDZ2-ZO1 functionalized PLGA-based nanoparticles to be used as intracellular decoy. Contrary to conventional strategies, this innovative approach aims to capitalize on the E protein-PDZ2 interaction to prevent virus assembly and replication. In fact, the conjugation of the PDZ2 domain to polymeric nanoparticles increases the affinity toward the E protein effectively creating a "molecular sponge" able to sequester E proteins within the intracellular environment of infected cells. Our in vitro studies on selected cellular models, show that these nanodevices significantly reduce SARS-CoV-2-mediated virulence, emphasizing the importance of exploiting viral-host interactions for therapeutic benefit.
Collapse
Affiliation(s)
- Noah Giacon
- Dipartimento di Scienze Biotecnologiche di Base, Cliniche Intensivologiche e Perioperatorie, Università Cattolica del Sacro Cuore, Largo F. Vito 1, 00168, Rome, Italy
| | - Ettore Lo Cascio
- Dipartimento di Scienze Biotecnologiche di Base, Cliniche Intensivologiche e Perioperatorie, Università Cattolica del Sacro Cuore, Largo F. Vito 1, 00168, Rome, Italy
| | - Valeria Pennacchietti
- Dipartimento di Scienze Biochimiche "A. Rossi Fanelli", Laboratory Affiliated to Istituto Pasteur Italia - Fondazione Cenci Bolognetti, Sapienza Università di Roma, P.le A. Moro 5, 00185, Rome, Italy
| | - Flavio De Maio
- Dipartimento di Scienze Biotecnologiche di Base, Cliniche Intensivologiche e Perioperatorie, Università Cattolica del Sacro Cuore, Largo F. Vito 1, 00168, Rome, Italy
- Dipartimento di Scienze di Laboratorio e Infettivologiche, Fondazione Policlinico Universitario "A. Gemelli", IRCCS, Largo A. Gemelli 8, 00168, Rome, Italy
| | - Giulia Santarelli
- Dipartimento di Scienze di Laboratorio e Infettivologiche, Fondazione Policlinico Universitario "A. Gemelli", IRCCS, Largo A. Gemelli 8, 00168, Rome, Italy
| | - Diego Sibilia
- Dipartimento di Scienze della Vita e Sanità Pubblica, Università Cattolica del Sacro Cuore, Largo F. Vito 1, 00168, Rome, Italy
| | - Federica Tiberio
- Dipartimento di Scienze della Vita e Sanità Pubblica, Università Cattolica del Sacro Cuore, Largo F. Vito 1, 00168, Rome, Italy
| | - Maurizio Sanguinetti
- Dipartimento di Scienze Biotecnologiche di Base, Cliniche Intensivologiche e Perioperatorie, Università Cattolica del Sacro Cuore, Largo F. Vito 1, 00168, Rome, Italy
- Dipartimento di Scienze di Laboratorio e Infettivologiche, Fondazione Policlinico Universitario "A. Gemelli", IRCCS, Largo A. Gemelli 8, 00168, Rome, Italy
| | - Wanda Lattanzi
- Dipartimento di Scienze della Vita e Sanità Pubblica, Università Cattolica del Sacro Cuore, Largo F. Vito 1, 00168, Rome, Italy
- Fondazione Policlinico Universitario "A. Gemelli", IRCCS, Largo A. Gemelli 8, 00168, Rome, Italy
| | - Angelo Toto
- Dipartimento di Scienze Biochimiche "A. Rossi Fanelli", Laboratory Affiliated to Istituto Pasteur Italia - Fondazione Cenci Bolognetti, Sapienza Università di Roma, P.le A. Moro 5, 00185, Rome, Italy.
| | - Alessandro Arcovito
- Dipartimento di Scienze Biotecnologiche di Base, Cliniche Intensivologiche e Perioperatorie, Università Cattolica del Sacro Cuore, Largo F. Vito 1, 00168, Rome, Italy.
- Dipartimento di Scienze della Vita e Sanità Pubblica, Università Cattolica del Sacro Cuore, Largo F. Vito 1, 00168, Rome, Italy.
| |
Collapse
|
34
|
Aliakbari S, Hasanzadeh L, Sayyah M, Amini N, Pourbadie HG. Induced expression of rabies glycoprotein in the dorsal hippocampus enhances hippocampal dependent memory in a rat model of Alzheimer's disease. J Neurovirol 2024; 30:274-285. [PMID: 38943023 DOI: 10.1007/s13365-024-01221-y] [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: 03/24/2024] [Revised: 06/12/2024] [Accepted: 06/12/2024] [Indexed: 06/30/2024]
Abstract
The Rabies virus is a neurotropic virus that manipulates the natural cell death processes of its host to ensure its own survival and replication. Studies have shown that the anti-apoptotic effect of the virus is mediated by one of its protein named, rabies glycoprotein (RVG). Alzheimer's disease (AD) is characterized by the loss of neural cells and memory impairment. We aim to examine whether expression of RVG in the hippocampal cells can shield the detrimental effects induced by Aβ. Oligomeric form of Aβ (oAβ) or vehicle was bilaterally microinjected into the dorsal hippocampus of male Wistar rats. One week later, two μl (108 T.U. /ml) of the lentiviral vector carrying RVG gene was injected into their dorsal hippocampus (post-treatment). In another experiment, the lentiviral vector was microinjected one week before Aβ injection (pre-treatment). One week later, the rat's brain was sliced into cross-sections, and the presence of RVG-expressing neuronal cells was confirmed using fluorescent microscopy. Rats were subjected to assessments of spatial learning and memory as well as passive avoidance using the Morris water maze (MWM) and the Shuttle box apparatuses, respectively. Protein expression of AMPA receptor subunit (GluA1) was determined using western blotting technique. In MWM, Aβ treated rats showed decelerated acquisition of the task and impairment of reference memory. RVG expression in the hippocampus prevented and restored the deficits in both pre- and post- treatment conditions, respectively. It also improved inhibitory memory in the oAβ treated rats. RVG increased the expression level of GluA1 level in the hippocampus. Based on our findings, the expression of RVG in the hippocampus has the potential to enhance both inhibitory and spatial learning abilities, ultimately improving memory performance in an AD rat model. This beneficial effect is likely attributed, at least in part, to the increased expression of GluA1-containing AMPA receptors.
