1
|
Bracaglia L, Oliveti S, Felli IC, Pierattelli R. Decoding Order and Disorder in Proteins by NMR Spectroscopy. J Am Chem Soc 2025; 147:13146-13157. [PMID: 40223218 DOI: 10.1021/jacs.4c14959] [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: 04/15/2025]
Abstract
Proteins often have a complex architecture, consisting of both globular ordered domains and intrinsically disordered regions (IDRs). These multidomain proteins pose challenges for traditional structural biology techniques. One major difficulty arises from the dynamic and flexible nature of IDRs, which lack a stable three-dimensional structure. Indeed, this feature further complicates the application of traditional structural biology techniques. Characterizing these systems is typically simplified by isolating individual domains, which can provide valuable insights into the structure and function of specific regions. However, this approach overlooks the interactions and regulatory mechanisms that occur between domains. To capture the full functional and structural complexity of multidomain proteins, it is crucial to study larger constructs. In this study, we focused on the CREB binding protein (CBP), a pivotal protein involved in numerous cellular processes. CBP is characterized by its modular structure, featuring alternating globular domains and IDRs. We specifically examined the TAZ4 construct, encompassing the TAZ2 globular domain and the ID4 flexible linker region. To characterize this multidomain system, we designed NMR experiments that take advantage of the dynamic differences between the two domains to obtain 2D and 3D spectra enabling the selection of the signals based on their nuclear relaxation properties. These experiments allowed the sequence-specific assignment of the TAZ4 construct to be extended revealing a crosstalk between the disordered region and the globular domain.
Collapse
Affiliation(s)
- Lorenzo Bracaglia
- Department of Chemistry "Ugo Schiff" and Magnetic Resonance Center, University of Florence, Sesto Fiorentino 50019, Italy
| | - Silvia Oliveti
- Department of Chemistry "Ugo Schiff" and Magnetic Resonance Center, University of Florence, Sesto Fiorentino 50019, Italy
| | - Isabella C Felli
- Department of Chemistry "Ugo Schiff" and Magnetic Resonance Center, University of Florence, Sesto Fiorentino 50019, Italy
| | - Roberta Pierattelli
- Department of Chemistry "Ugo Schiff" and Magnetic Resonance Center, University of Florence, Sesto Fiorentino 50019, Italy
| |
Collapse
|
2
|
Howe J, Barbar EJ. Dynamic interactions of dimeric hub proteins underlie their diverse functions and structures: A comparative analysis of 14-3-3 and LC8. J Biol Chem 2025; 301:108416. [PMID: 40107617 PMCID: PMC12017986 DOI: 10.1016/j.jbc.2025.108416] [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: 11/05/2024] [Revised: 02/06/2025] [Accepted: 02/08/2025] [Indexed: 03/22/2025] Open
Abstract
Hub proteins interact with a host of client proteins and regulate multiple cellular functions. Dynamic hubs have a single binding interface for one client at a time resulting in competition among clients with the highest affinity. Dynamic dimeric hubs with two identical sites bind either two different client proteins or two chains of the same client to form homogenous complexes and could also form heterogeneous mixtures of interconverting complexes. Here, we review the interactions of the dimeric hubs 14-3-3 and LC8. 14-3-3 is a phosphoserine/threonine binding protein involved in structuring client proteins and regulating their phosphorylation. LC8 is involved in promoting the dimerization of client peptides and the rigidification of their disordered regions. Both 14-3-3 and LC8 are essential genes, with 14-3-3 playing a crucial role in apoptosis and cell cycle regulation, while LC8 is critical for the assembly of proteins involved in transport, DNA repair, and transcription. Interestingly, both protein dimers can dissociate by phosphorylation, which results in their interactome-wide changes. Their interactions are also regulated by the phosphorylation of their clients. Both form heterogeneous complexes with various functions including phase separation, signaling, and viral hijacking where they restrict the conformational heterogeneity of their dimeric clients that bind nucleic acids. This comparative analysis highlights the importance of dynamic protein-protein interactions in the diversity of functions of 14-3-3 and LC8 and how small differences in structures of interfaces explain why 14-3-3 is primarily involved in the regulation of phosphorylation states while LC8 is primarily involved in the regulation of assembly of large dynamic complexes.
Collapse
Affiliation(s)
- Jesse Howe
- Oregon State University, Department of Biochemistry and Biophysics, Corvallis, Oregon, USA
| | - Elisar J Barbar
- Oregon State University, Department of Biochemistry and Biophysics, Corvallis, Oregon, USA.
| |
Collapse
|
3
|
Jiao P, Yang N, Jia Q, Fan B, Feng K, Yu J, Zhao S. A dual-reporter LDLR system integrating fluorescence and luminescence for understanding LDLR regulation and facilitating drug discovery. Front Mol Biosci 2025; 12:1552085. [PMID: 40182619 PMCID: PMC11966430 DOI: 10.3389/fmolb.2025.1552085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Accepted: 02/24/2025] [Indexed: 04/05/2025] Open
Abstract
Introduction The low-density lipoprotein receptor (LDLR) is integral to cholesterol metabolism and cardiovascular health. Enhancing LDLR expression is a promising strategy for treating hyperlipidemia and reducing the risk of atherosclerosis. However, current LDLR reporter systems have limitations in detecting both transcriptional and translational regulation. To address this, we developed a novel dual-reporter LDLR system incorporating Enhanced Green Fluorescent Protein (EGFP) and Gaussia luciferase (Gluc) to enable precise monitoring of LDLR expression and function. Methods A CRISPR/Cas9-mediated knock-in strategy was used to integrate EGFP and Gluc upstream of the stop codon located in exon 18 of the LDLR gene in HEK293 cells. The dual-reporter system allows real-time visualization of LDLR expression via EGFP fluorescence and quantitative assessment through secreted Gluc activity. The system was validated using western blotting, immunofluorescence, and functional assays, including DiI-LDL uptake and drug response analyses with statins and PCSK9 inhibitors. Results The established LDLR-EGFP-Gluc knock-in cell line faithfully recapitulates endogenous LDLR expression and function. EGFP fluorescence accurately reflects LDLR expression dynamics, while Gluc activity provides a highly sensitive and quantitative readout. Functional assays confirmed that LDLR expression responds appropriately to statins and PCSK9 inhibitors. Additionally, screening for transcriptional regulators identified FOXP3 and CREB as novel modulators of LDLR expression, with CREB-mediated regulation involving the sterol regulatory element-binding protein 2 (SREBP2) pathway. Discussion This dual-reporter system enables complementary monitoring of LDLR dynamics, providing enhanced sensitivity, accuracy, and versatility for studying LDLR regulation and function, as well as facilitating drug discovery targeting hyperlipidemia and cardiovascular diseases.
Collapse
Affiliation(s)
- Peng Jiao
- Department of Urology, Binzhou Medical University Hospital, Binzhou, Shandong, China
- Medical Integration and Practice Center, Shandong University, Jinan, Shandong, China
- Shandong Engineering Research Center of Molecular Medicine for Renal Diseases, Yantai, Shandong, China
- Department of Physiology, Binzhou Medical University, Yantai, Shandong, China
| | - Na Yang
- Shandong Engineering Research Center of Molecular Medicine for Renal Diseases, Yantai, Shandong, China
- Department of Physiology, Binzhou Medical University, Yantai, Shandong, China
| | - Qianfeng Jia
- Department of Urology, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, Shandong, China
| | - Baozhen Fan
- Department of Urology, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, Shandong, China
| | - Ke Feng
- Shandong Engineering Research Center of Molecular Medicine for Renal Diseases, Yantai, Shandong, China
- Department of Physiology, Binzhou Medical University, Yantai, Shandong, China
| | - Jian Yu
- Department of Basic Medical Education, Yantai Nursing School, Yantai, Shandong, China
| | - Shengtian Zhao
- Department of Urology, Binzhou Medical University Hospital, Binzhou, Shandong, China
- Shandong Provincial Engineering Laboratory of Urologic Tissue Reconstruction, Jinan, Shandong, China
- Department of Urology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| |
Collapse
|
4
|
Flores E, Camacho AR, Cuevas-Zepeda E, McCoy MB, Yu F, Staller MV, Sukenik S. Correlating disordered activation domain ensembles with gene expression levels. BIOPHYSICAL REPORTS 2025; 5:100195. [PMID: 39755236 PMCID: PMC11791265 DOI: 10.1016/j.bpr.2024.100195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Revised: 12/14/2024] [Accepted: 12/31/2024] [Indexed: 01/06/2025]
Abstract
Transcription factor proteins bind to specific DNA promoter sequences and initiate gene transcription. These proteins often contain intrinsically disordered activation domains (ADs) that regulate their transcriptional activity. Like other disordered protein regions, ADs do not have a fixed three-dimensional structure and instead exist in an ensemble of conformations. Disordered ensembles contain sequence-encoded structural preferences that are often linked to their function. We hypothesize that this link exists between the structural preferences of AD ensembles and their ability to induce gene expression. To test this, we measured the ensemble dimensions of two ADs, HIF-1α and CITED2, in live cells using fluorescence resonance energy transfer microscopy and correlated this structural information with their transcriptional activity. We find that mutations that expanded the ensemble of HIF-1α increased transcriptional activity, while compacting mutations reduced it, highlighting the critical role of structural plasticity in regulating HIF-1α function. Conversely, CITED2 showed no correlation between ensemble dimensions and activity. Our results highlight a possible link between AD ensemble dimensions and their transcriptional activity, with implications for transcriptional regulation and dysfunction.
Collapse
Affiliation(s)
- Eduardo Flores
- Department of Chemistry and Biochemistry, University of California, Merced, Merced, California
| | - Aleah R Camacho
- Department of Chemistry and Biochemistry, University of California, Merced, Merced, California
| | - Estefania Cuevas-Zepeda
- Department of Chemistry and Biochemistry, University of California, Merced, Merced, California
| | - Mary B McCoy
- Department of Chemistry and Biochemistry, University of California, Merced, Merced, California
| | - Feng Yu
- Department of Chemistry and Biochemistry, University of California, Merced, Merced, California; Molecular Biophysics and Integrated Bioimaging, Lawrence Berkeley National Laboratory, Berkeley, California
| | - Max V Staller
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, California; Center for Computational Biology, University of California, Berkeley, Berkeley, California; Chan Zuckerberg Biohub-San Francisco, San Francisco, California
| | - Shahar Sukenik
- Department of Chemistry and Biochemistry, University of California, Merced, Merced, California; Department of Chemistry, Syracuse University, Syracuse, New York.
| |
Collapse
|
5
|
Jemth P. Protein binding and folding through an evolutionary lens. Curr Opin Struct Biol 2025; 90:102980. [PMID: 39817990 DOI: 10.1016/j.sbi.2024.102980] [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: 09/17/2024] [Revised: 12/18/2024] [Accepted: 12/19/2024] [Indexed: 01/18/2025]
Abstract
Protein-protein associations are often mediated by an intrinsically disordered protein region interacting with a folded domain in a coupled binding and folding reaction. Classic physical organic chemistry approaches together with structural biology have shed light on mechanistic aspects of such reactions. Further insight into general principles may be obtained by interpreting the results through an evolutionary lens. This review attempts to provide an overview on how the analysis of binding and folding reactions can benefit from an evolutionary approach, and is aimed at protein scientists without a background in evolution. Evolution constantly reshapes existing proteins by sampling more or less fit variants. Most new variants are weeded out as generations and new species come and go over hundreds to hundreds of millions of years. The huge ongoing genome sequencing efforts have provided us with a snapshot of existing adapted fit-for-purpose protein homologs in thousands of different organisms. Comparison of present-day orthologs and paralogs highlights general principles of the evolution of coupled binding and folding reactions and demonstrate a great potential for evolution to operate on disordered regions and modulate affinity and specificity of the interactions.
Collapse
Affiliation(s)
- Per Jemth
- Department of Medical Biochemistry and Microbiology, Uppsala University, BMC, Box 582, SE-75123 Uppsala, Sweden.
| |
Collapse
|
6
|
Patalano SD, Fuxman Bass P, Fuxman Bass JI. Transcription factors in the development and treatment of immune disorders. Transcription 2025; 16:118-140. [PMID: 38100543 PMCID: PMC11970766 DOI: 10.1080/21541264.2023.2294623] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 12/05/2023] [Accepted: 12/08/2023] [Indexed: 12/17/2023] Open
Abstract
Immune function is highly controlled at the transcriptional level by the binding of transcription factors (TFs) to promoter and enhancer elements. Several TF families play major roles in immune gene expression, including NF-κB, STAT, IRF, AP-1, NRs, and NFAT, which trigger anti-pathogen responses, promote cell differentiation, and maintain immune system homeostasis. Aberrant expression, activation, or sequence of isoforms and variants of these TFs can result in autoimmune and inflammatory diseases as well as hematological and solid tumor cancers. For this reason, TFs have become attractive drug targets, even though most were previously deemed "undruggable" due to their lack of small molecule binding pockets and the presence of intrinsically disordered regions. However, several aspects of TF structure and function can be targeted for therapeutic intervention, such as ligand-binding domains, protein-protein interactions between TFs and with cofactors, TF-DNA binding, TF stability, upstream signaling pathways, and TF expression. In this review, we provide an overview of each of the important TF families, how they function in immunity, and some related diseases they are involved in. Additionally, we discuss the ways of targeting TFs with drugs along with recent research developments in these areas and their clinical applications, followed by the advantages and disadvantages of targeting TFs for the treatment of immune disorders.
Collapse
Affiliation(s)
- Samantha D. Patalano
- Biology Department, Boston University, Boston, MA, USA
- Molecular Biology, Cellular Biology and Biochemistry Program, Boston University, Boston, MA, USA
| | - Paula Fuxman Bass
- Facultad de Medicina, Universidad de Buenos Aires, Ciudad Autónoma de Buenos Aires, Argentina
| | - Juan I. Fuxman Bass
- Biology Department, Boston University, Boston, MA, USA
- Molecular Biology, Cellular Biology and Biochemistry Program, Boston University, Boston, MA, USA
- Bioinformatics Program, Boston University, Boston, MA, USA
| |
Collapse
|
7
|
Watanabe-Matsui M, Kadoya S, Segawa K, Shima H, Nakagawa T, Nagasawa Y, Hayashi S, Matsumoto M, Ikeda M, Muto A, Ochiai K, Nguyen LC, Doh-Ura K, Shirouzu M, Nakayama K, Murayama K, Igarashi K. Heme regulates protein interactions and phosphorylation of BACH2 intrinsically disordered region in humoral response. iScience 2025; 28:111529. [PMID: 39758820 PMCID: PMC11699347 DOI: 10.1016/j.isci.2024.111529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Revised: 09/03/2023] [Accepted: 12/02/2024] [Indexed: 01/07/2025] Open
Abstract
Heme is known to bind to the intrinsically disordered region (IDR) to regulate protein function. The binding of heme to the IDR of transcription factor BACH2 promotes plasma cell differentiation, but the molecular basis is unknown. Heme was found to increase BACH2 IDR interaction with TANK-binding kinase 1 (TBK1). TBK1 inactivated BACH2 by phosphorylation of its IDR, whereas BACH2 repressed TBK1 gene expression. BACH2 phosphorylation by TBK1 inhibited its interaction with the co-repressor NCOR1 and promoted plasma cell differentiation. Heme also induced BACH2 binding to ubiquitin E3 ligase adaptor FBXO22, which polyubiquitinated BACH2 only in the presence of heme in vitro. Mutations of some of the TBK1-mediated phosphorylation sites promoted BACH2-FBXO22 interaction, while additional mutations abrogated their interaction, suggesting that TBK1 can both inhibit and promote BACH2-FBXO22 interaction. Therefore, heme regulates phosphorylation of BACH2 IDR by TBK1 and its interaction with NCOR1 and FBXO22, leading to de-repression of BACH2 target genes in humoral immunity.