Collapse
Affiliation(s)
- Shayan Aliakbari
- Department of Physiology and Pharmacology, Pasteur Institute of Iran, Tehran, Iran
| | - Leila Hasanzadeh
- Department of Physiology and Pharmacology, Pasteur Institute of Iran, Tehran, Iran
| | - Mohammad Sayyah
- Department of Physiology and Pharmacology, Pasteur Institute of Iran, Tehran, Iran
| | - Niloufar Amini
- Department of Physiology and Pharmacology, Pasteur Institute of Iran, Tehran, Iran
| | | |
Collapse
|
35
|
Bondarenko V, Chen Q, Tillman TS, Xu Y, Tang P. Unconventional PDZ Recognition Revealed in α7 nAChR-PICK1 Complexes. ACS Chem Neurosci 2024; 15:2070-2079. [PMID: 38691676 PMCID: PMC11099923 DOI: 10.1021/acschemneuro.4c00138] [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: 03/05/2024] [Revised: 04/16/2024] [Accepted: 04/19/2024] [Indexed: 05/03/2024] Open
Abstract
PDZ domains are modular domains that conventionally bind to C terminal or internal motifs of target proteins to control cellular functions through the regulation of protein complex assemblies. Almost all reported structures of PDZ-target protein complexes rely on fragments or peptides as target proteins. No intact target protein complexed with PDZ was structurally characterized. In this study, we used NMR spectroscopy and other biochemistry and biophysics tools to uncover insights into structural coupling between the PDZ domain of protein interacting with C-kinase 1 (PICK1) and α7 nicotinic acetylcholine receptors (α7 nAChR). Notably, the intracellular domains of both α7 nAChR and PICK1 PDZ exhibit a high degree of plasticity in their coupling. Specifically, the MA helix of α7 nAChR interacts with residues lining the canonical binding site of the PICK1 PDZ, while flexible loops also engage in protein-protein interactions. Both hydrophobic and electrostatic interactions mediate the coupling. Overall, the resulting structure of the α7 nAChR-PICK1 complex reveals an unconventional PDZ binding mode, significantly expanding the repertoire of functionally important PDZ interactions.
Collapse
Affiliation(s)
- Vasyl Bondarenko
- Depatment
of Anesthesiology and Perioperative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
| | - Qiang Chen
- Depatment
of Anesthesiology and Perioperative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
| | - Tommy S. Tillman
- Depatment
of Anesthesiology and Perioperative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
| | - Yan Xu
- Depatment
of Anesthesiology and Perioperative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
- Department
of Structural Biology, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
- Department
of Pharmacology and Chemical Biology, University
of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
- Department
of Physics and Astronomy, University of
Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
| | - Pei Tang
- Depatment
of Anesthesiology and Perioperative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
- Department
of Pharmacology and Chemical Biology, University
of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
- Department
of Computational and Systems Biology, University
of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
| |
Collapse
|
36
|
Omble A, Mahajan S, Bhoite A, Kulkarni K. Dishevelled2 activates WGEF via its interaction with a unique internal peptide motif of the GEF. Commun Biol 2024; 7:543. [PMID: 38714795 PMCID: PMC11076555 DOI: 10.1038/s42003-024-06194-6] [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: 11/04/2023] [Accepted: 04/15/2024] [Indexed: 05/10/2024] Open
Abstract
The Wnt-planar cell polarity (Wnt-PCP) pathway is crucial in establishing cell polarity during development and tissue homoeostasis. This pathway is found to be dysregulated in many pathological conditions, including cancer and autoimmune disorders. The central event in Wnt-PCP pathway is the activation of Weak-similarity guanine nucleotide exchange factor (WGEF) by the adapter protein Dishevelled (Dvl). The PDZ domain of Dishevelled2 (Dvl2PDZ) binds and activates WGEF by releasing it from its autoinhibitory state. However, the actual Dvl2PDZ binding site of WGEF and the consequent activation mechanism of the GEF have remained elusive. Using biochemical and molecular dynamics studies, we show that a unique "internal-PDZ binding motif" (IPM) of WGEF mediates the WGEF-Dvl2PDZ interaction to activate the GEF. The residues at P2, P0, P-2 and P-3 positions of IPM play an important role in stabilizing the WGEFpep-Dvl2PDZ interaction. Furthermore, MD simulations of modelled Dvl2PDZ-WGEFIPM peptide complexes suggest that WGEF-Dvl2PDZ interaction may differ from the reported Dvl2PDZ-IPM interactions. Additionally, the apo structure of human Dvl2PDZ shows conformational dynamics different from its IPM peptide bound state, suggesting an induced fit mechanism for the Dvl2PDZ-peptide interaction. The current study provides a model for Dvl2 induced activation of WGEF.
Collapse
Affiliation(s)
- Aishwarya Omble
- Division of Biochemical Sciences, CSIR-National Chemical Laboratory, Dr. Homi Bhabha Road, Pune, 411008, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Shrutika Mahajan
- Division of Biochemical Sciences, CSIR-National Chemical Laboratory, Dr. Homi Bhabha Road, Pune, 411008, India
| | - Ashwini Bhoite
- Division of Biochemical Sciences, CSIR-National Chemical Laboratory, Dr. Homi Bhabha Road, Pune, 411008, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Kiran Kulkarni
- Division of Biochemical Sciences, CSIR-National Chemical Laboratory, Dr. Homi Bhabha Road, Pune, 411008, India.