Collapse
Affiliation(s)
- Miki Watanabe-Matsui
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Sendai, Japan
- The Japan Society for the Promotion of Science (JSPS), Tokyo, Japan
| | - Shun Kadoya
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Kei Segawa
- Pharmaceutical Discovery Research Laboratories, Teijin Pharma Limited, Tokyo, Japan
| | - Hiroki Shima
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Tadashi Nakagawa
- Division of Cell Proliferation, ART, Tohoku University Graduate School of Medicine, Sendai, Japan
- Department of Clinical Pharmacology, Sanyo-Onoda City University, Sanyo-Onoda, Japan
| | - Yuko Nagasawa
- Division of Cell Proliferation, ART, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Shuichiro Hayashi
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Mitsuyo Matsumoto
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Mariko Ikeda
- Laboratory for Protein Functional and Structural Biology, RIKEN Center for Biosystems Dynamics Research, Yokohama, Japan
| | - Akihiko Muto
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Kyoko Ochiai
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Long C. Nguyen
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Katsumi Doh-Ura
- Department of Neurochemistry, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Mikako Shirouzu
- Laboratory for Protein Functional and Structural Biology, RIKEN Center for Biosystems Dynamics Research, Yokohama, Japan
| | - Keiko Nakayama
- Division of Cell Proliferation, ART, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Kazutaka Murayama
- Division of Biomedical Measurements and Diagnostics, Tohoku University Graduate School of Biomedical Engineering, Sendai, Japan
| | - Kazuhiko Igarashi
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Sendai, Japan
| |
Collapse
|
8
|
Abdelaziz HA, Hamed MF, Ghoniem HA, Nader MA, Suddek GM. Empagliflozin Mitigates PTZ-Induced Seizures in Rats: Modulating Npas4 and CREB-BDNF Signaling Pathway. J Neuroimmune Pharmacol 2025; 20:5. [PMID: 39776284 PMCID: PMC11706855 DOI: 10.1007/s11481-024-10162-6] [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: 03/12/2024] [Accepted: 11/29/2024] [Indexed: 01/11/2025]
Abstract
Empagliflozin (EMPA) is one of the sodium/glucose cotransporter 2 (SGLT2) inhibitors that has been recently approved for the treatment of diabetes mellitus type II. Recently, EMPA has shown protective effects in different neurological disorders, besides its antidiabetic activity. Kindling is a relevant model to study epilepsy and neuroplasticity. This study aimed to investigate the potential protective effects of EMPA (1 and 3 mg/kg orally) against convulsant effects induced by pentylenetetrazole (PTZ) using a modified window- (win-) PTZ kindling protocol. The biochemical dysfunction and hippocampal damage induced by PTZ were profoundly reversed by EMPA treatment in a dose-dependent manner, as evidenced by the significant increase in reduced glutathione (GSH) and decrease in malondialdehyde (MDA) hippocampal contents. Furthermore, EMPA counteracted PTZ-induced neuronal damage in the hippocampal region, as confirmed by histopathological examination of the hippocampal tissues. EMPA impaired astrocytosis and showed an antiapoptotic effect through a significant reduction of glial fibrillary acidic protein (GFAP) and BCL2-Associated X Protein (BAX) expressions, respectively. Interestingly, EMPA exhibited an antiepileptic effect against PTZ-induced seizures through significantly reducing neuronal PAS domain Protein 4 (Npas4), cyclic adenosine monophosphate (cAMP) response element binding protein (CREB) hippocampal expressions, and enhancing the brain-derived neurotrophic factor (BDNF)-tropomyosin receptor kinase B (TrkB) pathway, which are found to be involved in epileptogenesis, eventually leading to significant improvement of behavioral impairments induced by PTZ. Hence, these results showed further prospective insights for EMPA as a neuroprotective agent.
Collapse
Affiliation(s)
- Heba A Abdelaziz
- Pharmacology and Biochemistry Department, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa, 35712, Egypt
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Mansoura University, Mansoura, 35516, Egypt
| | - Mohamed F Hamed
- Pathology Department, Faculty of Veterinary Medicine, Mansoura University, Mansoura, 35516, Egypt
| | - Hamdy A Ghoniem
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Mansoura University, Mansoura, 35516, Egypt
| | - Manar A Nader
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Mansoura University, Mansoura, 35516, Egypt
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Mansoura National University, Gamasa, 7731168, Egypt
| | - Ghada M Suddek
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Mansoura University, Mansoura, 35516, Egypt.
| |
Collapse
|
9
|
Huang S, Zhao Y, Phillips S, Wilms B, He C, Wu L. Single-base m 6A epitranscriptomics reveals novel HIV-1 host interaction targets in primary CD4 + T cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.12.31.630958. [PMID: 39803509 PMCID: PMC11722377 DOI: 10.1101/2024.12.31.630958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/16/2025]
Abstract
N 6-methyladenosine (m6A) is the most prevalent cellular mRNA modification and plays a critical role in regulating RNA stability, localization, and gene expression. m6A modification plays a vital role in modulating the expression of viral and cellular genes during HIV-1 infection. HIV-1 infection increases cellular RNA m6A levels in many cell types, which facilitates HIV-1 replication and infectivity in target cells. However, the function of m6A modification in regulating HIV-1 infection of primary CD4+ T cells remains unclear. Here, we demonstrate that HIV-1 infection of Jurkat CD4+ T cells and primary CD4+ T cells promotes the interaction between the m6A writer complex subunits methyltransferase-like 3 and 14 (METTL3/METTL14). Using single-base m6A-specific RNA sequencing, we identified several differentially m6A-modified cellular mRNAs, including perilipin 3 (PLIN3), during HIV-1 infection in primary CD4+ T cells. Interestingly, HIV-1 infection increased PLIN3 mRNA level by enhancing its stability, but PLIN3 protein level was decreased. Knocking down PLIN3 in primary CD4+ T cells reduced HIV-1 production but enhanced virion infectivity. In contrast, in Jurkat cells, PLIN3 mRNA and protein expression levels were unaffected by HIV-1 infection, and knocking out PLIN3 did not impact HIV-1 production or infectivity. These results indicate that the interplay between HIV-1 and PLIN3 is cell-type specific and only observed in primary CD4+ T cells. Overall, our results highlight the importance of m6A RNA modification in HIV-1-infected primary CD4+ T cells and suggest its significance as a regulatory mechanism in HIV-1 infection.
Collapse
Affiliation(s)
- Siyu Huang
- Department of Microbiology and Immunology, Carver College of Medicine, The University of Iowa, Iowa City, Iowa, USA
| | - Yutao Zhao
- Department of Chemistry, Department of Biochemistry and Molecular Biology, Institute for Biophysical Dynamics, University of Chicago, Chicago, Illinois, USA
| | - Stacia Phillips
- Department of Microbiology and Immunology, Carver College of Medicine, The University of Iowa, Iowa City, Iowa, USA
| | - Bethany Wilms
- Department of Microbiology and Immunology, Carver College of Medicine, The University of Iowa, Iowa City, Iowa, USA
| | - Chuan He
- Department of Chemistry, Department of Biochemistry and Molecular Biology, Institute for Biophysical Dynamics, University of Chicago, Chicago, Illinois, USA
- Howard Hughes Medical Institute, University of Chicago, Chicago, Illinois, USA
| | - Li Wu
- Department of Microbiology and Immunology, Carver College of Medicine, The University of Iowa, Iowa City, Iowa, USA
| |
Collapse
|
10
|
Cho SE, Lee Y, Kim JI, Jung KY, Jeong H, Park HW, Lee BI. Comparative crystal structure analysis of the human EP300 and CBP KIX domains. Biochem Biophys Res Commun 2024; 741:151064. [PMID: 39612645 DOI: 10.1016/j.bbrc.2024.151064] [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: 11/15/2024] [Accepted: 11/23/2024] [Indexed: 12/01/2024]
Abstract
Small-cell lung cancer (SCLC) is highly lethal because the tumors grow and metastasize rapidly. Effective treatments for SCLC are lacking currently. A recent study demonstrated that the E1A binding protein P300 (EP300) KIX domain has pro-tumorigenic activity and is selectively involved in the development and growth of SCLC. These findings suggest the possibility of developing small-molecule inhibitors of EP300 KIX as new targeted therapies for SCLC. In this study, we reported the crystal structure of the human EP300 KIX domain at 2.9 Å resolution except for a flexible loop and C-terminal end. The overall structure was almost identical to that of the cAMP response element-binding protein (CBP) KIX. Nine EP300 KIX residues were different from those of CBP KIX. Among these non-strictly conserved residues, Ala627, which corresponds to Asp647 in CBP KIX, reduces the negative surface potential. Asn581 and Arg613 contributed to the formation of additional hydrogen bonds in the EP300 KIX structure. Further structural analysis revealed that the hydrophobic residues that form the allosteric network in CBP KIX were well conserved in the EP300 KIX structure. This study lays the groundwork for structure-based drug design for SCLC.
Collapse
Affiliation(s)
- Sang Eun Cho
- Research Institute, National Cancer Center, Goyang-si, Gyeonggi-do, 10408, Republic of Korea; Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Yuno Lee
- Drug Information Research Center, Korea Research Institute of Chemical Technology, Daejeon, 34114, Republic of Korea
| | - Ji-In Kim
- Therapeutics and Biotechnology Division, Korea Research Institute of Chemical Technology, Daejeon, 34114, Republic of Korea; College of Pharmacy, Chungbuk National University, Cheongju-si, Chungcheongbuk-do, 28160, Republic of Korea
| | - Kwan-Young Jung
- Therapeutics and Biotechnology Division, Korea Research Institute of Chemical Technology, Daejeon, 34114, Republic of Korea; Department of Medicinal Chemistry and Pharmacology, University of Science & Technology, Daejeon, 34113, Republic of Korea
| | - Hyeongseop Jeong
- Center for Research Equipment, Korea Basic Science Institute, Cheongju-si, Chungcheongbuk-do, 28119, Republic of Korea
| | - Hyun Woo Park
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Byung Il Lee
- Research Institute, National Cancer Center, Goyang-si, Gyeonggi-do, 10408, Republic of Korea; Department of Cancer Biomedical Science, National Cancer Center Graduate School of Cancer Science and Policy, Goyang-si, Gyeonggi-do, 10408, Republic of Korea.
| |
Collapse
|
11
|
Chatterjee D, Zhang Z, Lin PY, Wang PH, Sidhu GK, Yennawar NH, Hsieh JWA, Chen PY, Song R, Meyers BC, Chopra S. Maize unstable factor for orange1 encodes a nuclear protein that affects redox accumulation during kernel development. THE PLANT CELL 2024; 37:koae301. [PMID: 39589935 DOI: 10.1093/plcell/koae301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Accepted: 10/17/2024] [Indexed: 11/28/2024]
Abstract
The basal endosperm transfer layer (BETL) of the maize (Zea mays L.) kernel is composed of transfer cells for nutrient transport to nourish the developing kernel. To understand the spatiotemporal processes required for BETL development, we characterized 2 unstable factor for orange1 (Zmufo1) mutant alleles. The BETL defects in these mutants were associated with high levels of reactive oxygen species, oxidative DNA damage, and cell death. Interestingly, antioxidant supplementation in in vitro cultured kernels alleviated the cellular defects in mutants. Transcriptome analysis of the loss-of-function Zmufo1 allele showed differential expression of tricarboxylic acid cycle, redox homeostasis, and BETL-related genes. The basal endosperms of the mutant alleles had high levels of acetyl-CoA and elevated histone acetyltransferase activity. The BETL cell nuclei showed reduced electron-dense regions, indicating sparse heterochromatin distribution in the mutants compared with wild-type. Zmufo1 overexpression further reduced histone methylation marks in the enhancer and gene body regions of the pericarp color1 (Zmp1) reporter gene. Zmufo1 encodes an intrinsically disordered nuclear protein with very low sequence similarity to known proteins. Yeast two-hybrid and luciferase complementation assays established that ZmUFO1 interacts with proteins that play a role in chromatin remodeling, nuclear transport, and transcriptional regulation. This study establishes the critical function of Zmufo1 during basal endosperm development in maize kernels.
Collapse
Affiliation(s)
- Debamalya Chatterjee
- Department of Plant Science, The Pennsylvania State University, University Park, PA 16802, USA
| | - Ziru Zhang
- National Center for Maize Improvement, China Agricultural University, Beijing 100083, China
| | - Pei-Yu Lin
- Institute of Plant and Microbial Biology, Academia Sinica, Nankang, Taipei 11529, Taiwan
| | - Po-Hao Wang
- Department of Plant Science, The Pennsylvania State University, University Park, PA 16802, USA
| | - Gurpreet K Sidhu
- Department of Plant Science, The Pennsylvania State University, University Park, PA 16802, USA
| | - Neela H Yennawar
- X-Ray Crystallography Facility, The Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA 16802, USA
| | - Jo-Wei Allison Hsieh
- Institute of Plant and Microbial Biology, Academia Sinica, Nankang, Taipei 11529, Taiwan
- Genome and Systems Biology Degree Program, Academia Sinica and National Taiwan University, Taipei 10617, Taiwan
| | - Pao-Yang Chen
- Institute of Plant and Microbial Biology, Academia Sinica, Nankang, Taipei 11529, Taiwan
- Genome and Systems Biology Degree Program, Academia Sinica and National Taiwan University, Taipei 10617, Taiwan
| | - Rentao Song
- National Center for Maize Improvement, China Agricultural University, Beijing 100083, China
| | - Blake C Meyers
- The Donald Danforth Plant Science Center, St. Louis, MO 63132, USA
- Division of Plant Science and Technology, University of Missouri, Columbia, MO 65201, USA
| | - Surinder Chopra
- Department of Plant Science, The Pennsylvania State University, University Park, PA 16802, USA
| |
Collapse
|
12
|
LeBlanc C, Stefani J, Soriano M, Lam A, Zintel MA, Kotha SR, Chase E, Pimentel-Solorio G, Vunnum A, Flug K, Fultineer A, Hummel N, Staller MV. Conservation of function without conservation of amino acid sequence in intrinsically disordered transcriptional activation domains. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.03.626510. [PMID: 39677729 PMCID: PMC11642888 DOI: 10.1101/2024.12.03.626510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
Protein function is canonically believed to be more conserved than amino acid sequence, but this idea is only well supported in folded domains, where highly diverged sequences can fold into equivalent 3D structures. In contrast, intrinsically disordered protein regions (IDRs) do not fold into a stable 3D structure, thus it remains unknown when and how function is conserved for IDRs that experience rapid amino acid sequence divergence. As a model system for studying the evolution of IDRs, we examined transcriptional activation domains, the regions of transcription factors that bind to coactivator complexes. We systematically identified activation domains on 502 orthologs of the transcriptional activator Gcn4 spanning 600 MY of fungal evolution. We find that the central activation domain shows strong conservation of function without conservation of sequence. This conservation of function without conservation of sequence is facilitated by evolutionary turnover (gain and loss) of key acidic and aromatic residues, the positions most important for function. This high sequence flexibility of functional orthologs mirrors the physical flexibility of the activation domain coactivator interaction interface, suggesting that physical flexibility enables evolutionary plasticity. We propose that turnover of short functional elements, sometimes individual amino acids, is a general mechanism for conservation of function without conservation of sequence during IDR evolution.
Collapse
Affiliation(s)
- Claire LeBlanc
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, 94720
- Center for Computational Biology, University of California Berkeley, Berkeley, 94720
| | - Jordan Stefani
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, 94720
- Center for Computational Biology, University of California Berkeley, Berkeley, 94720
| | - Melvin Soriano
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, 94720
- Center for Computational Biology, University of California Berkeley, Berkeley, 94720
| | - Angelica Lam
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, 94720
- Center for Computational Biology, University of California Berkeley, Berkeley, 94720
| | - Marissa A. Zintel
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, 94720
| | - Sanjana R. Kotha
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, 94720
- Center for Computational Biology, University of California Berkeley, Berkeley, 94720
| | - Emily Chase
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, 94720
- Center for Computational Biology, University of California Berkeley, Berkeley, 94720
| | - Giovani Pimentel-Solorio
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, 94720
- Center for Computational Biology, University of California Berkeley, Berkeley, 94720
| | - Aditya Vunnum
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, 94720
| | - Katherine Flug
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, 94720
| | - Aaron Fultineer
- Department of Physics, University of California Berkeley, Berkeley, 94720
| | - Niklas Hummel
- Department of Biology, Technische Universität Darmstadt, Darmstadt, Germany
| | - Max V. Staller
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, 94720
- Center for Computational Biology, University of California Berkeley, Berkeley, 94720
- Chan Zuckerberg Biohub–San Francisco, San Francisco, CA 94158
| |
Collapse
|
13
|
Mo K, Wang Y, Lu C, Li Z. Insight into the role of macrophages in periodontitis restoration and development. Virulence 2024; 15:2427234. [PMID: 39535076 PMCID: PMC11572313 DOI: 10.1080/21505594.2024.2427234] [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: 01/15/2024] [Revised: 08/31/2024] [Accepted: 10/31/2024] [Indexed: 11/16/2024] Open
Abstract
Periodontitis is one of the chronic diseases that have the greatest impact on human health, and it is associated with several other chronic diseases. Tissue damage associated with periodontitis is often connected with immune response. Immune cells are a crucial component of the human immune system and are directly involved in periodontitis during the inflammatory phase of the disease. Macrophages, as a key component of the immune system, are responsible for defence, antigen presentation and phagocytosis in healthy tissue. They are also closely linked to the development and resolution of periodontitis, through mechanisms such as macrophage polarization, pattern recognition receptors recognition, efferocytosis, and Specialized Pro-resolving Mediators (SPMs) production. Additionally, apoptosis and autophagy are also known to play a role in the recovery of periodontitis. This review aims to investigate the aforementioned mechanisms in more detail and identify novel therapeutic approaches for periodontitis.