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India.
| |
Collapse
|
37
|
Schuhmann F, Ramsay JL, Kattnig DR, Solov’yov IA. Structural Rearrangements of Pigeon Cryptochrome 4 Undergoing a Complete Redox Cycle. J Phys Chem B 2024; 128:3844-3855. [PMID: 38568745 PMCID: PMC11056986 DOI: 10.1021/acs.jpcb.4c00424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Revised: 03/04/2024] [Accepted: 03/11/2024] [Indexed: 04/26/2024]
Abstract
Cryptochrome is currently the major contender of a protein to underpin magnetoreception, the ability to sense the Earth's magnetic field. Among various types of cryptochromes, cryptochrome 4 has been identified as the likely magnetoreceptor in migratory birds. All-atom molecular dynamics (MD) studies have offered first insights into the structural dynamics of cryptochrome but are limited to a short time scale due to large computational demands. Here, we employ coarse-grained MD simulations to investigate the emergence of long-lived states and conformational changes in pigeon cryptochrome 4. Our coarse-grained simulations complete the picture by permitting observation on a significantly longer time scale. We observe conformational transitions in the phosphate-binding loop of pigeon cryptochrome 4 upon activation and identify prominent motions in residues 440-460, suggesting a possible role as a signaling state of the protein or as a gated interaction site for forming protein complexes that might facilitate downstream processes. The findings highlight the importance of considering longer time scales in studying cryptochrome dynamics and magnetoreception. Coarse-grained MD simulations offer a valuable tool to unravel the complex behavior of cryptochrome proteins and shed new light on the mechanisms underlying their role in magnetoreception. Further exploration of these conformational changes and their functional implications may contribute to a deeper understanding of the molecular mechanisms of magnetoreception in birds.
Collapse
Affiliation(s)
- Fabian Schuhmann
- Institute
of Physics, Carl von Ossietzky Universität
Oldenburg, Carl-von-Ossietzky Str. 9-11, Oldenburg 26129, Germany
- Niels
Bohr International Academy, Niels Bohr Institute, University of Copenhagen, Blegdamsvej 17, Copenhagen 2100, Denmark
| | - Jessica L. Ramsay
- Living
Systems Institute and Department of Physics, University of Exeter, Stocker Rd., Exeter EX4
4QD, U.K.
| | - Daniel R. Kattnig
- Living
Systems Institute and Department of Physics, University of Exeter, Stocker Rd., Exeter EX4
4QD, U.K.
| | - Ilia A. Solov’yov
- Institute
of Physics, Carl von Ossietzky Universität
Oldenburg, Carl-von-Ossietzky Str. 9-11, Oldenburg 26129, Germany
- Research
Centre for Neurosensory Science, Carl von
Ossietzky Universität Oldenburg, Carl-von-Ossietzky-Str. 9-11, Oldenburg 26129, Germany
- Center
for Nanoscale Dynamics (CENAD), Carl von
Ossietzky Universität Oldenburg, Ammerländer Heerstr. 114-118, Oldenburg 26129, Germany
| |
Collapse
|
38
|
Walker V. The Intricacies of Renal Phosphate Reabsorption-An Overview. Int J Mol Sci 2024; 25:4684. [PMID: 38731904 PMCID: PMC11083860 DOI: 10.3390/ijms25094684] [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: 03/24/2024] [Revised: 04/17/2024] [Accepted: 04/19/2024] [Indexed: 05/13/2024] Open
Abstract
To maintain an optimal body content of phosphorus throughout postnatal life, variable phosphate absorption from food must be finely matched with urinary excretion. This amazing feat is accomplished through synchronised phosphate transport by myriads of ciliated cells lining the renal proximal tubules. These respond in real time to changes in phosphate and composition of the renal filtrate and to hormonal instructions. How they do this has stimulated decades of research. New analytical techniques, coupled with incredible advances in computer technology, have opened new avenues for investigation at a sub-cellular level. There has been a surge of research into different aspects of the process. These have verified long-held beliefs and are also dramatically extending our vision of the intense, integrated, intracellular activity which mediates phosphate absorption. Already, some have indicated new approaches for pharmacological intervention to regulate phosphate in common conditions, including chronic renal failure and osteoporosis, as well as rare inherited biochemical disorders. It is a rapidly evolving field. The aim here is to provide an overview of our current knowledge, to show where it is leading, and where there are uncertainties. Hopefully, this will raise questions and stimulate new ideas for further research.
Collapse
Affiliation(s)
- Valerie Walker
- Department of Clinical Biochemistry, University Hospital Southampton NHS Foundation Trust, Southampton General Hospital, Southampton S016 6YD, UK
| |
Collapse
|
39
|
Fujiwara-Tani R, Sasaki T, Bhawal UK, Mori S, Ogata R, Sasaki R, Ikemoto A, Kishi S, Fujii K, Ohmori H, Sho M, Kuniyasu H. Nuclear MAST4 Suppresses FOXO3 through Interaction with AKT3 and Induces Chemoresistance in Pancreatic Ductal Carcinoma. Int J Mol Sci 2024; 25:4056. [PMID: 38612866 PMCID: PMC11012408 DOI: 10.3390/ijms25074056] [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: 01/25/2024] [Revised: 03/24/2024] [Accepted: 04/02/2024] [Indexed: 04/14/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is highly malignant, with a 5-year survival rate of less than 10%. Furthermore, the acquisition of anticancer drug resistance makes PDAC treatment difficult. We established MIA-GEM cells, a PDAC cell line resistant to gemcitabine (GEM), a first-line anticancer drug, using the human PDAC cell line-MIA-PaCa-2. Microtubule-associated serine/threonine kinase-4 (MAST4) expression was increased in MIA-GEM cells compared with the parent cell line. Through inhibitor screening, dysregulated AKT signaling was identified in MIA-GEM cells with overexpression of AKT3. MAST4 knockdown effectively suppressed AKT3 overexpression, and both MAST4 and AKT3 translocation into the nucleus, phosphorylating forkhead box O3a (FOXO3) in MIA-GEM cells. Modulating FOXO3 target gene expression in these cells inhibited apoptosis while promoting stemness and proliferation. Notably, nuclear MAST4 demonstrated higher expression in GEM-resistant PDAC cases compared with that in the GEM-sensitive cases. Elevated MAST4 expression correlated with a poorer prognosis in PDAC. Consequently, nuclear MAST4 emerges as a potential marker for GEM resistance and poor prognosis, representing a novel therapeutic target for PDAC.
Collapse
Grants
- 19K16564 Ministry of Education, Culture, Sports, Science and Technology
- 20K21659 Ministry of Education, Culture, Sports, Science and Technology
- 23K10481 Ministry of Education, Culture, Sports, Science and Technology
- 22K11396 Ministry of Education, Culture, Sports, Science and Technology
- 21K11223 Ministry of Education, Culture, Sports, Science and Technology
- 22H04922 Ministry of Education, Culture, Sports, Science and Technology
Collapse
Affiliation(s)
- Rina Fujiwara-Tani
- Department of Molecular Pathology, Nara Medical University, 840 Shijo-cho, Kashihara 634-8521, Nara, Japan; (T.S.); (S.M.); (R.O.); (A.I.); (S.K.); (K.F.); (H.O.)
| | - Takamitsu Sasaki
- Department of Molecular Pathology, Nara Medical University, 840 Shijo-cho, Kashihara 634-8521, Nara, Japan; (T.S.); (S.M.); (R.O.); (A.I.); (S.K.); (K.F.); (H.O.)
| | - Ujjal Kumar Bhawal
- Research Institute of Oral Science, Nihon University School of Dentistry at Matsudo, Matsudo 271-8587, Chiba, Japan;
| | - Shiori Mori
- Department of Molecular Pathology, Nara Medical University, 840 Shijo-cho, Kashihara 634-8521, Nara, Japan; (T.S.); (S.M.); (R.O.); (A.I.); (S.K.); (K.F.); (H.O.)
| | - Ruiko Ogata
- Department of Molecular Pathology, Nara Medical University, 840 Shijo-cho, Kashihara 634-8521, Nara, Japan; (T.S.); (S.M.); (R.O.); (A.I.); (S.K.); (K.F.); (H.O.)
| | - Rika Sasaki
- Department of Molecular Pathology, Nara Medical University, 840 Shijo-cho, Kashihara 634-8521, Nara, Japan; (T.S.); (S.M.); (R.O.); (A.I.); (S.K.); (K.F.); (H.O.)
| | - Ayaka Ikemoto
- Department of Molecular Pathology, Nara Medical University, 840 Shijo-cho, Kashihara 634-8521, Nara, Japan; (T.S.); (S.M.); (R.O.); (A.I.); (S.K.); (K.F.); (H.O.)
| | - Shingo Kishi
- Department of Molecular Pathology, Nara Medical University, 840 Shijo-cho, Kashihara 634-8521, Nara, Japan; (T.S.); (S.M.); (R.O.); (A.I.); (S.K.); (K.F.); (H.O.)
- Pathology Laboratory, Research Institute, Tokushukai Nozaki Hospital, 2-10-50 Tanigawa, Daito 574-0074, Osaka, Japan
| | - Kiyomu Fujii
- Department of Molecular Pathology, Nara Medical University, 840 Shijo-cho, Kashihara 634-8521, Nara, Japan; (T.S.); (S.M.); (R.O.); (A.I.); (S.K.); (K.F.); (H.O.)
| | - Hitoshi Ohmori
- Department of Molecular Pathology, Nara Medical University, 840 Shijo-cho, Kashihara 634-8521, Nara, Japan; (T.S.); (S.M.); (R.O.); (A.I.); (S.K.); (K.F.); (H.O.)
| | - Masayuki Sho
- Department of Surgery, Nara Medical University, Kashihara 634-8522, Nara, Japan;
| | - Hiroki Kuniyasu
- Department of Molecular Pathology, Nara Medical University, 840 Shijo-cho, Kashihara 634-8521, Nara, Japan; (T.S.); (S.M.); (R.O.); (A.I.); (S.K.); (K.F.); (H.O.)
| |
Collapse
|
40
|
Ajel M, Jazayeri SM, Behboudi E, Poorebrahim M, Ahangar Oskouee M, Bannazadeh Baghi H, Hasani A, Varshochi M, Shekarchi AA, Sabbaghian M, Poortahmasebi V. Investigation of the Mutations in the SARS-CoV-2 Envelope Protein and Its Interaction with the PALS1 by Molecular Docking. Rep Biochem Mol Biol 2024; 13:124-136. [PMID: 39582830 PMCID: PMC11580137 DOI: 10.61186/rbmb.13.1.124] [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: 04/22/2024] [Accepted: 08/25/2024] [Indexed: 11/26/2024]
Abstract
Background The envelope (E) protein of globally circulating severe acute respiratory syndrome coronavirus 2 (SARS CoV 2) is highly conserved. This study aimed to find the mutation rate of the E genes in COVID-19 patients, and also to evaluate the conformational characteristics of viral E protein. Methods In this study, 120 patients with SARS-CoV-2 positive test results were selected according to real-time PCR assay. Specific primers for conventional PCR have been used to amplify E gene; furthermore, to identify the E gene mutations, direct sequencing of the E genes was also done. Bioinformatics techniques were used to investigate the possible effects of antigenic changes and 3D characteristics of amino acid substitutions. Also, the immunogenicity of wild-type and mutant E was analyzed utilizing a ClusPro docking server and the IEDB online platform. Results A total of 120 COVID-19 patients were included (57.5% were male and 42.5% female), with an overall mean age of 55.70±10.61 years old. Of 10 nucleotide changes, 8 (80%) were silent. Also, 2 (20%) missense mutations (amino acid altering) were found in the E gene (L73F and S68F). Conclusions These mutations insert some new helix structures in the E mutants. Also, the results of molecular docking studies indicated that both S68F and L73F mutations could notably enhance the stability and binding affinity of protein E's C-terminal motif to the Protein Associated with LIN7 1, MAGUK P55 Family Member (PALS1) which may probably increase local viral spread, and infiltration of immune cells into lung alveolar spaces.
Collapse
Affiliation(s)
- Maryam Ajel
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
- Department of Bacteriology and Virology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Seyed Mohammad Jazayeri
- Research Center of Pediatric Infectious Diseases, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran.
| | - Emad Behboudi
- Department of Basic Medical Sciences, Khoy University of Medical Sciences, Khoy, Iran.
| | - Mansour Poorebrahim
- Targeted Tumor Vaccines Group, Clinical Cooperation Unit Applied Tumor Immunity, German Cancer Research Center (DKFZ), Heidelberg, Germany.