Collapse
Affiliation(s)
- Keyin Mo
- School of Stomatology, Jinan University, Guangzhou, China
| | - Yijue Wang
- West China School of Stomatology, Sichuan University, Chengdu, China
| | - Chunting Lu
- Science and Education Office, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, China
| | - Zejian Li
- Hospital of Stomatology, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, China
| |
Collapse
|
14
|
Llewellyn J, Baratam R, Culig L, Beerman I. Cellular stress and epigenetic regulation in adult stem cells. Life Sci Alliance 2024; 7:e202302083. [PMID: 39348938 PMCID: PMC11443024 DOI: 10.26508/lsa.202302083] [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: 04/07/2023] [Revised: 09/16/2024] [Accepted: 09/16/2024] [Indexed: 10/02/2024] Open
Abstract
Stem cells are a unique class of cells that possess the ability to differentiate and self-renew, enabling them to repair and replenish tissues. To protect and maintain the potential of stem cells, the cells and the environment surrounding these cells (stem cell niche) are highly responsive and tightly regulated. However, various stresses can affect the stem cells and their niches. These stresses are both systemic and cellular and can arise from intrinsic or extrinsic factors which would have strong implications on overall aging and certain disease states. Therefore, understanding the breadth of drivers, namely epigenetic alterations, involved in cellular stress is important for the development of interventions aimed at maintaining healthy stem cells and tissue homeostasis. In this review, we summarize published findings of epigenetic responses to replicative, oxidative, mechanical, and inflammatory stress on various types of adult stem cells.
Collapse
Affiliation(s)
- Joey Llewellyn
- Epigenetics and Stem Cell Unit, Translational Gerontology Branch, National Institute on Aging, Baltimore, MD, USA
| | - Rithvik Baratam
- Epigenetics and Stem Cell Unit, Translational Gerontology Branch, National Institute on Aging, Baltimore, MD, USA
| | - Luka Culig
- Epigenetics and Stem Cell Unit, Translational Gerontology Branch, National Institute on Aging, Baltimore, MD, USA
| | - Isabel Beerman
- Epigenetics and Stem Cell Unit, Translational Gerontology Branch, National Institute on Aging, Baltimore, MD, USA
| |
Collapse
|
15
|
Tandon S, Sarkar S. Myc functions downstream of InR and their concurrent upregulation additively restricts pathogenesis of human poly(Q) disorders in Drosophila disease models. Int J Biochem Cell Biol 2024; 177:106690. [PMID: 39521038 DOI: 10.1016/j.biocel.2024.106690] [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: 05/30/2024] [Revised: 11/05/2024] [Accepted: 11/07/2024] [Indexed: 11/16/2024]
Abstract
Human polyglutamine [poly(Q)] disorders are caused by abnormal expansion of CAG repeats in one gene (disease specific), yet a plethora of cellular pathways are found to be involved in their pathogenesis and progression. Despite the tremendous effort, all pursuits for the development of intervention therapy against these disorders seem futile. Recent reports suggest combination therapy as a potential strategy to combat the complex pathogenesis of such neurodegenerative disorders. The present study attempted to identify a combinatorial intervention strategy against human poly(Q) disorders in Drosophila disease models. Due to its immense potential to be stimulated by drugs, the evolutionarily conserved insulin signalling cascade which is well-established modifier of human poly(Q) pathogenesis was selected for the study. Genetic screening studies identified Drosophila Myc as a potential partner of insulin receptor (InR) that conferred additive rescue against poly(Q) induced neurodegeneration. Comprehensive analyses demonstrated InR and Myc to confer additive rescue against several events of pathogenesis, including aggregation of expanded poly(Q) containing proteins, transcriptional dysregulation, upsurge of cell death cascades, etc. Also, the synergistic rescue efficiency of InR and Myc was equally efficient in mitigating poly(Q) induced structural and functional deficits. The study also demonstrates that Myc functions downstream of InR signalling cascade to deliver rescue against human poly(Q) mediated toxicity in Drosophila disease models. In conclusion, the present study suggests that InR and Myc have the potential to be developed as a combinatorial therapeutic approach against human poly(Q) diseases.
Collapse
Affiliation(s)
- Shweta Tandon
- Department of Genetics, University of Delhi South Campus, Benito Juarez Road, New Delhi 110021, India
| | - Surajit Sarkar
- Department of Genetics, University of Delhi South Campus, Benito Juarez Road, New Delhi 110021, India.
| |
Collapse
|
16
|
Flores E, Camacho AR, Cuevas-Zepeda E, McCoy MB, Yu F, Staller MV, Sukenik S. Correlating Disordered Activation Domain Ensembles with Gene Expression Levels. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.19.619222. [PMID: 39484498 PMCID: PMC11527027 DOI: 10.1101/2024.10.19.619222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Transcription factor proteins bind to specific DNA promoter sequences and initiate gene transcription. In eukaryotes, most transcription factors contain intrinsically disordered activation domains (ADs) that regulate their transcriptional activity. Like other disordered protein regions, ADs do not have a fixed three-dimensional structure and instead exist in an ensemble of conformations. Disordered ensembles contain sequence-encoded structural preferences which are often linked to their function. We hypothesize this link exists between the structural preferences of disordered AD ensembles and their ability to induce gene expression. To test this, we used FRET microscopy to measure the ensemble dimensions of two activation domains, HIF-1α and CITED2, in live cells, and correlate this structural information with transcriptional activity. We find that point mutations that expanded the HIF-1α ensemble increased transcriptional activity, while those that compacted it reduced activity. Conversely, CITED2 showed no correlation between ensemble dimensions and activity. Our results reveal a sequence-dependent relationship between AD ensemble dimensions and their transcriptional activity.
Collapse
Affiliation(s)
- Eduardo Flores
- Department of Chemistry and Biochemistry, University of California Merced, Merced, 95343
| | - Aleah R Camacho
- Department of Chemistry and Biochemistry, University of California Merced, Merced, 95343
| | | | - Mary B McCoy
- Department of Chemistry and Biochemistry, University of California Merced, Merced, 95343
| | - Feng Yu
- Department of Chemistry and Biochemistry, University of California Merced, Merced, 95343
- Molecular Biophysics and Integrated Bioimaging, Lawrence Berkeley National Laboratory, 94720 Berkeley, CA, USA
| | - Max V Staller
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, 94720
- Center for Computational Biology, University of California Berkeley, Berkeley, 94720
- Chan Zuckerberg Biohub-San Francisco, San Francisco, CA 94158
| | - Shahar Sukenik
- Department of Chemistry and Biochemistry, University of California Merced, Merced, 95343
- Department of Chemistry, Syracuse University, Syracuse, 13244
| |
Collapse
|
17
|
Inge M, Miller R, Hook H, Bray D, Keenan J, Zhao R, Gilmore T, Siggers T. Rapid profiling of transcription factor-cofactor interaction networks reveals principles of epigenetic regulation. Nucleic Acids Res 2024; 52:10276-10296. [PMID: 39166482 PMCID: PMC11417405 DOI: 10.1093/nar/gkae706] [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: 04/16/2024] [Revised: 06/14/2024] [Accepted: 08/19/2024] [Indexed: 08/23/2024] Open
Abstract
Transcription factor (TF)-cofactor (COF) interactions define dynamic, cell-specific networks that govern gene expression; however, these networks are understudied due to a lack of methods for high-throughput profiling of DNA-bound TF-COF complexes. Here, we describe the Cofactor Recruitment (CoRec) method for rapid profiling of cell-specific TF-COF complexes. We define a lysine acetyltransferase (KAT)-TF network in resting and stimulated T cells. We find promiscuous recruitment of KATs for many TFs and that 35% of KAT-TF interactions are condition specific. KAT-TF interactions identify NF-κB as a primary regulator of acutely induced histone 3 lysine 27 acetylation (H3K27ac). Finally, we find that heterotypic clustering of CBP/P300-recruiting TFs is a strong predictor of total promoter H3K27ac. Our data support clustering of TF sites that broadly recruit KATs as a mechanism for widespread co-occurring histone acetylation marks. CoRec can be readily applied to different cell systems and provides a powerful approach to define TF-COF networks impacting chromatin state and gene regulation.
Collapse
Affiliation(s)
- Melissa M Inge
- Department of Biology, Boston University, Boston, MA 02215, USA
- Biological Design Center, Boston University, Boston, MA 02215, USA
| | - Rebekah Miller
- Department of Biology, Boston University, Boston, MA 02215, USA
- Biological Design Center, Boston University, Boston, MA 02215, USA
- Bioinformatics Program, Boston University, Boston, MA 02215, USA
| | - Heather Hook
- Department of Biology, Boston University, Boston, MA 02215, USA
| | - David Bray
- Department of Biology, Boston University, Boston, MA 02215, USA
- Bioinformatics Program, Boston University, Boston, MA 02215, USA
| | - Jessica L Keenan
- Department of Biology, Boston University, Boston, MA 02215, USA
- Bioinformatics Program, Boston University, Boston, MA 02215, USA
| | - Rose Zhao
- Department of Biology, Boston University, Boston, MA 02215, USA
| | | | - Trevor Siggers
- Department of Biology, Boston University, Boston, MA 02215, USA
- Biological Design Center, Boston University, Boston, MA 02215, USA
- Bioinformatics Program, Boston University, Boston, MA 02215, USA
| |
Collapse
|
18
|
Karunatilleke NC, Brickenden A, Choy WY. Molecular basis of the interactions between the disordered Neh4 and Neh5 domains of Nrf2 and CBP/p300 in oxidative stress response. Protein Sci 2024; 33:e5137. [PMID: 39150085 PMCID: PMC11328122 DOI: 10.1002/pro.5137] [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: 04/26/2024] [Revised: 06/21/2024] [Accepted: 07/22/2024] [Indexed: 08/17/2024]
Abstract
Nuclear factor erythroid 2-related factor 2 (Nrf2) is a major transcription factor that functions in maintaining redox homeostasis in cells. It mediates the transcription of cytoprotective genes in response to environmental and endogenous stresses to prevent oxidative damage. Thus, Nrf2 plays a significant role in chemoprevention. However, aberrant activation of Nrf2 has been shown to protect cancer cells from apoptosis and contribute to their chemoresistance. The interaction between Nrf2 and CBP is critical for the gene transcription activation. CBP and its homologue p300 interact with two transactivation domains in Nrf2, Neh4, and Neh5 domains through their TAZ1 and TAZ2 domains. To date, the molecular basis of this crucial interaction is not known, hindering a more detailed understanding of the regulation of Nrf2. To close this knowledge gap, we have used a set of biophysical experiments to dissect the Nrf2-CBP/p300 interactions. Structural properties of Neh4 and Neh5 and their binding with the TAZ1 and TAZ2 domains of CBP/p300 were characterized. Our results show that the Neh4 and Neh5 domains of Nrf2 are intrinsically disordered, and they both can bind the TAZ1 and TAZ2 domains of CBP/p300 with micromolar affinities. The findings provide molecular insight into the regulation of Nrf2 by CBP/p300 through multi-domain interactions.
Collapse
Affiliation(s)
- Nadun C Karunatilleke
- Department of Biochemistry, The University of Western Ontario, London, Ontario, Canada
| | - Anne Brickenden
- Department of Biochemistry, The University of Western Ontario, London, Ontario, Canada
| | - Wing-Yiu Choy
- Department of Biochemistry, The University of Western Ontario, London, Ontario, Canada
| |
Collapse
|
19
|
DelRosso N, Suzuki PH, Griffith D, Lotthammer JM, Novak B, Kocalar S, Sheth MU, Holehouse AS, Bintu L, Fordyce P. High-throughput affinity measurements of direct interactions between activation domains and co-activators. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.19.608698. [PMID: 39229005 PMCID: PMC11370418 DOI: 10.1101/2024.08.19.608698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
Sequence-specific activation by transcription factors is essential for gene regulation1,2. Key to this are activation domains, which often fall within disordered regions of transcription factors3,4 and recruit co-activators to initiate transcription5. These interactions are difficult to characterize via most experimental techniques because they are typically weak and transient6,7. Consequently, we know very little about whether these interactions are promiscuous or specific, the mechanisms of binding, and how these interactions tune the strength of gene activation. To address these questions, we developed a microfluidic platform for expression and purification of hundreds of activation domains in parallel followed by direct measurement of co-activator binding affinities (STAMMPPING, for Simultaneous Trapping of Affinity Measurements via a Microfluidic Protein-Protein INteraction Generator). By applying STAMMPPING to quantify direct interactions between eight co-activators and 204 human activation domains (>1,500 K ds), we provide the first quantitative map of these interactions and reveal 334 novel binding pairs. We find that the metazoan-specific co-activator P300 directly binds >100 activation domains, potentially explaining its widespread recruitment across the genome to influence transcriptional activation. Despite sharing similar molecular properties (e.g. enrichment of negative and hydrophobic residues), activation domains utilize distinct biophysical properties to recruit certain co-activator domains. Co-activator domain affinity and occupancy are well-predicted by analytical models that account for multivalency, and in vitro affinities quantitatively predict activation in cells with an ultrasensitive response. Not only do our results demonstrate the ability to measure affinities between even weak protein-protein interactions in high throughput, but they also provide a necessary resource of over 1,500 activation domain/co-activator affinities which lays the foundation for understanding the molecular basis of transcriptional activation.
Collapse
Affiliation(s)
| | - Peter H Suzuki
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Daniel Griffith
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, MO, USA
- Center for Biomolecular Condensates, Washington University in St. Louis, St. Louis, MO, USA
| | - Jeffrey M Lotthammer
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, MO, USA
- Center for Biomolecular Condensates, Washington University in St. Louis, St. Louis, MO, USA
| | - Borna Novak
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, MO, USA
- Center for Biomolecular Condensates, Washington University in St. Louis, St. Louis, MO, USA
| | - Selin Kocalar
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Maya U Sheth
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Alex S Holehouse
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, MO, USA
- Center for Biomolecular Condensates, Washington University in St. Louis, St. Louis, MO, USA
| | - Lacramioara Bintu
- Biophysics Program, Stanford University, Stanford, CA, USA
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Polly Fordyce
- Biophysics Program, Stanford University, Stanford, CA, USA
- Department of Bioengineering, Stanford University, Stanford, CA, USA
- Sarafan ChEM-H Institute, Stanford University, Stanford, CA, USA
- Chan Zuckerberg Biohub San Francisco, CA, USA
| |
Collapse
|
20
|
Chen K, Wang L, Yu Z, Yu J, Ren Y, Wang Q, Xu Y. Structure of the human TIP60 complex. Nat Commun 2024; 15:7092. [PMID: 39154037 PMCID: PMC11330486 DOI: 10.1038/s41467-024-51259-z] [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: 01/08/2024] [Accepted: 08/02/2024] [Indexed: 08/19/2024] Open
Abstract
Mammalian TIP60 is a multi-functional enzyme with histone acetylation and histone dimer exchange activities. It plays roles in diverse cellular processes including transcription, DNA repair, cell cycle control, and embryonic development. Here we report the cryo-electron microscopy structures of the human TIP60 complex with the core subcomplex and TRRAP module refined to 3.2-Å resolution. The structures show that EP400 acts as a backbone integrating the motor module, the ARP module, and the TRRAP module. The RUVBL1-RUVBL2 hexamer serves as a rigid core for the assembly of EP400 ATPase and YL1 in the motor module. In the ARP module, an ACTL6A-ACTB heterodimer and an extra ACTL6A make hydrophobic contacts with EP400 HSA helix, buttressed by network interactions among DMAP1, EPC1, and EP400. The ARP module stably associates with the motor module but is flexibly tethered to the TRRAP module, exhibiting a unique feature of human TIP60. The architecture of the nucleosome-bound human TIP60 reveals an unengaged nucleosome that is located between the core subcomplex and the TRRAP module. Our work illustrates the molecular architecture of human TIP60 and provides architectural insights into how this complex is bound by the nucleosome.
Collapse
Affiliation(s)
- Ke Chen
- Fudan University Shanghai Cancer Center, Institutes of Biomedical Sciences, State Key Laboratory of Genetic Engineering and Shanghai Key Laboratory of Medical Epigenetics, Shanghai Medical College of Fudan University, Shanghai, 200032, China
| | - Li Wang
- Fudan University Shanghai Cancer Center, Institutes of Biomedical Sciences, State Key Laboratory of Genetic Engineering and Shanghai Key Laboratory of Medical Epigenetics, Shanghai Medical College of Fudan University, Shanghai, 200032, China.
| | - Zishuo Yu
- Fudan University Shanghai Cancer Center, Institutes of Biomedical Sciences, State Key Laboratory of Genetic Engineering and Shanghai Key Laboratory of Medical Epigenetics, Shanghai Medical College of Fudan University, Shanghai, 200032, China
| | - Jiali Yu
- Fudan University Shanghai Cancer Center, Institutes of Biomedical Sciences, State Key Laboratory of Genetic Engineering and Shanghai Key Laboratory of Medical Epigenetics, Shanghai Medical College of Fudan University, Shanghai, 200032, China
| | - Yulei Ren
- Fudan University Shanghai Cancer Center, Institutes of Biomedical Sciences, State Key Laboratory of Genetic Engineering and Shanghai Key Laboratory of Medical Epigenetics, Shanghai Medical College of Fudan University, Shanghai, 200032, China
| | - Qianmin Wang
- Fudan University Shanghai Cancer Center, Institutes of Biomedical Sciences, State Key Laboratory of Genetic Engineering and Shanghai Key Laboratory of Medical Epigenetics, Shanghai Medical College of Fudan University, Shanghai, 200032, China.
| | - Yanhui Xu
- Fudan University Shanghai Cancer Center, Institutes of Biomedical Sciences, State Key Laboratory of Genetic Engineering and Shanghai Key Laboratory of Medical Epigenetics, Shanghai Medical College of Fudan University, Shanghai, 200032, China.