| | - Mahin Ahangar Oskouee
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
- Department of Bacteriology and Virology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Hossein Bannazadeh Baghi
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
- Department of Bacteriology and Virology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Alka Hasani
- Department of Bacteriology and Virology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Mojtaba Varshochi
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Ali Akbar Shekarchi
- Department of Pathology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Mohammad Sabbaghian
- Department of Bacteriology and Virology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Vahdat Poortahmasebi
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
- Department of Bacteriology and Virology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
41
|
Dhupar R, Powers AA, Eisenberg SH, Gemmill RM, Bardawil CE, Udoh HM, Cubitt A, Nangle LA, Soloff AC. Orchestrating Resilience: How Neuropilin-2 and Macrophages Contribute to Cardiothoracic Disease. J Clin Med 2024; 13:1446. [PMID: 38592275 PMCID: PMC10934188 DOI: 10.3390/jcm13051446] [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: 01/16/2024] [Revised: 02/21/2024] [Accepted: 02/24/2024] [Indexed: 04/10/2024] Open
Abstract
Immunity has evolved to balance the destructive nature of inflammation with wound healing to overcome trauma, infection, environmental insults, and rogue malignant cells. The inflammatory response is marked by overlapping phases of initiation, resolution, and post-resolution remodeling. However, the disruption of these events can lead to prolonged tissue damage and organ dysfunction, resulting long-term disease states. Macrophages are the archetypic phagocytes present within all tissues and are important contributors to these processes. Pleiotropic and highly plastic in their responses, macrophages support tissue homeostasis, repair, and regeneration, all while balancing immunologic self-tolerance with the clearance of noxious stimuli, pathogens, and malignant threats. Neuropilin-2 (Nrp2), a promiscuous co-receptor for growth factors, semaphorins, and integrins, has increasingly been recognized for its unique role in tissue homeostasis and immune regulation. Notably, recent studies have begun to elucidate the role of Nrp2 in both non-hematopoietic cells and macrophages with cardiothoracic disease. Herein, we describe the unique role of Nrp2 in diseases of the heart and lung, with an emphasis on Nrp2 in macrophages, and explore the potential to target Nrp2 as a therapeutic intervention.
Collapse
Affiliation(s)
- Rajeev Dhupar
- Department of Cardiothoracic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; (R.D.); (H.M.U.)
- UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Surgical and Research Services, VA Pittsburgh Healthcare System, Pittsburgh, PA 15240, USA
| | - Amy A. Powers
- Department of Cardiothoracic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; (R.D.); (H.M.U.)
| | - Seth H. Eisenberg
- Department of Cardiothoracic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; (R.D.); (H.M.U.)
| | - Robert M. Gemmill
- Division of Hematology/Oncology, Department of Medicine, Medical University of South Carolina, Charleston, SC 29425, USA;
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Charles E. Bardawil
- Department of Cardiothoracic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; (R.D.); (H.M.U.)
| | - Hannah M. Udoh
- Department of Cardiothoracic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; (R.D.); (H.M.U.)
| | - Andrea Cubitt
- aTyr Pharma, San Diego, CA 92121, USA; (A.C.); (L.A.N.)
| | | | - Adam C. Soloff
- Department of Cardiothoracic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; (R.D.); (H.M.U.)
- UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Surgical and Research Services, VA Pittsburgh Healthcare System, Pittsburgh, PA 15240, USA
| |
Collapse
|
42
|
Guerin N, Childs H, Zhou P, Donald BR. DexDesign: A new OSPREY-based algorithm for designing de novo D-peptide inhibitors. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.12.579944. [PMID: 38405797 PMCID: PMC10888900 DOI: 10.1101/2024.02.12.579944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/27/2024]
Abstract
With over 270 unique occurrences in the human genome, peptide-recognizing PDZ domains play a central role in modulating polarization, signaling, and trafficking pathways. Mutations in PDZ domains lead to diseases such as cancer and cystic fibrosis, making PDZ domains attractive targets for therapeutic intervention. D-peptide inhibitors offer unique advantages as therapeutics, including increased metabolic stability and low immunogenicity. Here, we introduce DexDesign, a novel OSPREY-based algorithm for computationally designing de novo D-peptide inhibitors. DexDesign leverages three novel techniques that are broadly applicable to computational protein design: the Minimum Flexible Set, K*-based Mutational Scan, and Inverse Alanine Scan, which enable exponential reductions in the size of the peptide sequence search space. We apply these techniques and DexDesign to generate novel D-peptide inhibitors of two biomedically important PDZ domain targets: CAL and MAST2. We introduce a new framework for analyzing de novo peptides-evaluation along a replication/restitution axis-and apply it to the DexDesign-generated D-peptides. Notably, the peptides we generated are predicted to bind their targets tighter than their targets' endogenous ligands, validating the peptides' potential as lead therapeutic candidates. We provide an implementation of DexDesign in the free and open source computational protein design software OSPREY.
Collapse
|
43
|
Guerin N, Childs H, Zhou P, Donald BR. DexDesign: an OSPREY-based algorithm for designing de novo D-peptide inhibitors. Protein Eng Des Sel 2024; 37:gzae007. [PMID: 38757573 PMCID: PMC11099876 DOI: 10.1093/protein/gzae007] [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: 09/03/2023] [Revised: 04/17/2024] [Indexed: 05/18/2024] Open
Abstract
With over 270 unique occurrences in the human genome, peptide-recognizing PDZ domains play a central role in modulating polarization, signaling, and trafficking pathways. Mutations in PDZ domains lead to diseases such as cancer and cystic fibrosis, making PDZ domains attractive targets for therapeutic intervention. D-peptide inhibitors offer unique advantages as therapeutics, including increased metabolic stability and low immunogenicity. Here, we introduce DexDesign, a novel OSPREY-based algorithm for computationally designing de novo D-peptide inhibitors. DexDesign leverages three novel techniques that are broadly applicable to computational protein design: the Minimum Flexible Set, K*-based Mutational Scan, and Inverse Alanine Scan. We apply these techniques and DexDesign to generate novel D-peptide inhibitors of two biomedically important PDZ domain targets: CAL and MAST2. We introduce a framework for analyzing de novo peptides-evaluation along a replication/restitution axis-and apply it to the DexDesign-generated D-peptides. Notably, the peptides we generated are predicted to bind their targets tighter than their targets' endogenous ligands, validating the peptides' potential as lead inhibitors. We also provide an implementation of DexDesign in the free and open source computational protein design software OSPREY.