- The International Co-laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, China, Department of Systems Biology for Medicine, School of Basic Medical Sciences, Shanghai Medical College of Fudan University, Shanghai, 200032, China.
| |
Collapse
|
21
|
Lackner A, Cabral JE, Qiu Y, Zhou H, Leonidas L, Pham MA, Macapagal A, Lin S, Armanus E, McNulty R. Small molecule inhibitor binds to NOD-like receptor family pyrin domain containing 3 and prevents inflammasome activation. iScience 2024; 27:110459. [PMID: 39104412 PMCID: PMC11298654 DOI: 10.1016/j.isci.2024.110459] [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: 02/02/2024] [Revised: 05/10/2024] [Accepted: 07/02/2024] [Indexed: 08/07/2024] Open
Abstract
Despite recent advances in the mechanism of oxidized DNA activating NLRP3, the molecular mechanism and consequence of oxidized DNA associating with NLRP3 remains unknown. Cytosolic NLRP3 binds oxidized DNA which has been released from the mitochondria, which subsequently triggers inflammasome activation. Human glycosylase (hOGG1) repairs oxidized DNA damage which inhibits inflammasome activation. The fold of NLRP3 pyrin domain contains amino acids and a protein fold similar to hOGG1. Amino acids that enable hOGG1 to bind and cleave oxidized DNA are conserved in NLRP3. We found NLRP3 could bind and cleave oxidized guanine within mitochondrial DNA. The binding of oxidized DNA to NLRP3 was prevented by small molecule drugs which also inhibit hOGG1. These same drugs also inhibited inflammasome activation. Elucidating this mechanism will enable the design of drug memetics that treat inflammasome pathologies, illustrated herein by NLRP3 pyrin domain inhibitors which suppressed interleukin-1β (IL-1β) production in macrophages.
Collapse
Affiliation(s)
- Angela Lackner
- Laboratory of Macromolecular Structure, Department of Molecular Biology and Biochemistry, Charlie Dunlop School of Biological Sciences, University of California, Irvine, Steinhaus Hall, Irvine, CA 92694-3900, USA
| | - Julia Elise Cabral
- Laboratory of Macromolecular Structure, Department of Molecular Biology and Biochemistry, Charlie Dunlop School of Biological Sciences, University of California, Irvine, Steinhaus Hall, Irvine, CA 92694-3900, USA
| | - Yanfei Qiu
- Laboratory of Macromolecular Structure, Department of Molecular Biology and Biochemistry, Charlie Dunlop School of Biological Sciences, University of California, Irvine, Steinhaus Hall, Irvine, CA 92694-3900, USA
| | - Haitian Zhou
- Laboratory of Macromolecular Structure, Department of Molecular Biology and Biochemistry, Charlie Dunlop School of Biological Sciences, University of California, Irvine, Steinhaus Hall, Irvine, CA 92694-3900, USA
| | - Lemuel Leonidas
- Laboratory of Macromolecular Structure, Department of Molecular Biology and Biochemistry, Charlie Dunlop School of Biological Sciences, University of California, Irvine, Steinhaus Hall, Irvine, CA 92694-3900, USA
| | - Minh Anh Pham
- Laboratory of Macromolecular Structure, Department of Molecular Biology and Biochemistry, Charlie Dunlop School of Biological Sciences, University of California, Irvine, Steinhaus Hall, Irvine, CA 92694-3900, USA
| | - Alijah Macapagal
- Laboratory of Macromolecular Structure, Department of Molecular Biology and Biochemistry, Charlie Dunlop School of Biological Sciences, University of California, Irvine, Steinhaus Hall, Irvine, CA 92694-3900, USA
| | - Sophia Lin
- Laboratory of Macromolecular Structure, Department of Molecular Biology and Biochemistry, Charlie Dunlop School of Biological Sciences, University of California, Irvine, Steinhaus Hall, Irvine, CA 92694-3900, USA
| | - Emy Armanus
- Laboratory of Macromolecular Structure, Department of Molecular Biology and Biochemistry, Charlie Dunlop School of Biological Sciences, University of California, Irvine, Steinhaus Hall, Irvine, CA 92694-3900, USA
| | - Reginald McNulty
- Laboratory of Macromolecular Structure, Department of Molecular Biology and Biochemistry, Charlie Dunlop School of Biological Sciences, University of California, Irvine, Steinhaus Hall, Irvine, CA 92694-3900, USA
- Department of Pharmaceutical Sciences, University of California, Irvine, Steinhaus Hall, Irvine, CA 92694-3900, USA
| |
Collapse
|
22
|
McIvor JAP, Larsen DS, Mercadante D. Charge Relaying within a Phospho-Motif Rescue Binding Competency of a Disordered Transcription Factor. J Chem Inf Model 2024; 64:6041-6052. [PMID: 39074869 DOI: 10.1021/acs.jcim.4c00286] [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
Structural disorder in proteins is central to cellular signaling, where conformational plasticity equips molecules to promiscuously interact with different partners. By engaging with multiple binding partners via the rearrangement of its three helices, the nuclear coactivator binding domain (NCBD) of the CBP/p300 transcription factor is a paradigmatic example of promiscuity. Recently, molecular simulations and experiments revealed that, through the establishment of long-range electrostatic interactions, intended as salt-bridges formed between the post-translationally inserted phosphate and positively charged residues in helix H3 of NCBD, phosphorylation triggers NCBD compaction, lowering its affinity for binding partners. By means of extensive molecular simulations, we here investigated the effect of short-range electrostatics on the conformational ensemble of NCBD, by monitoring the interactions between a phosphorylated serine and conserved positively charged residues within the NCBD phospho-motif. We found that empowering proximal electrostatic interactions, as opposed to long-range electrostatics, can reshape the NCBD ensemble rescuing the binding competency of phosphorylated NCBD. Given the conservation of positive charges in phospho-motifs, proximal electrostatic interactions might dampen the effects of phosphorylation and act as a relay to regulate phosphorylated intrinsically disordered proteins, ultimately tuning the binding affinity for different cellular partners.
Collapse
Affiliation(s)
- Jordan A P McIvor
- School of Chemical Sciences, The University of Auckland, 23 Symonds Street, Auckland 1010, New Zealand
| | - Danaé S Larsen
- School of Chemical Sciences, The University of Auckland, 23 Symonds Street, Auckland 1010, New Zealand
| | - Davide Mercadante
- School of Chemical Sciences, The University of Auckland, 23 Symonds Street, Auckland 1010, New Zealand
| |
Collapse
|
23
|
Morffy N, Van den Broeck L, Miller C, Emenecker RJ, Bryant JA, Lee TM, Sageman-Furnas K, Wilkinson EG, Pathak S, Kotha SR, Lam A, Mahatma S, Pande V, Waoo A, Wright RC, Holehouse AS, Staller MV, Sozzani R, Strader LC. Identification of plant transcriptional activation domains. Nature 2024; 632:166-173. [PMID: 39020176 PMCID: PMC11589624 DOI: 10.1038/s41586-024-07707-3] [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: 06/26/2023] [Accepted: 06/12/2024] [Indexed: 07/19/2024]
Abstract
Gene expression in Arabidopsis is regulated by more than 1,900 transcription factors (TFs), which have been identified genome-wide by the presence of well-conserved DNA-binding domains. Activator TFs contain activation domains (ADs) that recruit coactivator complexes; however, for nearly all Arabidopsis TFs, we lack knowledge about the presence, location and transcriptional strength of their ADs1. To address this gap, here we use a yeast library approach to experimentally identify Arabidopsis ADs on a proteome-wide scale, and find that more than half of the Arabidopsis TFs contain an AD. We annotate 1,553 ADs, the vast majority of which are, to our knowledge, previously unknown. Using the dataset generated, we develop a neural network to accurately predict ADs and to identify sequence features that are necessary to recruit coactivator complexes. We uncover six distinct combinations of sequence features that result in activation activity, providing a framework to interrogate the subfunctionalization of ADs. Furthermore, we identify ADs in the ancient AUXIN RESPONSE FACTOR family of TFs, revealing that AD positioning is conserved in distinct clades. Our findings provide a deep resource for understanding transcriptional activation, a framework for examining function in intrinsically disordered regions and a predictive model of ADs.
Collapse
Affiliation(s)
| | - Lisa Van den Broeck
- Department of Plant and Microbial Biology, North Carolina State University, Raleigh, NC, USA
| | - Caelan Miller
- Department of Biology, Duke University, Durham, NC, USA
| | - Ryan J Emenecker
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, MO, USA
- Center for Biomolecular Condensates, Washington University in St. Louis, St. Louis, MO, USA
| | - John A Bryant
- Biological Systems Engineering, Virginia Tech, Blacksburg, VA, USA
| | - Tyler M Lee
- Department of Biology, Duke University, Durham, NC, USA
| | | | | | - Sunita Pathak
- Department of Biology, Duke University, Durham, NC, USA
| | - Sanjana R Kotha
- Center for Computational Biology, University of California, Berkeley, Berkeley, CA, USA
| | - Angelica Lam
- Center for Computational Biology, University of California, Berkeley, Berkeley, CA, USA
| | - Saloni Mahatma
- Department of Plant and Microbial Biology, North Carolina State University, Raleigh, NC, USA
| | - Vikram Pande
- Department of Plant and Microbial Biology, North Carolina State University, Raleigh, NC, USA
| | - Aman Waoo
- Department of Plant and Microbial Biology, North Carolina State University, Raleigh, NC, USA
| | - R Clay Wright
- Biological Systems Engineering, Virginia Tech, Blacksburg, VA, USA
| | - Alex S Holehouse
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, MO, USA
- Center for Biomolecular Condensates, Washington University in St. Louis, St. Louis, MO, USA
| | - Max V Staller
- Center for Computational Biology, University of California, Berkeley, Berkeley, CA, USA
| | - Rosangela Sozzani
- Department of Plant and Microbial Biology, North Carolina State University, Raleigh, NC, USA
| | | |
Collapse
|
24
|
Liang Z, Hu S, Dong Y, Miao L, Zhu W, Feng B, Fu J, Luo M, Wang L, Dong Z. Molecular characterization and function of hif1a and fih1 in response to acute thermal stress in American shad (Alosa sapidissima). FISH PHYSIOLOGY AND BIOCHEMISTRY 2024; 50:1563-1581. [PMID: 38789648 DOI: 10.1007/s10695-024-01356-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 05/03/2024] [Indexed: 05/26/2024]
Abstract
In order to evaluate the function of hypoxia-inducible factor-1 alpha (hif1α) and factor inhibiting hif1α (fih1) in response to thermal stress, we first conducted a functional analysis of A. sapidissima hif1α and fih1, and determined hif1α and fih1 expressions in different tissues in response to thermal stress based on identified housekeeping genes (HKGs). The results showed that hif1α and fih1 were mainly located in the nucleus and cytoplasm. The full length cDNA sequence of hif1α and fih1 was 4073 bp and 2759 bp, respectively. The cDNA sequence of hif1α includes 15 exons encoding 750 amino acid residues, and the full length cDNA sequence of fih1 contains 9 exons encoding 354 amino acid residues. During the acute thermal stress transferring from 16 ± 0.5 °C (control) to 20 ± 0.5 °C, 25 ± 0.5 °C, and 30 ± 0.5 °C for 15 min, it was found that the expression trends of hif1α and fih1 showed an inhibitory regulation in the heart, while they consistently expressed in brain, intestine, muscle, gill, kidney and liver. In conclusion, this is the first study to identify the tissue-specific HKGs in A. sapidissima and found that ef1α and β-actin are the most suitable HKGs. Hif1α and Fih1 are mainly the nuclear and cytoplasmic proteins, respectively, having high levels in the heart and brain. Alosa sapidissima countered a temperature increase from 16 to 25 ℃ by regulating the expressions of hif1α and fih1, but their physiological regulatory functions were unable to cope with acute thermal stress when the temperature difference was 14 ℃ (from 16 to 30 ℃).
Collapse
Affiliation(s)
- Zhengyuan Liang
- Wuxi Fisheries College, Nanjing Agricultural University, No.9 East Shanshui Road, Wuxi Jiangsu, 214081, China
- Wuxi Raysun Fishery Science and Technology Company, Xingyuan North Road No. 401, P.O. Box D20-501, Wuxi Jiangsu, 214000, China
| | - Songqin Hu
- Wuxi Fisheries College, Nanjing Agricultural University, No.9 East Shanshui Road, Wuxi Jiangsu, 214081, China
| | - Yalun Dong
- Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture and Rural Affairs; Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi Jiangsu, 214081, China
| | - Linghong Miao
- Wuxi Fisheries College, Nanjing Agricultural University, No.9 East Shanshui Road, Wuxi Jiangsu, 214081, China
- Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture and Rural Affairs; Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi Jiangsu, 214081, China
| | - Wenbin Zhu
- Wuxi Fisheries College, Nanjing Agricultural University, No.9 East Shanshui Road, Wuxi Jiangsu, 214081, China
- Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture and Rural Affairs; Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi Jiangsu, 214081, China
| | - Bingbing Feng
- Fisheries Technology Extension Center of Jiangsu Province, Hanzhongmen Street No. 300, Nanjing Jiangsu, 210036, China
| | - Jianjun Fu
- Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture and Rural Affairs; Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi Jiangsu, 214081, China
| | - Mingkun Luo
- Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture and Rural Affairs; Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi Jiangsu, 214081, China
| | - Lanmei Wang
- Wuxi Fisheries College, Nanjing Agricultural University, No.9 East Shanshui Road, Wuxi Jiangsu, 214081, China
- Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture and Rural Affairs; Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi Jiangsu, 214081, China
| | - Zaije Dong
- Wuxi Fisheries College, Nanjing Agricultural University, No.9 East Shanshui Road, Wuxi Jiangsu, 214081, China.
- Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture and Rural Affairs; Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi Jiangsu, 214081, China.
| |
Collapse
|
25
|
Petrovicz VL, Pasztuhov I, Martinek TA, Hegedüs Z. Site-directed allostery perturbation to probe the negative regulation of hypoxia inducible factor-1α. RSC Chem Biol 2024; 5:711-720. [PMID: 39092442 PMCID: PMC11289882 DOI: 10.1039/d4cb00066h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 05/27/2024] [Indexed: 08/04/2024] Open
Abstract
The interaction between the intrinsically disordered transcription factor HIF-1α and the coactivator proteins p300/CBP is essential in the fast response to low oxygenation. The negative feedback regulator, CITED2, switches off the hypoxic response through a very efficient irreversible mechanism. The negative cooperativity with HIF-1α relies on the formation of a ternary intermediate that leads to allosteric structural changes in p300/CBP, in which the cooperative folding/binding of the CITED2 sequence motifs plays a key role. Understanding the contribution of a binding motif to the structural changes in relation to competition efficiency provides invaluable insights into the molecular mechanism. Our strategy is to site-directedly perturb the p300-CITED2 complex's structure without significantly affecting binding thermodynamics. In this way, the contribution of a sequence motif to the negative cooperativity with HIF-1α would mainly depend on the induced structural changes, and to a lesser extent on binding affinity. Using biophysical assays and NMR measurements, we show here that the interplay between the N-terminal tail and the rest of the binding motifs of CITED2 is crucial for the unidirectional displacement of HIF-1α. We introduce an advantageous approach for evaluating the roles of the different sequence parts with the help of motif-by-motif backbone perturbations.
Collapse
Affiliation(s)
- Vencel L Petrovicz
- University of Szeged, Department of Medical Chemistry 8 Dóm tér Szeged 6720 Hungary
| | - István Pasztuhov
- University of Szeged, Department of Medical Chemistry 8 Dóm tér Szeged 6720 Hungary
| | - Tamás A Martinek
- University of Szeged, Department of Medical Chemistry 8 Dóm tér Szeged 6720 Hungary
- HUN-REN SZTE Biomimetic Systems Research Group 8 Dóm tér Szeged 6720 Hungary
| | - Zsófia Hegedüs
- University of Szeged, Department of Medical Chemistry 8 Dóm tér Szeged 6720 Hungary
| |
Collapse
|
26
|
Munshi R. How Transcription Factor Clusters Shape the Transcriptional Landscape. Biomolecules 2024; 14:875. [PMID: 39062589 PMCID: PMC11274464 DOI: 10.3390/biom14070875] [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: 06/01/2024] [Revised: 07/14/2024] [Accepted: 07/16/2024] [Indexed: 07/28/2024] Open
Abstract
In eukaryotic cells, gene transcription typically occurs in discrete periods of promoter activity, interspersed with intervals of inactivity. This pattern deviates from simple stochastic events and warrants a closer examination of the molecular interactions that activate the promoter. Recent studies have identified transcription factor (TF) clusters as key precursors to transcriptional bursting. Often, these TF clusters form at chromatin segments that are physically distant from the promoter, making changes in chromatin conformation crucial for promoter-TF cluster interactions. In this review, I explore the formation and constituents of TF clusters, examining how the dynamic interplay between chromatin architecture and TF clustering influences transcriptional bursting. Additionally, I discuss techniques for visualizing TF clusters and provide an outlook on understanding the remaining gaps in this field.
Collapse
Affiliation(s)
- Rahul Munshi
- Joseph Henry Laboratories of Physics and Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544, USA
| |
Collapse
|
27
|
Haghshenas S, Bout HJ, Schijns JM, Levy MA, Kerkhof J, Bhai P, McConkey H, Jenkins ZA, Williams EM, Halliday BJ, Huisman SA, Lauffer P, de Waard V, Witteveen L, Banka S, Brady AF, Galazzi E, van Gils J, Hurst ACE, Kaiser FJ, Lacombe D, Martinez-Monseny AF, Fergelot P, Monteiro FP, Parenti I, Persani L, Santos-Simarro F, Simpson BN, Alders M, Robertson SP, Sadikovic B, Menke LA. Menke-Hennekam syndrome; delineation of domain-specific subtypes with distinct clinical and DNA methylation profiles. HGG ADVANCES 2024; 5:100287. [PMID: 38553851 PMCID: PMC11040166 DOI: 10.1016/j.xhgg.2024.100287] [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: 10/20/2023] [Revised: 03/26/2024] [Accepted: 03/26/2024] [Indexed: 04/18/2024] Open
Abstract
CREB-binding protein (CBP, encoded by CREBBP) and its paralog E1A-associated protein (p300, encoded by EP300) are involved in histone acetylation and transcriptional regulation. Variants that produce a null allele or disrupt the catalytic domain of either protein cause Rubinstein-Taybi syndrome (RSTS), while pathogenic missense and in-frame indel variants in parts of exons 30 and 31 cause phenotypes recently described as Menke-Hennekam syndrome (MKHK). To distinguish MKHK subtypes and define their characteristics, molecular and extended clinical data on 82 individuals (54 unpublished) with variants affecting CBP (n = 71) or p300 (n = 11) (NP_004371.2 residues 1,705-1,875 and NP_001420.2 residues 1,668-1,833, respectively) were summarized. Additionally, genome-wide DNA methylation profiles were assessed in DNA extracted from whole peripheral blood from 54 individuals. Most variants clustered closely around the zinc-binding residues of two zinc-finger domains (ZZ and TAZ2) and within the first α helix of the fourth intrinsically disordered linker (ID4) of CBP/p300. Domain-specific methylation profiles were discerned for the ZZ domain in CBP/p300 (found in nine out of 10 tested individuals) and TAZ2 domain in CBP (in 14 out of 20), while a domain-specific diagnostic episignature was refined for the ID4 domain in CBP/p300 (in 21 out of 21). Phenotypes including intellectual disability of varying degree and distinct physical features were defined for each of the regions. These findings demonstrate existence of at least three MKHK subtypes, which are domain specific (MKHK-ZZ, MKHK-TAZ2, and MKHK-ID4) rather than gene specific (CREBBP/EP300). DNA methylation episignatures enable stratification of molecular pathophysiologic entities within a gene or across a family of paralogous genes.
Collapse
Affiliation(s)
- Sadegheh Haghshenas
- Verspeeten Clinical Genome Centre, London Health Sciences Centre, London ON N6A 5W9, Canada
| | - Hidde J Bout
- Department of Pediatrics, Emma Children's Hospital, Amsterdam UMC, University of Amsterdam, Amsterdam Reproduction and Development Research Institute, 1105 Amsterdam, AZ, the Netherlands
| | - Josephine M Schijns
- Department of Pediatrics, Emma Children's Hospital, Amsterdam UMC, University of Amsterdam, Amsterdam Reproduction and Development Research Institute, 1105 Amsterdam, AZ, the Netherlands
| | - Michael A Levy
- Verspeeten Clinical Genome Centre, London Health Sciences Centre, London ON N6A 5W9, Canada
| | - Jennifer Kerkhof
- Verspeeten Clinical Genome Centre, London Health Sciences Centre, London ON N6A 5W9, Canada
| | - Pratibha Bhai
- Verspeeten Clinical Genome Centre, London Health Sciences Centre, London ON N6A 5W9, Canada
| | - Haley McConkey
- Verspeeten Clinical Genome Centre, London Health Sciences Centre, London ON N6A 5W9, Canada
| | - Zandra A Jenkins
- Department of Women's and Children's Health, Dunedin School of Medicine, University of Otago, Dunedin 9016, New Zealand
| | - Ella M Williams
- Department of Women's and Children's Health, Dunedin School of Medicine, University of Otago, Dunedin 9016, New Zealand
| | - Benjamin J Halliday
- Department of Women's and Children's Health, Dunedin School of Medicine, University of Otago, Dunedin 9016, New Zealand
| | - Sylvia A Huisman
- Department of Pediatrics, Emma Children's Hospital, Amsterdam UMC, University of Amsterdam, Amsterdam Reproduction and Development Research Institute, 1105 Amsterdam, AZ, the Netherlands; Zodiak, Prinsenstichting, Purmerend, JE 1444, the Netherlands
| | - Peter Lauffer
- Department of Human Genetics, Amsterdam UMC, University of Amsterdam, Amsterdam Reproduction and Development Research Institute, Amsterdam 1105 AZ, the Netherlands
| | - Vivian de Waard
- Department of Medical Biochemistry, Amsterdam UMC, University of Amsterdam, Amsterdam Cardiovascular Sciences, Amsterdam, AZ 1105, the Netherlands
| | - Laura Witteveen
- Department of Pediatrics, Emma Children's Hospital, Amsterdam UMC, University of Amsterdam, Amsterdam Reproduction and Development Research Institute, 1105 Amsterdam, AZ, the Netherlands
| | - Siddharth Banka
- Division of Evolution, Infection and Genomics, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9WL, UK; Manchester Centre for Genomic Medicine, Saint Mary's Hospital, Manchester University NHS Foundation Trust, Manchester M13 9WL, UK
| | - Angela F Brady
- North West Thames Regional Genetics Service, Northwick Park Hospital, Harrow HA1 3UJ, UK
| | - Elena Galazzi
- Department of Endocrine & Metabolic Diseases, San Luca Hospital, IRCCS Istituto Auxologico Italiano, 20100 Milan, Italy
| | - Julien van Gils
- Centre Hospitalier Universitaire Bordeaux, 33404 Bordeaux, France
| | - Anna C E Hurst
- Department of Genetics, University of Alabama, Birmingham, AL 35294-0024, USA
| | - Frank J Kaiser
- Institute of Human Genetics, University of Duisburg-Essen, 45122 Essen, Germany; Center for Rare Diseases, University Hospital Essen, 45122 Essen, Germany
| | - Didier Lacombe
- Centre Hospitalier Universitaire Bordeaux, 33404 Bordeaux, France
| | - Antonio F Martinez-Monseny
- Genètica Clínica, Servei de Medicina Genètica i Molecular, Hospital Sant Joan de Déu, 08950 Barcelona, Spain
| | | | | | - Ilaria Parenti
- Institute of Human Genetics, University of Duisburg-Essen, 45122 Essen, Germany
| | - Luca Persani
- Department of Endocrine & Metabolic Diseases, San Luca Hospital, IRCCS Istituto Auxologico Italiano, 20100 Milan, Italy; Department of Medical Biotechnology and Translational Medicine, University of Milan, 20100 Milan, Italy
| | - Fernando Santos-Simarro
- Institute of Medical and Molecular Genetics (INGEMM), Hospital Universitario La Paz, IdiPAZ, CIBERER, ISCIII, 28029 Madrid, Spain; Unit of Molecular Diagnostics and Clinical Genetics, Hospital Universitari Son Espases, Health Research Institute of the Balearic Islands (IdISBa), 07120 Palma, Spain
| | - Brittany N Simpson
- Department of Pediatrics, Division of Human Genetics, Cincinnati Children's Hospital Medical Center, University of Cincinnati School of Medicine, Cincinnati, OH 45206, USA
| | - Mariëlle Alders
- Department of Human Genetics, Amsterdam UMC, University of Amsterdam, Amsterdam Reproduction and Development Research Institute, Amsterdam 1105 AZ, the Netherlands
| | - Stephen P Robertson
- Department of Women's and Children's Health, Dunedin School of Medicine, University of Otago, Dunedin 9016, New Zealand
| | - Bekim Sadikovic
- Verspeeten Clinical Genome Centre, London Health Sciences Centre, London ON N6A 5W9, Canada; Department of Pathology and Laboratory Medicine, Western University, London, ON N6A3K7, Canada.
| | - Leonie A Menke
- Department of Pediatrics, Emma Children's Hospital, Amsterdam UMC, University of Amsterdam, Amsterdam Reproduction and Development Research Institute, 1105 Amsterdam, AZ, the Netherlands.
| |
Collapse
|
28
|
Kato H, Salgado M, Mendez D, Gonzalez N, Rawson J, Ligot D, Balandran B, Orr C, Quijano JC, Omori K, Qi M, Al-Abdullah IH, Mullen Y, Ku HT, Kandeel F, Komatsu H. Biological hypoxia in pre-transplant human pancreatic islets induces transplant failure in diabetic mice. Sci Rep 2024; 14:12402. [PMID: 38811610 PMCID: PMC11137081 DOI: 10.1038/s41598-024-61604-3] [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: 02/05/2024] [Accepted: 05/07/2024] [Indexed: 05/31/2024] Open
Abstract
Evaluating the quality of isolated human islets before transplantation is crucial for predicting the success in treating Type 1 diabetes. The current gold standard involves time-intensive in vivo transplantation into diabetic immunodeficient mice. Given the susceptibility of isolated islets to hypoxia, we hypothesized that hypoxia present in islets before transplantation could indicate compromised islet quality, potentially leading to unfavorable outcomes. To test this hypothesis, we analyzed expression of 39 hypoxia-related genes in human islets from 85 deceased donors. We correlated gene expression profiles with transplantation outcomes in 327 diabetic mice, each receiving 1200 islet equivalents grafted into the kidney capsule. Transplantation outcome was post-transplant glycemic control based on area under the curve of blood glucose over 4 weeks. In linear regression analysis, DDIT4 (R = 0.4971, P < 0.0001), SLC2A8 (R = 0.3531, P = 0.0009) and HK1 (R = 0.3444, P = 0.0012) had the highest correlation with transplantation outcome. A multiple regression model of 11 genes increased the correlation (R = 0.6117, P < 0.0001). We conclude that assessing pre-transplant hypoxia in human islets via gene expression analysis is a rapid, viable alternative to conventional in vivo assessments. This approach also underscores the importance of mitigating pre-transplant hypoxia in isolated islets to improve the success rate of islet transplantation.
Collapse
Affiliation(s)
- Hiroyuki Kato
- Department of Translational Research and Cellular Therapeutics, Arthur Riggs Diabetes AND Metabolism Research Institute of City of Hope, 1500 E. Duarte Rd., Duarte, CA, 91010, USA
- Department of Surgery, University of California, San Francisco, 513 Parnassus Ave., San Francisco, CA, 94143, USA
| | - Mayra Salgado
- Department of Translational Research and Cellular Therapeutics, Arthur Riggs Diabetes AND Metabolism Research Institute of City of Hope, 1500 E. Duarte Rd., Duarte, CA, 91010, USA
| | - Daniel Mendez
- Department of Translational Research and Cellular Therapeutics, Arthur Riggs Diabetes AND Metabolism Research Institute of City of Hope, 1500 E. Duarte Rd., Duarte, CA, 91010, USA
| | - Nelson Gonzalez
- Department of Translational Research and Cellular Therapeutics, Arthur Riggs Diabetes AND Metabolism Research Institute of City of Hope, 1500 E. Duarte Rd., Duarte, CA, 91010, USA
| | - Jeffrey Rawson
- Department of Translational Research and Cellular Therapeutics, Arthur Riggs Diabetes AND Metabolism Research Institute of City of Hope, 1500 E. Duarte Rd., Duarte, CA, 91010, USA
| | - Doreen Ligot
- Department of Translational Research and Cellular Therapeutics, Arthur Riggs Diabetes AND Metabolism Research Institute of City of Hope, 1500 E. Duarte Rd., Duarte, CA, 91010, USA
| | - Bennie Balandran
- Department of Translational Research and Cellular Therapeutics, Arthur Riggs Diabetes AND Metabolism Research Institute of City of Hope, 1500 E. Duarte Rd., Duarte, CA, 91010, USA
| | - Chris Orr
- Department of Translational Research and Cellular Therapeutics, Arthur Riggs Diabetes AND Metabolism Research Institute of City of Hope, 1500 E. Duarte Rd., Duarte, CA, 91010, USA
| | - Janine C Quijano
- Department of Translational Research and Cellular Therapeutics, Arthur Riggs Diabetes AND Metabolism Research Institute of City of Hope, 1500 E. Duarte Rd., Duarte, CA, 91010, USA
| | - Keiko Omori
- Department of Translational Research and Cellular Therapeutics, Arthur Riggs Diabetes AND Metabolism Research Institute of City of Hope, 1500 E. Duarte Rd., Duarte, CA, 91010, USA
| | - Meirigeng Qi
- Department of Translational Research and Cellular Therapeutics, Arthur Riggs Diabetes AND Metabolism Research Institute of City of Hope, 1500 E. Duarte Rd., Duarte, CA, 91010, USA
| | - Ismail H Al-Abdullah
- Department of Translational Research and Cellular Therapeutics, Arthur Riggs Diabetes AND Metabolism Research Institute of City of Hope, 1500 E. Duarte Rd., Duarte, CA, 91010, USA
| | - Yoko Mullen
- Department of Translational Research and Cellular Therapeutics, Arthur Riggs Diabetes AND Metabolism Research Institute of City of Hope, 1500 E. Duarte Rd., Duarte, CA, 91010, USA
| | - Hsun Teresa Ku
- Department of Translational Research and Cellular Therapeutics, Arthur Riggs Diabetes AND Metabolism Research Institute of City of Hope, 1500 E. Duarte Rd., Duarte, CA, 91010, USA
| | - Fouad Kandeel
- Department of Translational Research and Cellular Therapeutics, Arthur Riggs Diabetes AND Metabolism Research Institute of City of Hope, 1500 E. Duarte Rd., Duarte, CA, 91010, USA
| | - Hirotake Komatsu
- Department of Translational Research and Cellular Therapeutics, Arthur Riggs Diabetes AND Metabolism Research Institute of City of Hope, 1500 E. Duarte Rd., Duarte, CA, 91010, USA.
- Department of Surgery, University of California, San Francisco, 513 Parnassus Ave., San Francisco, CA, 94143, USA.
| |
Collapse
|
29
|
Dai Q, Yuan Z, Sun Q, Ao Z, He B, Jiang Y. Discovery of novel nucleoside derivatives as selective lysine acetyltransferase p300 inhibitors for cancer therapy. Bioorg Med Chem Lett 2024; 104:129742. [PMID: 38604299 DOI: 10.1016/j.bmcl.2024.129742] [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: 11/28/2023] [Revised: 04/05/2024] [Accepted: 04/08/2024] [Indexed: 04/13/2024]
Abstract
P300 and CBP are two closely related histone acetyltransferases that are important transcriptional coactivators of many cellular processes. Inhibition of the transcriptional regulator p300/CBP is a promising therapeutic approach in oncology. However, there are no reported single selective p300 or CBP inhibitors to date. In this study, we designed and optimized a series of lysine acetyltransferase p300 selective inhibitors bearing a nucleoside scaffold. Most compounds showed excellent inhibitory activity against p300 with IC50 ranging from 0.18 to 9.90 μM, except for J16, J29, J40, and J48. None of the compounds showed inhibitory activity against CBP (inhibition rate < 50 % at 10 µM). Then the cytotoxicity of the compounds against a series of cancer cells were evaluated. Compounds J31 and J32 showed excellent proliferation inhibitory activity on cancer cells T47D and H520 with desirable selectivity profile of p300 over CBP. These compounds could be promising lead compounds for the development of novel epigenetic inhibitors as antitumor agents.