Collapse
Affiliation(s)
- Nathan Guerin
- Department of Computer Science, Duke University, 308 Research Drive, Durham, NC 27708, United States
| | - Henry Childs
- Department of Chemistry, Duke University, 124 Science Drive, Durham, NC 27708, United States
| | - Pei Zhou
- Department of Biochemistry, Duke University School of Medicine, 307 Research Drive, Durham, NC 22710, United States
| | - Bruce R Donald
- Department of Computer Science, Duke University, 308 Research Drive, Durham, NC 27708, United States
- Department of Chemistry, Duke University, 124 Science Drive, Durham, NC 27708, United States
- Department of Biochemistry, Duke University School of Medicine, 307 Research Drive, Durham, NC 22710, United States
- Department of Mathematics, Duke University, 120 Science Drive, Durham, NC 27708, United States
| |
Collapse
|
44
|
Bhayana S, Dougherty JA, Kamigaki Y, Agrawal S, Wijeratne S, Fitch J, Waller AP, Wolfgang KJ, White P, Kerlin BA, Smoyer WE. Glucocorticoid- and pioglitazone-induced proteinuria reduction in experimental NS both correlate with glomerular ECM modulation. iScience 2024; 27:108631. [PMID: 38188512 PMCID: PMC10770536 DOI: 10.1016/j.isci.2023.108631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 10/31/2023] [Accepted: 11/30/2023] [Indexed: 01/09/2024] Open
Abstract
Idiopathic nephrotic syndrome (NS) is a common glomerular disease. Although glucocorticoids (GC) are the primary treatment, the PPARγ agonist pioglitazone (Pio) also reduces proteinuria in patients with NS and directly protects podocytes from injury. Because both drugs reduce proteinuria, we hypothesized these effects result from overlapping transcriptional patterns. Systems biology approaches compared glomerular transcriptomes from rats with PAN-induced NS treated with GC vs. Pio and identified 29 commonly regulated genes-of-interest, primarily involved in extracellular matrix (ECM) remodeling. Correlation with clinical idiopathic NS patient datasets confirmed glomerular ECM dysregulation as a potential mechanism of injury. Cellular deconvolution in silico revealed GC- and Pio-induced amelioration of altered genes primarily within podocytes and mesangial cells. While validation studies are indicated, these analyses identified molecular pathways involved in the early stages of NS (prior to scarring), suggesting that targeting glomerular ECM dysregulation may enable a future non-immunosuppressive approach for proteinuria reduction in idiopathic NS.
Collapse
Affiliation(s)
- Sagar Bhayana
- Center for Clinical and Translational Research, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43205, USA
| | - Julie A. Dougherty
- Center for Clinical and Translational Research, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43205, USA
| | - Yu Kamigaki
- Center for Clinical and Translational Research, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43205, USA
| | - Shipra Agrawal
- Center for Clinical and Translational Research, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43205, USA
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH 43210, USA
| | - Saranga Wijeratne
- Institute for Genomic Medicine, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43205, USA
| | - James Fitch
- Institute for Genomic Medicine, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43205, USA
| | - Amanda P. Waller
- Center for Clinical and Translational Research, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43205, USA
| | - Katelyn J. Wolfgang
- Center for Clinical and Translational Research, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43205, USA
| | - Peter White
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH 43210, USA
- Institute for Genomic Medicine, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43205, USA
| | - Bryce A. Kerlin
- Center for Clinical and Translational Research, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43205, USA
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH 43210, USA
| | - William E. Smoyer
- Center for Clinical and Translational Research, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43205, USA
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH 43210, USA
| |
Collapse
|
45
|
Wang T, Wang Z, Wang R, Zhang L, Zhang Y, Lu H. Highly efficient and chemoselective blocking of free amino group by ortho-phthalaldehyde (OPA) for comprehensive analysis of protein terminome. Talanta 2024; 267:125262. [PMID: 37804787 DOI: 10.1016/j.talanta.2023.125262] [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: 07/08/2023] [Revised: 09/24/2023] [Accepted: 09/30/2023] [Indexed: 10/09/2023]
Abstract
Herein, we introduced ortho-phthalaldehyde (OPA) for blocking free amino groups and established a simple and robust method for comprehensive profiling of protein terminome based on strong cation exchange chromatography (SCX) fractionation. With the highly efficient and chemoseletive amine-group blocking, we identified 2271 canonical human protein N-termini, 1650 canonical human protein C-termini, as well as 645 protein neo-N-termini from HeLa cells.
Collapse
Affiliation(s)
- Ting Wang
- Department of Chemistry and Liver Cancer Institute of Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Zhongjie Wang
- Institutes of Biomedical Sciences and NHC Key Laboratory of Glycoconjugates Research, Fudan University, Shanghai, 200032, China
| | - Rui Wang
- Institutes of Biomedical Sciences and NHC Key Laboratory of Glycoconjugates Research, Fudan University, Shanghai, 200032, China
| | - Lei Zhang
- Institutes of Biomedical Sciences and NHC Key Laboratory of Glycoconjugates Research, Fudan University, Shanghai, 200032, China
| | - Ying Zhang
- Department of Chemistry and Liver Cancer Institute of Zhongshan Hospital, Fudan University, Shanghai, 200032, China; Institutes of Biomedical Sciences and NHC Key Laboratory of Glycoconjugates Research, Fudan University, Shanghai, 200032, China.
| | - Haojie Lu
- Department of Chemistry and Liver Cancer Institute of Zhongshan Hospital, Fudan University, Shanghai, 200032, China; Institutes of Biomedical Sciences and NHC Key Laboratory of Glycoconjugates Research, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
46
|
Park S, Fan J, Chamakuri S, Palaniappan M, Sharma K, Qin X, Wang J, Tan Z, Judge A, Hu L, Sankaran B, Li F, Prasad BVV, Matzuk MM, Palzkill T. Exploiting the Carboxylate-Binding Pocket of β-Lactamase Enzymes Using a Focused DNA-Encoded Chemical Library. J Med Chem 2024; 67:620-642. [PMID: 38117688 PMCID: PMC11489902 DOI: 10.1021/acs.jmedchem.3c01834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2023]
Abstract
β-Lactamase enzymes hydrolyze and thereby provide bacterial resistance to the important β-lactam class of antibiotics. The OXA-48 and NDM-1 β-lactamases cause resistance to the last-resort β-lactams, carbapenems, leading to a serious public health threat. Here, we utilized DNA-encoded chemical library (DECL) technology to discover novel β-lactamase inhibitors. We exploited the β-lactamase enzyme-substrate binding interactions and created a DECL targeting the carboxylate-binding pocket present in all β-lactamases. A library of 106 compounds, each containing a carboxylic acid or a tetrazole as an enzyme recognition element, was designed, constructed, and used to identify OXA-48 and NDM-1 inhibitors with micromolar to nanomolar potency. Further optimization led to NDM-1 inhibitors with increased potencies and biological activities. This work demonstrates that the carboxylate-binding pocket-targeting DECL, designed based on substrate binding information, aids in inhibitor identification and led to the discovery of novel non-β-lactam pharmacophores for the development of β-lactamase inhibitors for enzymes of different structural and mechanistic classes.