Collapse
Affiliation(s)
- Qiuzi Dai
- The Hunan Provincial University Key Laboratory of the Fundamental and Clinical Research on Functional Nucleic Acid, Hunan Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations, Changsha Medical University, Changsha 410219, China
| | - Zigao Yuan
- Shenzhen Kivita Innovative Drug Discovery Institute, Shenzhen 518055, China
| | - Qinsheng Sun
- Shenzhen Kivita Innovative Drug Discovery Institute, Shenzhen 518055, China
| | - Zhuolin Ao
- Division of Biosciences, Department of Biochemistry, University College London, London WC1E6AA, UK
| | - Binsheng He
- The Hunan Provincial University Key Laboratory of the Fundamental and Clinical Research on Functional Nucleic Acid, Hunan Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations, Changsha Medical University, Changsha 410219, China.
| | - Yuyang Jiang
- Shenzhen Kivita Innovative Drug Discovery Institute, Shenzhen 518055, China; State Key Laboratory of Chemical Oncogenomics, Tsinghua Shenzhen International Graduate School, Shenzhen 518055, China; School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, China.
| |
Collapse
|
30
|
Ciesla J, Huang KL, Wagner EJ, Munger J. A UL26-PIAS1 complex antagonizes anti-viral gene expression during Human Cytomegalovirus infection. PLoS Pathog 2024; 20:e1012058. [PMID: 38768227 PMCID: PMC11142722 DOI: 10.1371/journal.ppat.1012058] [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: 02/18/2024] [Revised: 05/31/2024] [Accepted: 04/29/2024] [Indexed: 05/22/2024] Open
Abstract
Viral disruption of innate immune signaling is a critical determinant of productive infection. The Human Cytomegalovirus (HCMV) UL26 protein prevents anti-viral gene expression during infection, yet the mechanisms involved are unclear. We used TurboID-driven proximity proteomics to identify putative UL26 interacting proteins during infection to address this issue. We find that UL26 forms a complex with several immuno-regulatory proteins, including several STAT family members and various PIAS proteins, a family of E3 SUMO ligases. Our results indicate that UL26 prevents STAT phosphorylation during infection and antagonizes transcriptional activation induced by either interferon α (IFNA) or tumor necrosis factor α (TNFα). Additionally, we find that the inactivation of PIAS1 sensitizes cells to inflammatory stimulation, resulting in an anti-viral transcriptional environment similar to ΔUL26 infection. Further, PIAS1 is important for HCMV cell-to-cell spread, which depends on the presence of UL26, suggesting that the UL26-PIAS1 interaction is vital for modulating intrinsic anti-viral defense.
Collapse
Affiliation(s)
- Jessica Ciesla
- Department of Biochemistry and Biophysics, University of Rochester School of Medicine and Dentistry, Rochester, New York, United States of America
| | - Kai-Lieh Huang
- Department of Biochemistry and Biophysics, University of Rochester School of Medicine and Dentistry, Rochester, New York, United States of America
| | - Eric J. Wagner
- Department of Biochemistry and Biophysics, University of Rochester School of Medicine and Dentistry, Rochester, New York, United States of America
| | - Joshua Munger
- Department of Biochemistry and Biophysics, University of Rochester School of Medicine and Dentistry, Rochester, New York, United States of America
| |
Collapse
|
31
|
Tomasello B, Bellia F, Naletova I, Magrì A, Tabbì G, Attanasio F, Tomasello MF, Cairns WRL, Fortino M, Pietropaolo A, Greco V, La Mendola D, Sciuto S, Arena G, Rizzarelli E. BDNF- and VEGF-Responsive Stimulus to an NGF Mimic Cyclic Peptide with Copper Ionophore Capability and Ctr1/CCS-Driven Signaling. ACS Chem Neurosci 2024; 15:1755-1769. [PMID: 38602894 DOI: 10.1021/acschemneuro.3c00716] [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] [Indexed: 04/13/2024] Open
Abstract
Neurotrophins are a family of growth factors that play a key role in the development and regulation of the functioning of the central nervous system. Their use as drugs is made difficult by their poor stability, cellular permeability, and side effects. Continuing our effort to use peptides that mimic the neurotrophic growth factor (NGF), the family model protein, and specifically the N-terminus of the protein, here we report on the spectroscopic characterization and resistance to hydrolysis of the 14-membered cyclic peptide reproducing the N-terminus sequence (SSSHPIFHRGEFSV (c-NGF(1-14)). Far-UV CD spectra and a computational study show that this peptide has a rigid conformation and left-handed chirality typical of polyproline II that favors its interaction with the D5 domain of the NGF receptor TrkA. c-NGF(1-14) is able to bind Cu2+ with good affinity; the resulting complexes have been characterized by potentiometric and spectroscopic measurements. Experiments on PC12 cells show that c-NGF(1-14) acts as an ionophore, influencing the degree and the localization of both the membrane transporter (Ctr1) and the copper intracellular transporter (CCS). c-NGF(1-14) induces PC12 differentiation, mimics the protein in TrkA phosphorylation, and activates the kinase cascade, inducing Erk1/2 phosphorylation. c-NGF(1-14) biological activities are enhanced when the peptide interacts with Cu2+ even with the submicromolar quantities present in the culture media as demonstrated by ICP-OES measurements. Finally, c-NGF(1-14) and Cu2+ concur to activate the cAMP response element-binding protein CREB that, in turn, induces the brain-derived neurotrophic factor (BDNF) and the vascular endothelial growth factor (VEGF) release.
Collapse
Affiliation(s)
- Barbara Tomasello
- Department of Drug and Health Sciences, University of Catania, V.le Andrea Doria 6, Catania 95125, Italy
| | - Francesco Bellia
- Institute of Crystallography, CNR, P. Gaifami 18, Catania 95126, Italy
| | - Irina Naletova
- Institute of Crystallography, CNR, P. Gaifami 18, Catania 95126, Italy
| | - Antonio Magrì
- Institute of Crystallography, CNR, P. Gaifami 18, Catania 95126, Italy
| | - Giovanni Tabbì
- Institute of Crystallography, CNR, P. Gaifami 18, Catania 95126, Italy
| | | | | | - Warren R L Cairns
- Istituto di Scienze Polari (ISP), c/o Campus Scientifico, Università Ca' Foscari Venezia Via Torino, Venezia Mestre 155-30170, Italy
| | - Mariagrazia Fortino
- Dipartimento di Scienze della Salute, Università di Catanzaro, Viale Europa, Catanzaro 88100, Italy
| | - Adriana Pietropaolo
- Dipartimento di Scienze della Salute, Università di Catanzaro, Viale Europa, Catanzaro 88100, Italy
| | - Valentina Greco
- Department of Chemical Sciences, University of Catania, A. Doria 6, Catania 95125, Italy
| | - Diego La Mendola
- Department of Pharmaceutical Sciences, University of Pisa, Bonanno Pisano 12, Pisa 56126, Italy
| | - Sebastiano Sciuto
- Department of Chemical Sciences, University of Catania, A. Doria 6, Catania 95125, Italy
| | - Giuseppe Arena
- Department of Chemical Sciences, University of Catania, A. Doria 6, Catania 95125, Italy
| | - Enrico Rizzarelli
- Institute of Crystallography, CNR, P. Gaifami 18, Catania 95126, Italy
- Department of Chemical Sciences, University of Catania, A. Doria 6, Catania 95125, Italy
| |
Collapse
|
32
|
Kim S, Park J, Choi Y, Jeon H, Lim N. Investigating the Relevance of Cyclic Adenosine Monophosphate Response Element-Binding Protein to the Wound Healing Process: An In Vivo Study Using Photobiomodulation Treatment. Int J Mol Sci 2024; 25:4838. [PMID: 38732058 PMCID: PMC11084265 DOI: 10.3390/ijms25094838] [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: 02/24/2024] [Revised: 04/05/2024] [Accepted: 04/25/2024] [Indexed: 05/13/2024] Open
Abstract
Monitoring inflammatory cytokines is crucial for assessing healing process and photobiomodulation (PBM) enhances wound healing. Meanwhile, cAMP response element-binding protein (CREB) is a regulator of cellular metabolism and proliferation. This study explored potential links between inflammatory cytokines and the activity of CREB in PBM-treated wounds. A total of 48 seven-week-old male SD rats were divided into four groups (wound location, skin or oral; treatment method, natural healing or PBM treatment). Wounds with a 6 mm diameter round shape were treated five times with an 808 nm laser every other day (total 60 J). The wound area was measured with a caliper and calculated using the elliptical formula. Histological analysis assessed the epidermal regeneration and collagen expression of skin and oral tissue with H&E and Masson's trichrome staining. Pro-inflammatory (TNF-α) and anti-inflammatory (TGF-β) cytokines were quantified by RT-PCR. The ratio of phosphorylated CREB (p-CREB) to unphosphorylated CREB was identified through Western blot. PBM treatment significantly reduced the size of the wounds on day 3 and day 7, particularly in the skin wound group (p < 0.05 on day 3, p < 0.001 on day 7). The density of collagen expression was significantly higher in the PBM treatment group (in skin wound, p < 0.05 on day 3, p < 0.001 on day 7, and p < 0.05 on day 14; in oral wound, p < 0.01 on day 7). The TGF-β/TNF-α ratio and the p-CREB/CREB ratio showed a parallel trend during wound healing. Our findings suggested that the CREB has potential as a meaningful marker to track the wound healing process.
Collapse
Affiliation(s)
- Sungyeon Kim
- Department of Plastic and Reconstructive Surgery, Dankook University College of Medicine, Cheonan 31116, Chungnam, Republic of Korea; (S.K.); (H.J.)
| | - Jion Park
- Department of Medical Laser, Graduate School, Dankook University, Cheonan 31116, Chungnam, Republic of Korea;
| | - Younghoon Choi
- Institute of Medical Science, Dankook University Hospital, Cheonan 31116, Chungnam, Republic of Korea;
| | - Hongbae Jeon
- Department of Plastic and Reconstructive Surgery, Dankook University College of Medicine, Cheonan 31116, Chungnam, Republic of Korea; (S.K.); (H.J.)
- Dankook Physician Scientist Research Center (DPSRC), Dankook University Hospital, Cheonan 31116, Chungnam, Republic of Korea
| | - Namkyu Lim
- Department of Plastic and Reconstructive Surgery, Dankook University College of Medicine, Cheonan 31116, Chungnam, Republic of Korea; (S.K.); (H.J.)
- Dankook Physician Scientist Research Center (DPSRC), Dankook University Hospital, Cheonan 31116, Chungnam, Republic of Korea
| |
Collapse
|
33
|
Inge MM, Miller R, Hook H, Bray D, Keenan JL, Zhao R, Gilmore TD, Siggers T. Rapid profiling of transcription factor-cofactor interaction networks reveals principles of epigenetic regulation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.05.588333. [PMID: 38617258 PMCID: PMC11014505 DOI: 10.1101/2024.04.05.588333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/16/2024]
Abstract
Transcription factor (TF)-cofactor (COF) interactions define dynamic, cell-specific networks that govern gene expression; however, these networks are understudied due to a lack of methods for high-throughput profiling of DNA-bound TF-COF complexes. Here we describe the Cofactor Recruitment (CoRec) method for rapid profiling of cell-specific TF-COF complexes. We define a lysine acetyltransferase (KAT)-TF network in resting and stimulated T cells. We find promiscuous recruitment of KATs for many TFs and that 35% of KAT-TF interactions are condition specific. KAT-TF interactions identify NF-κB as a primary regulator of acutely induced H3K27ac. Finally, we find that heterotypic clustering of CBP/P300-recruiting TFs is a strong predictor of total promoter H3K27ac. Our data supports clustering of TF sites that broadly recruit KATs as a mechanism for widespread co-occurring histone acetylation marks. CoRec can be readily applied to different cell systems and provides a powerful approach to define TF-COF networks impacting chromatin state and gene regulation.
Collapse
Affiliation(s)
- M M Inge
- Department of Biology, Boston University, Boston, MA, USA
- Biological Design Center, Boston University, Boston, MA, USA
- These authors contributed equally
| | - R Miller
- Department of Biology, Boston University, Boston, MA, USA
- Bioinformatics Program, Boston University, Boston, MA, USA
- Biological Design Center, Boston University, Boston, MA, USA
- These authors contributed equally
| | - H Hook
- Department of Biology, Boston University, Boston, MA, USA
| | - D Bray
- Department of Biology, Boston University, Boston, MA, USA
- Bioinformatics Program, Boston University, Boston, MA, USA
| | - J L Keenan
- Department of Biology, Boston University, Boston, MA, USA
- Bioinformatics Program, Boston University, Boston, MA, USA
| | - R Zhao
- Department of Biology, Boston University, Boston, MA, USA
| | - T D Gilmore
- Department of Biology, Boston University, Boston, MA, USA
| | - T Siggers
- Department of Biology, Boston University, Boston, MA, USA
- Bioinformatics Program, Boston University, Boston, MA, USA
- Biological Design Center, Boston University, Boston, MA, USA
| |
Collapse
|
34
|
Qian P, Wang S, Zhang T, Wu J. Transcriptional Expression of Histone Acetyltransferases and Deacetylases During the Recovery of Acute Exercise in Mouse Hippocampus. J Mol Neurosci 2024; 74:34. [PMID: 38565829 DOI: 10.1007/s12031-024-02215-5] [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: 11/06/2023] [Accepted: 03/21/2024] [Indexed: 04/04/2024]
Abstract
Protein acetylation, which is dynamically maintained by histone acetyltransferases (HATs) and deacetylases (HDACs), might play essential roles in hippocampal exercise physiology. However, whether HATs/HDACs are imbalanced during the recovery phase following acute exercise has not been determined. Groups of exercised mice with different recovery periods after acute exercise (0 h, 0.5 h, 1 h, 4 h, 7 h, and 24 h) were constructed, and a group of sham-exercised mice was used as the control. The mRNA levels of HATs and HDACs were detected via real-time quantitative polymerase chain reaction. Lysine acetylation on the total proteins and some specific locations on histones were detected via western blotting, as were various acylation modifications on the total proteins. Except for four unaffected genes (Hdac4, Ncoa1, Ncoa2, and Sirt1), the mRNA expression trajectories of 21 other HATs or HDACs affected by exercise could be categorized into three clusters. The genes in Cluster 1 increased quickly following exercise, with a peak at 0.5 h and/or 1 h, and remained at high levels until 24 h. Cluster 2 genes presented a gradual increase with a delayed peak at 4 h or 7 h postexercise before returning to baseline. The expression of Cluster 3 genes decreased at 0.5 h and/or 1 h, with some returning to overexpression (Hdac1 and Sirt3). Although most HATs were upregulated and half of the affected HDACs were downregulated at 0.5 h postexercise, the global or residue-specific histone acetylation levels were unchanged. In contrast, the levels of several metabolism-related acylation products of total proteins, including acetylation, succinylation, 2-hydroxyisobutyryllysine, β-hydroxybutyryllysine, and lactylation, decreased and mainly occurred on nonhistones immediately after exercise. During the 24-h recovery phase after acute exercise, the transcriptional trajectory of HATs or the same class of HDACs in the hippocampus exhibited heterogeneity. Although acute exercise did not affect the selected sites on histone lysine residues, it possibly incurred changes in acetylation and other acylation on nonhistone proteins.
Collapse
Affiliation(s)
- Ping Qian
- Beijing Municipal Key Laboratory of Child Development and Nutriomics, Capital Institute of Pediatrics, Beijing, 100020, China
- Department of Internal Medicine, Affiliated Children Hospital of Capital Institute of Pediatrics, Beijing, 100020, China
| | - Shan Wang
- Beijing Municipal Key Laboratory of Child Development and Nutriomics, Capital Institute of Pediatrics, Beijing, 100020, China
| | - Ting Zhang
- Beijing Municipal Key Laboratory of Child Development and Nutriomics, Capital Institute of Pediatrics, Beijing, 100020, China.
| | - Jianxin Wu
- Beijing Municipal Key Laboratory of Child Development and Nutriomics, Capital Institute of Pediatrics, Beijing, 100020, China.
- Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China.
| |
Collapse
|
35
|
Zavrtanik U, Medved T, Purič S, Vranken W, Lah J, Hadži S. Leucine Motifs Stabilize Residual Helical Structure in Disordered Proteins. J Mol Biol 2024; 436:168444. [PMID: 38218366 DOI: 10.1016/j.jmb.2024.168444] [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: 12/03/2023] [Revised: 12/31/2023] [Accepted: 01/09/2024] [Indexed: 01/15/2024]
Abstract
Many examples are known of regions of intrinsically disordered proteins that fold into α-helices upon binding to their targets. These helical binding motifs (HBMs) can be partially helical also in the unbound state, and this so-called residual structure can affect binding affinity and kinetics. To investigate the underlying mechanisms governing the formation of residual helical structure, we assembled a dataset of experimental helix contents of 65 peptides containing HBM that fold-upon-binding. The average residual helicity is 17% and increases to 60% upon target binding. The helix contents of residual and target-bound structures do not correlate, however the relative location of helix elements in both states shows a strong overlap. Compared to the general disordered regions, HBMs are enriched in amino acids with high helix preference and these residues are typically involved in target binding, explaining the overlap in helix positions. In particular, we find that leucine residues and leucine motifs in HBMs are the major contributors to helix stabilization and target-binding. For the two model peptides, we show that substitution of leucine motifs to other hydrophobic residues (valine or isoleucine) leads to reduction of residual helicity, supporting the role of leucine as helix stabilizer. From the three hydrophobic residues only leucine can efficiently stabilize residual helical structure. We suggest that the high occurrence of leucine motifs and a general preference for leucine at binding interfaces in HBMs can be explained by its unique ability to stabilize helical elements.
Collapse
Affiliation(s)
- Uroš Zavrtanik
- Department of Physical Chemistry, Faculty of Chemistry and Chemical Technology, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Tadej Medved
- Department of Physical Chemistry, Faculty of Chemistry and Chemical Technology, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Samo Purič
- Graduate Study Program, Faculty of Chemistry and Chemical Technology, University of Ljubljana, SI-1000 Ljubljana, Slovenia
| | - Wim Vranken
- Artificial Intelligence Laboratory, Vrije Universiteit Brussel, Pleinlaan 2, 1050 Brussels, Belgium; Interuniversity Institute of Bioinformatics in Brussels, ULB/VUB, Triomflaan, 1050 Brussels, Belgium; Structural Biology Brussels, Vrije Universiteit Brussel, Brussels 1050, Belgium; VIB Structural Biology Research Centre, Brussels 1050, Belgium
| | - Jurij Lah
- Department of Physical Chemistry, Faculty of Chemistry and Chemical Technology, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - San Hadži
- Department of Physical Chemistry, Faculty of Chemistry and Chemical Technology, University of Ljubljana, 1000 Ljubljana, Slovenia.
| |
Collapse
|
36
|
Udupa A, Kotha SR, Staller MV. Commonly asked questions about transcriptional activation domains. Curr Opin Struct Biol 2024; 84:102732. [PMID: 38056064 PMCID: PMC11193542 DOI: 10.1016/j.sbi.2023.102732] [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: 07/21/2023] [Revised: 10/23/2023] [Accepted: 10/27/2023] [Indexed: 12/08/2023]
Abstract
Eukaryotic transcription factors activate gene expression with their DNA-binding domains and activation domains. DNA-binding domains bind the genome by recognizing structurally related DNA sequences; they are structured, conserved, and predictable from protein sequences. Activation domains recruit chromatin modifiers, coactivator complexes, or basal transcriptional machinery via structurally diverse protein-protein interactions. Activation domains and DNA-binding domains have been called independent, modular units, but there are many departures from modularity, including interactions between these regions and overlap in function. Compared to DNA-binding domains, activation domains are poorly understood because they are poorly conserved, intrinsically disordered, and difficult to predict from protein sequences. This review, organized around commonly asked questions, describes recent progress that the field has made in understanding the sequence features that control activation domains and predicting them from sequence.
Collapse
Affiliation(s)
- Aditya Udupa
- Department of Molecular and Cell Biology, University of California, Berkeley, 94720, USA
| | - Sanjana R Kotha
- Department of Molecular and Cell Biology, University of California, Berkeley, 94720, USA; Center for Computational Biology, University of California, Berkeley, 94720, USA
| | - Max V Staller
- Department of Molecular and Cell Biology, University of California, Berkeley, 94720, USA; Center for Computational Biology, University of California, Berkeley, 94720, USA; Chan Zuckerberg Biohub-San Francisco, San Francisco, CA 94158, USA.
| |
Collapse
|
37
|
Shimoda LA. KAT's in the Cradle: p300/CBP and Regulation of HIF-1 and Blood Pressure during Intermittent Hypoxia. Am J Respir Cell Mol Biol 2024; 70:87-88. [PMID: 38109691 PMCID: PMC10848693 DOI: 10.1165/rcmb.2023-0435ed] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 12/18/2023] [Indexed: 12/20/2023] Open
Affiliation(s)
- Larissa A Shimoda
- Division of Pulmonary and Critical Care Medicine Johns Hopkins School of Medicine Baltimore, Maryland
| |
Collapse
|
38
|
Gou P, Zhang W. Protein lysine acetyltransferase CBP/p300: A promising target for small molecules in cancer treatment. Biomed Pharmacother 2024; 171:116130. [PMID: 38215693 DOI: 10.1016/j.biopha.2024.116130] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 01/02/2024] [Accepted: 01/02/2024] [Indexed: 01/14/2024] Open
Abstract
CBP and p300 are homologous proteins exhibiting remarkable structural and functional similarity. Both proteins function as acetyltransferase and coactivator, underscoring their significant roles in cellular processes. The function of histone acetyltransferases is to facilitate the release of DNA from nucleosomes and act as transcriptional co-activators to promote gene transcription. Transcription factors recruit CBP/p300 by co-condensation and induce transcriptional bursting. Disruption of CBP or p300 functions is associated with different diseases, especially cancer, which can result from either loss of function or gain of function. CBP and p300 are multidomain proteins containing HAT (histone acetyltransferase) and BRD (bromodomain) domains, which perform acetyltransferase activity and maintenance of HAT signaling, respectively. Inhibitors targeting HAT and BRD have been explored for decades, and some BRD inhibitors have been evaluated in clinical trials for treating hematologic malignancies or advanced solid tumors. Here, we review the development and application of CBP/p300 inhibitors. Several inhibitors have been evaluated in vivo, exhibiting notable potency but limited selectivity. Exploring these inhibitors emphasizes the promise of targeting CBP and p300 with small molecules in cancer therapy.
Collapse
Affiliation(s)
- Panhong Gou
- Department of Lymphoma and Myeloma, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Wenchao Zhang
- Department of Lymphoma and Myeloma, University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
39
|
Corda PO, Bollen M, Ribeiro D, Fardilha M. Emerging roles of the Protein Phosphatase 1 (PP1) in the context of viral infections. Cell Commun Signal 2024; 22:65. [PMID: 38267954 PMCID: PMC10807198 DOI: 10.1186/s12964-023-01468-8] [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: 10/16/2023] [Accepted: 12/30/2023] [Indexed: 01/26/2024] Open
Abstract
Protein Phosphatase 1 (PP1) is a major serine/threonine phosphatase in eukaryotes, participating in several cellular processes and metabolic pathways. Due to their low substrate specificity, PP1's catalytic subunits do not exist as free entities but instead bind to Regulatory Interactors of Protein Phosphatase One (RIPPO), which regulate PP1's substrate specificity and subcellular localization. Most RIPPOs bind to PP1 through combinations of short linear motifs (4-12 residues), forming highly specific PP1 holoenzymes. These PP1-binding motifs may, hence, represent attractive targets for the development of specific drugs that interfere with a subset of PP1 holoenzymes. Several viruses exploit the host cell protein (de)phosphorylation machinery to ensure efficient virus particle formation and propagation. While the role of many host cell kinases in viral life cycles has been extensively studied, the targeting of phosphatases by viral proteins has been studied in less detail. Here, we compile and review what is known concerning the role of PP1 in the context of viral infections and discuss how it may constitute a putative host-based target for the development of novel antiviral strategies.
Collapse
Affiliation(s)
- Pedro O Corda
- Institute of Biomedicine (iBiMED), Department of Medical Sciences, University of Aveiro, Aveiro, Portugal
| | - Mathieu Bollen
- Department of Cellular and Molecular Medicine, Laboratory of Biosignaling & Therapeutics, Katholieke Universiteit Leuven, Louvain, Belgium
| | - Daniela Ribeiro
- Institute of Biomedicine (iBiMED), Department of Medical Sciences, University of Aveiro, Aveiro, Portugal.
| | - Margarida Fardilha
- Institute of Biomedicine (iBiMED), Department of Medical Sciences, University of Aveiro, Aveiro, Portugal.
| |
Collapse
|
40
|
Mellios N, Papageorgiou G, Gorgievski V, Maxson G, Hernandez M, Otero M, Varangis M, Dell'Orco M, Perrone-Bizzozero N, Tzavara E. Regulation of neuronal circHomer1 biogenesis by PKA/CREB/ERK-mediated pathways and effects of glutamate and dopamine receptor blockade. RESEARCH SQUARE 2024:rs.3.rs-3547375. [PMID: 38260249 PMCID: PMC10802743 DOI: 10.21203/rs.3.rs-3547375/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
There are currently only very few efficacious drug treatments for SCZ and BD, none of which can significantly ameliorate cognitive symptoms. Thus, further research is needed in elucidating molecular pathways linked to cognitive function and antipsychotic treatment. Circular RNAs (circRNAs) are stable brain-enriched non-coding RNAs, derived from the covalent back-splicing of precursor mRNA molecules. CircHomer1 is a neuronal-enriched, activity-dependent circRNA, derived from the precursor of the long HOMER1B mRNA isoform, which is significantly downregulated in the prefrontal cortex of subjects with psychosis and is able to regulate cognitive function. Even though its relevance to psychiatric disorders and its role in brain function and synaptic plasticity have been well established, little is known about the molecular mechanisms that underlie circHomer1 biogenesis in response to neuronal activity and psychiatric drug treatment. Here we suggest that the RNA-binding protein (RBP) FUS positively regulates neuronal circHomer1 expression. Furthermore, we show that the MEK/ERK and PKA/CREB pathways positively regulate neuronal circHomer1 expression, as well as promote the transcription of Fus and Eif4a3, another RBP previously shown to activate circHomer1 biogenesis. We then demonstrate via both in vitro and in vivo studies that NMDA and mGluR5 receptors are upstream modulators of circHomer1 expression. Lastly, we report that in vivo D2R antagonism increases circHomer1 expression, whereas 5HT2AR blockade reduces circHomer1 levels in multiple brain regions. Taken together, this study allows us to gain novel insights into the molecular circuits that underlie the biogenesis of a psychiatric disease-associated circRNA.
Collapse
|
41
|
Liu Y, Joy ST, Henley MJ, Croskey A, Yates JA, Merajver SD, Mapp AK. Inhibition of CREB Binding and Function with a Dual-Targeting Ligand. Biochemistry 2024; 63:1-8. [PMID: 38086054 PMCID: PMC10836052 DOI: 10.1021/acs.biochem.3c00469] [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: 01/03/2024]
Abstract
CBP/p300 is a master transcriptional coactivator that regulates gene activation by interacting with multiple transcriptional activators. Dysregulation of protein-protein interactions (PPIs) between the CBP/p300 KIX domain and its activators is implicated in a number of cancers, including breast, leukemia, and colorectal cancer. However, KIX is typically considered "undruggable" because of its shallow binding surfaces lacking both significant topology and promiscuous binding profiles. We previously reported a dual-targeting peptide (MybLL-tide) that inhibits the KIX-Myb interaction with excellent specificity and potency. Here, we demonstrate a branched, second-generation analogue, CREBLL-tide, that inhibits the KIX-CREB PPI with higher potency and selectivity. Additionally, the best of these CREBLL-tide analogues shows excellent and selective antiproliferation activity in breast cancer cells. These results indicate that CREBLL-tide is an effective tool for assessing the role of KIX-activator interactions in breast cancer and expanding the dual-targeting strategy for inhibiting KIX and other coactivators that contain multiple binding surfaces.
Collapse
Affiliation(s)
- Yejun Liu
- Life Sciences Institute, University of Michigan, Ann Arbor, Michigan 48109, United States
- Program in Chemical Biology, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Stephen T Joy
- Life Sciences Institute, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Madeleine J Henley
- Life Sciences Institute, University of Michigan, Ann Arbor, Michigan 48109, United States
- Program in Chemical Biology, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Ayza Croskey
- Life Sciences Institute, University of Michigan, Ann Arbor, Michigan 48109, United States
- Program in Chemical Biology, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Joel A Yates
- Department of Internal Medicine, Hematology/Oncology, University of Michigan Medical School, Ann Arbor, Michigan 48109, United States
| | - Sofia D Merajver
- Department of Internal Medicine, Hematology/Oncology, University of Michigan Medical School, Ann Arbor, Michigan 48109, United States
| | - Anna K Mapp
- Life Sciences Institute, University of Michigan, Ann Arbor, Michigan 48109, United States
- Program in Chemical Biology, University of Michigan, Ann Arbor, Michigan 48109, United States
- Department of Chemistry, University of Michigan, Ann Arbor, Michigan 48109, United States
| |
Collapse
|
42
|
Shahrajabian MH, Sun W. Characterization of Intrinsically Disordered Proteins in Healthy and Diseased States by Nuclear Magnetic Resonance. Rev Recent Clin Trials 2024; 19:176-188. [PMID: 38409704 DOI: 10.2174/0115748871271420240213064251] [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: 09/13/2023] [Revised: 11/10/2023] [Accepted: 12/13/2023] [Indexed: 02/28/2024]
Abstract
INTRODUCTION Intrinsically Disordered Proteins (IDPs) are active in different cellular procedures like ordered assembly of chromatin and ribosomes, interaction with membrane, protein, and ligand binding, molecular recognition, binding, and transportation via nuclear pores, microfilaments and microtubules process and disassembly, protein functions, RNA chaperone, and nucleic acid binding, modulation of the central dogma, cell cycle, and other cellular activities, post-translational qualification and substitute splicing, and flexible entropic linker and management of signaling pathways. METHODS The intrinsic disorder is a precise structural characteristic that permits IDPs/IDPRs to be involved in both one-to-many and many-to-one signaling. IDPs/IDPRs also exert some dynamical and structural ordering, being much less constrained in their activities than folded proteins. Nuclear magnetic resonance (NMR) spectroscopy is a major technique for the characterization of IDPs, and it can be used for dynamic and structural studies of IDPs. RESULTS AND CONCLUSION This review was carried out to discuss intrinsically disordered proteins and their different goals, as well as the importance and effectiveness of NMR in characterizing intrinsically disordered proteins in healthy and diseased states.
Collapse
Affiliation(s)
- Mohamad Hesam Shahrajabian
- National Key Laboratory of Agricultural Microbiology, Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Wenli Sun
- National Key Laboratory of Agricultural Microbiology, Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| |
Collapse
|
43
|
DelRosso N, Bintu L. Using High-Throughput Measurements to Identify Principles of Transcriptional and Epigenetic Regulators. Methods Mol Biol 2024; 2842:79-101. [PMID: 39012591 DOI: 10.1007/978-1-0716-4051-7_4] [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] [Indexed: 07/17/2024]
Abstract
To achieve exquisite control over the epigenome, we need a better predictive understanding of how transcription factors, chromatin regulators, and their individual domain's function, both as modular parts and as full proteins. Transcriptional effector domains are one class of protein domains that regulate transcription and chromatin. These effector domains either repress or activate gene expression by interacting with chromatin-modifying enzymes, transcriptional cofactors, and/or general transcriptional machinery. Here, we discuss important design considerations for high-throughput investigations of effector domains, recent advances in discovering new domains in human cells and testing how domain function depends on amino acid sequence. For every effector domain, we would like to know the following: What role does the cell type, signaling state, and targeted context have on activation, silencing, and epigenetic memory? Large-scale measurements of transcriptional activities can help systematically answer these questions and identify general rules for how all these parameters affect effector domain activities. Last, we discuss what steps need to be taken to turn a newly discovered effector domain into a robust, precise epigenome editor. With more carefully considered high-throughput investigations, soon we will have better predictive control over the epigenome.
Collapse
|
44
|
Lackner A, Cabral JE, Qiu Y, Zhou H, Leonidas L, Pham MA, Macapagal A, Lin S, Armanus E, McNulty R. Small molecule inhibitor binds to NLRP3 and prevents inflammasome activation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.13.571573. [PMID: 38168343 PMCID: PMC10760093 DOI: 10.1101/2023.12.13.571573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
Despite recent advances in the mechanism of oxidized DNA activating NLRP3, the molecular mechanism and consequence of oxidized DNA associating with NLRP3 remains unknown. Cytosolic NLRP3 binds oxidized DNA which has been released from the mitochondria, which subsequently triggers inflammasome activation. Human glycosylase (hOGG1) repairs oxidized DNA damage which inhibits inflammasome activation. The fold of NLRP3 pyrin domain contains amino acids and a protein fold similar to hOGG1. Amino acids that enable hOGG1 to bind and cleave oxidized DNA are conserved in NLRP3. We found NLRP3 could bind and cleave oxidized guanine within mitochondrial DNA. The binding of oxidized DNA to NLRP3 was prevented by small molecule drugs which also inhibit hOGG1. These same drugs also inhibited inflammasome activation. Elucidating this mechanism will enable design of drug memetics that treat inflammasome pathologies, illustrated herein by NLRP3 pyrin domain inhibitors which suppressed interleukin-1β (IL-1β) production in macrophages. One-Sentence Summary NLRP3 cleaves oxidized DNA and small molecule drug binding inhibits inflammasome activation.
Collapse
|
45
|
Nicosia L, Spencer GJ, Brooks N, Amaral FMR, Basma NJ, Chadwick JA, Revell B, Wingelhofer B, Maiques-Diaz A, Sinclair O, Camera F, Ciceri F, Wiseman DH, Pegg N, West W, Knurowski T, Frese K, Clegg K, Campbell VL, Cavet J, Copland M, Searle E, Somervaille TCP. Therapeutic targeting of EP300/CBP by bromodomain inhibition in hematologic malignancies. Cancer Cell 2023; 41:2136-2153.e13. [PMID: 37995682 DOI: 10.1016/j.ccell.2023.11.001] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 08/07/2023] [Accepted: 11/01/2023] [Indexed: 11/25/2023]
Abstract
CCS1477 (inobrodib) is a potent, selective EP300/CBP bromodomain inhibitor which induces cell-cycle arrest and differentiation in hematologic malignancy model systems. In myeloid leukemia cells, it promotes rapid eviction of EP300/CBP from an enhancer subset marked by strong MYB occupancy and high H3K27 acetylation, with downregulation of the subordinate oncogenic network and redistribution to sites close to differentiation genes. In myeloma cells, CCS1477 induces eviction of EP300/CBP from FGFR3, the target of the common (4; 14) translocation, with redistribution away from IRF4-occupied sites to TCF3/E2A-occupied sites. In a subset of patients with relapsed or refractory disease, CCS1477 monotherapy induces differentiation responses in AML and objective responses in heavily pre-treated multiple myeloma. In vivo preclinical combination studies reveal synergistic responses to treatment with standard-of-care agents. Thus, CCS1477 exhibits encouraging preclinical and early-phase clinical activity by disrupting recruitment of EP300/CBP to enhancer networks occupied by critical transcription factors.