Collapse
Affiliation(s)
- Suhyeorn Park
- Verna and Marrs McLean Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, Houston, Texas 77030, United States
| | - Jiayi Fan
- Verna and Marrs McLean Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, Houston, Texas 77030, United States
| | - Srinivas Chamakuri
- Center for Drug Discovery, Department of Pathology & Immunology, Baylor College of Medicine, Houston, Texas 77030, United States
| | - Murugesan Palaniappan
- Center for Drug Discovery, Department of Pathology & Immunology, Baylor College of Medicine, Houston, Texas 77030, United States
| | - Kiran Sharma
- Center for Drug Discovery, Department of Pathology & Immunology, Baylor College of Medicine, Houston, Texas 77030, United States
| | - Xuan Qin
- Center for Drug Discovery, Department of Pathology & Immunology, Baylor College of Medicine, Houston, Texas 77030, United States
| | - Jian Wang
- Center for Drug Discovery, Department of Pathology & Immunology, Baylor College of Medicine, Houston, Texas 77030, United States
| | - Zhi Tan
- Verna and Marrs McLean Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, Houston, Texas 77030, United States
- Center for Drug Discovery, Department of Pathology & Immunology, Baylor College of Medicine, Houston, Texas 77030, United States
| | - Allison Judge
- Verna and Marrs McLean Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, Houston, Texas 77030, United States
| | - Liya Hu
- Verna and Marrs McLean Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, Houston, Texas 77030, United States
| | - Banumathi Sankaran
- Berkeley Center for Structural Biology, Advanced Light Source, Lawrence Berkeley National Lab, Berkeley, California 94720, United States
| | - Feng Li
- Center for Drug Discovery, Department of Pathology & Immunology, Baylor College of Medicine, Houston, Texas 77030, United States
| | - B V Venkataram Prasad
- Verna and Marrs McLean Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, Houston, Texas 77030, United States
| | - Martin M Matzuk
- Verna and Marrs McLean Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, Houston, Texas 77030, United States
- Center for Drug Discovery, Department of Pathology & Immunology, Baylor College of Medicine, Houston, Texas 77030, United States
| | - Timothy Palzkill
- Verna and Marrs McLean Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, Houston, Texas 77030, United States
| |
Collapse
|
47
|
Naik MT, Naik N, Hu T, Wang SH, Marshall J. Structure-based design of peptidomimetic inhibitors of PSD-95 with improved affinity for the PDZ3 domain. FEBS Lett 2024; 598:233-241. [PMID: 37904289 PMCID: PMC10842001 DOI: 10.1002/1873-3468.14767] [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: 08/09/2023] [Revised: 09/28/2023] [Accepted: 10/12/2023] [Indexed: 11/01/2023]
Abstract
Aberrant brain-derived neurotrophic factor (BDNF) signaling has been proposed to contribute to the pathophysiology of depression and other neurological disorders such as Angelman syndrome. We have previously shown that targeting the tropomyosin receptor kinase B/postsynaptic density protein-95 (PSD-95) nexus in the BDNF signaling pathway by peptidomimetic inhibitors is a promising approach for therapeutic intervention. Here, we used structure-based knowledge to develop a new Syn3 peptidomimetic compound series that fuses peptides derived from the PSD-95-binding protein SynGAP to our prototype compound CN2097. The new compounds target the PSD-95 PDZ3 domain and adjoining αC helix to achieve bivalent binding that results in up to 7-fold stronger affinity compared to CN2097. These compounds were designed to improve CN2097 specificity for the PSD-95 PDZ3 domain, and structure-activity relationship studies were performed to improve their resistance to proteolysis.
Collapse
Affiliation(s)
- Mandar T. Naik
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, Rhode Island, 02912, United States of America
- Department of Molecular Pharmacology, Physiology and Biotechnology, Brown University, Providence, Rhode Island, 02912, United States of America
| | - Nandita Naik
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, Rhode Island, 02912, United States of America
- Department of Molecular Pharmacology, Physiology and Biotechnology, Brown University, Providence, Rhode Island, 02912, United States of America
| | - Tony Hu
- Department of Molecular Pharmacology, Physiology and Biotechnology, Brown University, Providence, Rhode Island, 02912, United States of America
| | - Szu-Huan Wang
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, Rhode Island, 02912, United States of America
- Department of Molecular Pharmacology, Physiology and Biotechnology, Brown University, Providence, Rhode Island, 02912, United States of America
| | - John Marshall
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, Rhode Island, 02912, United States of America
- Department of Molecular Pharmacology, Physiology and Biotechnology, Brown University, Providence, Rhode Island, 02912, United States of America
| |
Collapse
|
48
|
Healy MD, Collins BM. The PDLIM family of actin-associated proteins and their emerging role in membrane trafficking. Biochem Soc Trans 2023; 51:2005-2016. [PMID: 38095060 PMCID: PMC10754285 DOI: 10.1042/bst20220804] [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: 09/29/2023] [Revised: 11/29/2023] [Accepted: 12/04/2023] [Indexed: 12/21/2023]
Abstract
The PDZ and LIM domain (PDLIM) proteins are associated with the actin cytoskeleton and have conserved in roles in metazoan actin organisation and function. They primarily function as scaffolds linking various proteins to actin and its binding partner α-actinin via two conserved domains; an N-terminal postsynaptic density 95, discs large and zonula occludens-1 (PDZ) domain, and either single or multiple C-terminal LIN-11, Isl-1 and MEC-3 (LIM) domains in the actinin-associated LIM protein (ALP)- and Enigma-related proteins, respectively. While their role in actin organisation, such as in stress fibres or in the Z-disc of muscle fibres is well known, emerging evidence also suggests a role in actin-dependent membrane trafficking in the endosomal system. This is mediated by a recently identified interaction with the sorting nexin 17 (SNX17) protein, an adaptor for the trafficking complex Commander which is itself intimately linked to actin-directed formation of endosomal recycling domains. In this review we focus on the currently understood structural basis for PDLIM function. The PDZ domains mediate direct binding to distinct classes of PDZ-binding motifs (PDZbms), including α-actinin and other actin-associated proteins, and a highly specific interaction with the type III PDZbm such as the one found in the C-terminus of SNX17. The structures of the LIM domains are less well characterised and how they engage with their ligands is completely unknown. Despite the lack of experimental structural data, we find that recently developed machine learning-based structure prediction methods provide insights into their potential interactions and provide a template for further studies of their molecular functions.