Collapse
Affiliation(s)
- Luciano Nicosia
- Leukaemia Biology Laboratory, Cancer Research UK Manchester Institute, The University of Manchester, Manchester M20 4BX, UK
| | - Gary J Spencer
- Leukaemia Biology Laboratory, Cancer Research UK Manchester Institute, The University of Manchester, Manchester M20 4BX, UK
| | | | - Fabio M R Amaral
- Leukaemia Biology Laboratory, Cancer Research UK Manchester Institute, The University of Manchester, Manchester M20 4BX, UK
| | - Naseer J Basma
- Leukaemia Biology Laboratory, Cancer Research UK Manchester Institute, The University of Manchester, Manchester M20 4BX, UK
| | - John A Chadwick
- Leukaemia Biology Laboratory, Cancer Research UK Manchester Institute, The University of Manchester, Manchester M20 4BX, UK
| | - Bradley Revell
- Leukaemia Biology Laboratory, Cancer Research UK Manchester Institute, The University of Manchester, Manchester M20 4BX, UK
| | - Bettina Wingelhofer
- Leukaemia Biology Laboratory, Cancer Research UK Manchester Institute, The University of Manchester, Manchester M20 4BX, UK
| | - Alba Maiques-Diaz
- Leukaemia Biology Laboratory, Cancer Research UK Manchester Institute, The University of Manchester, Manchester M20 4BX, UK
| | - Oliver Sinclair
- Leukaemia Biology Laboratory, Cancer Research UK Manchester Institute, The University of Manchester, Manchester M20 4BX, UK
| | - Francesco Camera
- Leukaemia Biology Laboratory, Cancer Research UK Manchester Institute, The University of Manchester, Manchester M20 4BX, UK
| | - Filippo Ciceri
- Leukaemia Biology Laboratory, Cancer Research UK Manchester Institute, The University of Manchester, Manchester M20 4BX, UK
| | - Daniel H Wiseman
- Epigenetics of Haematopoiesis Group, The University of Manchester, Manchester M20 4BX, UK
| | - Neil Pegg
- CellCentric Ltd., Cambridge CB10 1XL, UK
| | - Will West
- CellCentric Ltd., Cambridge CB10 1XL, UK
| | | | - Kris Frese
- CellCentric Ltd., Cambridge CB10 1XL, UK
| | | | | | - James Cavet
- The Christie NHS Foundation Trust, Manchester M20 4BX, UK
| | - Mhairi Copland
- Paul O'Gorman Leukaemia Research Centre, University of Glasgow, Glasgow G12 0YN, UK
| | - Emma Searle
- The Christie NHS Foundation Trust, Manchester M20 4BX, UK
| | - Tim C P Somervaille
- Leukaemia Biology Laboratory, Cancer Research UK Manchester Institute, The University of Manchester, Manchester M20 4BX, UK; The Christie NHS Foundation Trust, Manchester M20 4BX, UK.
| |
Collapse
|
46
|
Zhang L, Zhu K, Xu J, Chen X, Sheng C, Zhang D, Yang Y, Sun L, Zhao H, Wang X, Tao B, Zhou L, Liu J. Acetyltransferases CBP/p300 Control Transcriptional Switch of β-Catenin and Stat1 Promoting Osteoblast Differentiation. J Bone Miner Res 2023; 38:1885-1899. [PMID: 37850815 DOI: 10.1002/jbmr.4925] [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: 08/03/2023] [Revised: 10/02/2023] [Accepted: 10/13/2023] [Indexed: 10/19/2023]
Abstract
CREB-binding protein (CBP) (CREBBP) and p300 (EP300) are multifunctional histone acetyltransferases (HATs) with extensive homology. Germline mutations of CBP or p300 cause skeletal abnormalities in humans and mice. However, the precise roles of CBP/p300 in bone homeostasis remain elusive. Here, we report that conditional knockout of CBP or p300 in osteoblasts results in reduced bone mass and strength due to suppressed bone formation. The HAT activity is further confirmed to be responsible for CBP/p300-mediated osteogenesis using A-485, a selective inhibitor of CBP/p300 HAT. Mechanistically, CBP/p300 HAT governs osteogenic gene expression in part through transcriptional activation of β-catenin and inhibition of Stat1. Furthermore, acetylation of histone H3K27 and the transcription factor Foxo1 are demonstrated to be involved in CBP/p300 HAT-regulated β-catenin and Stat1 transcription, respectively. Taken together, these data identify acetyltransferases CBP/p300 as critical regulators that promote osteoblast differentiation and reveal an epigenetic mechanism responsible for maintaining bone homeostasis. © 2023 American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Linlin Zhang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Endocrinology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Kecheng Zhu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jingzun Xu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaojing Chen
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chunxiang Sheng
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Deng Zhang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuying Yang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lihao Sun
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hongyan Zhao
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiao Wang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bei Tao
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Libin Zhou
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jianmin Liu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
47
|
Maruyama S, Yamazaki M, Abé T, Cheng J, Saku T, Tanuma JI. Hypoxia-Induced Biosynthesis of the Extracellular Matrix Molecules, Perlecan and Fibronectin, Promotes the Growth of Pleomorphic Adenoma Cells In Vitro Models. Biomedicines 2023; 11:2981. [PMID: 38001981 PMCID: PMC10669301 DOI: 10.3390/biomedicines11112981] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 10/25/2023] [Accepted: 10/30/2023] [Indexed: 11/26/2023] Open
Abstract
Salivary pleomorphic adenoma is histopathologically characterized by its colorful stroma with myxoid, chondroid, and hyaline appearances, due to enhanced biosynthesis of extracellular matrix (ECM) molecules and poor vascularity. Thus, pleomorphic adenoma cells embedded in the stroma typically survive under hypoxic conditions. We determined the expression kinetics of ECM molecules, such as perlecan and fibronectin (FN), under hypoxia in SM-AP1 cells which are duct epithelial differentiated cells, and in SM-AP4 cells, which are myoepithelial differentiated cells, cloned from pleomorphic adenoma of the parotid gland. We investigated hypoxia-inducible factor-1α (HIF-1α)-inducing pathways through a variety of ECM molecules in association with their cellular proliferation and migration. We observed that hypoxic conditions with elevated HIF-1α protein levels induced increased expression of perlecan and FN in SM-AP cells than in controls. Moreover, perlecan and FN knockdown reduced the proliferation of SM-AP1 and SM-AP4 cells under hypoxia. Further, SM-AP1 cell migration was enhanced by both perlecan and FN knockdown, whereas SM-AP4 cell migration was increased by perlecan knockdown and inhibited by fibronectin knockdown. The results indicated that pleomorphic adenoma cells can survive under hypoxic conditions by promoting cell proliferation via enhanced synthesis of ECM molecules. Overall, ECM molecules may be a new anti-tumor target under hypoxic conditions.
Collapse
Affiliation(s)
- Satoshi Maruyama
- Oral Pathology Section, Department of Surgical Pathology, Niigata University Hospital, 1-754 Asahimachi-dori, Chuo-ku, Niigata 951-8520, Japan
| | - Manabu Yamazaki
- Division of Oral Pathology, Department of Tissue Regeneration and Reconstruction, Faculty of Dentistry & Niigata University Graduate School of Medical and Dental Sciences, 2-5274 Gakkoucho-dori, Chuo-ku, Niigata 951-8514, Japan (T.A.); (J.-i.T.)
| | - Tatsuya Abé
- Division of Oral Pathology, Department of Tissue Regeneration and Reconstruction, Faculty of Dentistry & Niigata University Graduate School of Medical and Dental Sciences, 2-5274 Gakkoucho-dori, Chuo-ku, Niigata 951-8514, Japan (T.A.); (J.-i.T.)
| | - Jun Cheng
- Division of Oral Pathology, Department of Tissue Regeneration and Reconstruction, Faculty of Dentistry & Niigata University Graduate School of Medical and Dental Sciences, 2-5274 Gakkoucho-dori, Chuo-ku, Niigata 951-8514, Japan (T.A.); (J.-i.T.)
| | - Takashi Saku
- PCL Fukuoka Pathology Cytology Center, 4-11-32 Yoshizuka, Hakata-ku, Fukuoka 812-0041, Japan
| | - Jun-ichi Tanuma
- Division of Oral Pathology, Department of Tissue Regeneration and Reconstruction, Faculty of Dentistry & Niigata University Graduate School of Medical and Dental Sciences, 2-5274 Gakkoucho-dori, Chuo-ku, Niigata 951-8514, Japan (T.A.); (J.-i.T.)
| |
Collapse
|
48
|
Khan K, Zafar S, Badshah Y, Ashraf NM, Rafiq M, Danish L, Shabbir M, Trembley JH, Afsar T, Almajwal A, Razak S. Cross talk of tumor protein D52 (TPD52) with KLF9, PKCε, and MicroRNA 223 in ovarian cancer. J Ovarian Res 2023; 16:202. [PMID: 37833790 PMCID: PMC10571360 DOI: 10.1186/s13048-023-01292-1] [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: 04/26/2023] [Accepted: 10/02/2023] [Indexed: 10/15/2023] Open
Abstract
BACKGROUND Gynecologic cancers comprise malignancies in the female reproductive organs. Ovarian cancer ranks sixth in terms of incidence rates while seventh in terms of mortality rates. The stage at which ovarian cancer is diagnosed mainly determines the survival outcomes of patients. Various screening approaches are presently employed for diagnosing ovarian cancer; however, these techniques have low accuracy and are non-specific, resulting in high mortality rates of patients due to this disease. Hence, it is crucial to identify improved screening and diagnostic markers to overcome this cancer. This study aimed to find new biomarkers to facilitate the prognosis and diagnosis of ovarian cancer. METHODS Bioinformatics approaches were used to predict the tertiary structure and cellular localization along with phylogenetic analysis of TPD52. Its molecular interactions were determined through KEGG analysis, and real-time PCR-based expression analysis was performed to assess its co-expression with another oncogenic cellular pathway (miR-223, KLF9, and PKCε) proteins in ovarian cancer. RESULTS Bioinformatics analysis depicted the cytoplasmic localization of TPD52 and the high conservation of its coiled-coil domains. Further study revealed that TPD52 mRNA and miRNA-223 expression was elevated, while the expression of KLF 9 and PKCε was reduced in the blood of ovarian cancer patients. Furthermore, TPD52 and miR-223 expression were upregulated in the early stages of cancer and non-metastatic cancers. CONCLUSION TPD52, miR-223, PKCε, and KLF9, can be used as a blood based markers for disease prognosis, metastasis, and treatment response. The study outcomes hold great potential to be translated at the clinical level after further validation on larger cohorts.
Collapse
Affiliation(s)
- Khushbukhat Khan
- Atta-ur-Rahman School of Applied Biosciences (ASAB), National University of Sciences and Technology (NUST), Islamabad, 44000, Pakistan
| | - Sameen Zafar
- Atta-ur-Rahman School of Applied Biosciences (ASAB), National University of Sciences and Technology (NUST), Islamabad, 44000, Pakistan
| | - Yasmin Badshah
- Atta-ur-Rahman School of Applied Biosciences (ASAB), National University of Sciences and Technology (NUST), Islamabad, 44000, Pakistan
| | - Naeem Mahmood Ashraf
- School of Biochemistry & Biotechnology, University of the Punjab, Lahore, Pakistan
| | - Mehak Rafiq
- School of Interdisciplinary Engineering & Sciences (SINES), National University of Sciences and Technology, Islamabad, 44000, Pakistan
| | - Lubna Danish
- Agricultural Research Institute, Tarnab, Peshawar, Pakistan
| | - Maria Shabbir
- Atta-ur-Rahman School of Applied Biosciences (ASAB), National University of Sciences and Technology (NUST), Islamabad, 44000, Pakistan.
- Department of Healthcare Biotechnology, Atta-ur-Rahman School of Applied Biosciences, National University of Sciences and Technology, Islamabad, Pakistan.
| | - Janeen H Trembley
- Research Service, Minneapolis VA Health Care System, Minneapolis, MN, USA
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
| | - Tayyaba Afsar
- Department of Community Health Sciences, College of Applied Medical Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Ali Almajwal
- Department of Community Health Sciences, College of Applied Medical Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Suhail Razak
- Department of Community Health Sciences, College of Applied Medical Sciences, King Saud University, Riyadh, Saudi Arabia.
| |
Collapse
|
49
|
Kotha SR, Staller MV. Clusters of acidic and hydrophobic residues can predict acidic transcriptional activation domains from protein sequence. Genetics 2023; 225:iyad131. [PMID: 37462277 PMCID: PMC10550315 DOI: 10.1093/genetics/iyad131] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 07/03/2023] [Indexed: 10/06/2023] Open
Abstract
Transcription factors activate gene expression in development, homeostasis, and stress with DNA binding domains and activation domains. Although there exist excellent computational models for predicting DNA binding domains from protein sequence, models for predicting activation domains from protein sequence have lagged, particularly in metazoans. We recently developed a simple and accurate predictor of acidic activation domains on human transcription factors. Here, we show how the accuracy of this human predictor arises from the clustering of aromatic, leucine, and acidic residues, which together are necessary for acidic activation domain function. When we combine our predictor with the predictions of convolutional neural network (CNN) models trained in yeast, the intersection is more accurate than individual models, emphasizing that each approach carries orthogonal information. We synthesize these findings into a new set of activation domain predictions on human transcription factors.
Collapse
Affiliation(s)
- Sanjana R Kotha
- Department of Molecular and Cell Biology, University of California, Berkeley, CA 94720, USA
- Center for Computational Biology, University of California, Berkeley, CA 94720, USA
| | - Max Valentín Staller
- Department of Molecular and Cell Biology, University of California, Berkeley, CA 94720, USA
- Center for Computational Biology, University of California, Berkeley, CA 94720, USA
- Chan Zuckerberg Biohub—San Francisco, San Francisco, CA 94158, USA
| |
Collapse
|
50
|
Brown AD, Vergunst KL, Branch M, Blair CM, Dupré DJ, Baillie GS, Langelaan DN. Structural basis of CBP/p300 recruitment by the microphthalmia-associated transcription factor. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2023; 1870:119520. [PMID: 37353163 DOI: 10.1016/j.bbamcr.2023.119520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 05/19/2023] [Accepted: 06/08/2023] [Indexed: 06/25/2023]
Abstract
The microphthalmia-associated transcription factor (MITF) is a master regulator of the melanocyte cell lineage. Aberrant MITF activity can lead to multiple malignancies including skin cancer, where it modulates the progression and invasiveness of melanoma. MITF-regulated gene expression requires recruitment of the transcriptional co-regulator CBP/p300, but details of this process are not fully defined. In this study, we investigate the structural and functional interaction between the MITF N-terminal transactivation domain (MITFTAD) and CBP/p300. Using pulldown assays and nuclear magnetic resonance spectroscopy we determined that MITFTAD is intrinsically disordered and binds to the TAZ1 and TAZ2 domains of CBP/p300 with moderate affinity. The solution-state structure of the MITFTAD:TAZ2 complex reveals that MITF interacts with a hydrophobic surface of TAZ2, while remaining somewhat dynamic. Peptide array and mutagenesis experiments determined that an acidic motif is integral to the MITFTAD:TAZ2 interaction and is necessary for transcriptional activity of MITF. Peptides that bind to the same surface of TAZ2 as MITFTAD, such as the adenoviral protein E1A, are capable of displacing MITF from TAZ2 and inhibiting transactivation. These findings provide insight into co-activator recruitment by MITF that are fundamental to our understanding of MITF targeted gene regulation and melanoma biology.
Collapse
Affiliation(s)
- Alexandra D Brown
- Department of Biochemistry & Molecular Biology, Dalhousie University, Halifax, NS B3H 4R2, Canada
| | - Kathleen L Vergunst
- Department of Biochemistry & Molecular Biology, Dalhousie University, Halifax, NS B3H 4R2, Canada
| | - Makenzie Branch
- Department of Biochemistry & Molecular Biology, Dalhousie University, Halifax, NS B3H 4R2, Canada
| | - Connor M Blair
- School of Cardiovascular & Metabolic Health, University of Glasgow, Glasgow G12 8QQ, Scotland, United Kingdom of Great Britain and Northern Ireland
| | - Denis J Dupré
- Department of Pharmacology, Dalhousie University, Halifax, NS B3H 4R2, Canada
| | - George S Baillie
- School of Cardiovascular & Metabolic Health, University of Glasgow, Glasgow G12 8QQ, Scotland, United Kingdom of Great Britain and Northern Ireland
| | - David N Langelaan
- Department of Biochemistry & Molecular Biology, Dalhousie University, Halifax, NS B3H 4R2, Canada.
| |
Collapse
|