Collapse
Affiliation(s)
- Michael D. Healy
- The University of Queensland, Institute for Molecular Bioscience, St Lucia, Queensland 4072, Australia
| | - Brett M. Collins
- The University of Queensland, Institute for Molecular Bioscience, St Lucia, Queensland 4072, Australia
| |
Collapse
|
49
|
Casanova-Sepúlveda G, Sexton JA, Turk BE, Boggon TJ. Autoregulation of the LIM kinases by their PDZ domain. Nat Commun 2023; 14:8441. [PMID: 38114480 PMCID: PMC10730565 DOI: 10.1038/s41467-023-44148-4] [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: 03/15/2023] [Accepted: 12/01/2023] [Indexed: 12/21/2023] Open
Abstract
LIM domain kinases (LIMK) are important regulators of actin cytoskeletal remodeling. These protein kinases phosphorylate the actin depolymerizing factor cofilin to suppress filament severing, and are key nodes between Rho GTPase cascades and actin. The two mammalian LIMKs, LIMK1 and LIMK2, contain consecutive LIM domains and a PDZ domain upstream of the C-terminal kinase domain. The roles of the N-terminal regions are not fully understood, and the function of the PDZ domain remains elusive. Here, we determine the 2.0 Å crystal structure of the PDZ domain of LIMK2 and reveal features not previously observed in PDZ domains including a core-facing arginine residue located at the second position of the 'x-Φ-G-Φ' motif, and that the expected peptide binding cleft is shallow and poorly conserved. We find a distal extended surface to be highly conserved, and when LIMK1 was ectopically expressed in yeast we find targeted mutagenesis of this surface decreases growth, implying increased LIMK activity. PDZ domain LIMK1 mutants expressed in yeast are hyperphosphorylated and show elevated activity in vitro. This surface in both LIMK1 and LIMK2 is critical for autoregulation independent of activation loop phosphorylation. Overall, our study demonstrates the functional importance of the PDZ domain to autoregulation of LIMKs.
Collapse
Affiliation(s)
| | - Joel A Sexton
- Department of Pharmacology, Yale University, New Haven, CT, 06520, USA
| | - Benjamin E Turk
- Department of Pharmacology, Yale University, New Haven, CT, 06520, USA
| | - Titus J Boggon
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT, 06520, USA.
- Department of Pharmacology, Yale University, New Haven, CT, 06520, USA.
| |
Collapse
|
50
|
Seo H, Lee HC, Lee KC, Kim D, Kim J, Kang D, Chung HJ, Cha HJ, Kim J, Song KS. PDZ Peptide of the ZO-1 Protein Significantly Increases UTP-Induced MUC8 Anti-Inflammatory Mucin Overproduction in Human Airway Epithelial Cells. Mol Cells 2023; 46:700-709. [PMID: 37750239 PMCID: PMC10654460 DOI: 10.14348/molcells.2023.0107] [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: 07/03/2023] [Revised: 08/29/2023] [Accepted: 09/03/2023] [Indexed: 09/27/2023] Open
Abstract
Mucus hyperproduction and hypersecretion are observed often in respiratory diseases. MUC8 is a glycoprotein synthesized by epithelial cells and generally expressed in the respiratory track. However, the physiological mechanism by which extracellular nucleotides induce MUC8 gene expression in human airway epithelial cells is unclear. Here, we show that UTP could induce MUC8 gene expression through P2Y2-PLCβ3-Ca2+ activation. Because the full-length cDNA sequence of MUC8 has not been identified, a specific siRNA-MUC8 was designed based on the partial cDNA sequence of MUC8. siRNA-MUC8 significantly increased TNF-α production and decreased IL-1Ra production, suggesting that MUC8 may downregulate UTP/P2Y2-induced airway inflammation. Interestingly, the PDZ peptide of ZO-1 protein strongly abolished UTP-induced TNF-α production and increased IL-1Ra production and MUC8 gene expression. In addition, the PDZ peptide dramatically increased the levels of UTP-induced ZO proteins and TEER (trans-epithelial electrical resistance). These results show that the anti-inflammatory mucin MUC8 may contribute to homeostasis, and the PDZ peptide can be a novel therapeutic candidate for UTP-induced airway inflammation.
Collapse
Affiliation(s)
- Han Seo
- Department of Anesthesiology and Pain Medicine, Kosin University College of Medicine, Busan 49267, Korea
| | - Hyun-Chae Lee
- Department of Medical Science, Kosin University College of Medicine, Busan 49267, Korea
| | - Ki Chul Lee
- Department of Anesthesiology and Pain Medicine, Kosin University College of Medicine, Busan 49267, Korea
| | - Doosik Kim
- Department of Anesthesiology and Pain Medicine, Kosin University College of Medicine, Busan 49267, Korea
| | - Jiwook Kim
- Department of Anesthesiology and Pain Medicine, Kosin University College of Medicine, Busan 49267, Korea
| | - Donghee Kang
- Department of Anesthesiology and Pain Medicine, Kosin University College of Medicine, Busan 49267, Korea
| | - Hyung-Joo Chung
- Department of Anesthesiology and Pain Medicine, Kosin University College of Medicine, Busan 49267, Korea
| | - Hee-Jae Cha
- Department of Genetics, Kosin University College of Medicine, Busan 49267, Korea
| | - Jeongtae Kim
- Department of Anatomy, Kosin University College of Medicine, Busan 49267, Korea
| | - Kyoung Seob Song
- Department of Medical Science, Kosin University College of Medicine, Busan 49267, Korea
| |
Collapse
|