1
|
Xu X, Closson JD, Marcelino LP, Favaro DC, Silvestrini ML, Solazzo R, Chong LT, Gardner KH. Identification of small-molecule ligand-binding sites on and in the ARNT PAS-B domain. J Biol Chem 2024; 300:107606. [PMID: 39059491 PMCID: PMC11381877 DOI: 10.1016/j.jbc.2024.107606] [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: 06/11/2024] [Revised: 07/16/2024] [Accepted: 07/18/2024] [Indexed: 07/28/2024] Open
Abstract
Transcription factors are challenging to target with small-molecule inhibitors due to their structural plasticity and lack of catalytic sites. Notable exceptions include naturally ligand-regulated transcription factors, including our prior work with the hypoxia-inducible factor (HIF)-2 transcription factor, showing that small-molecule binding within an internal pocket of the HIF-2α Per-Aryl hydrocarbon Receptor Nuclear Translocator (ARNT)-Sim (PAS)-B domain can disrupt its interactions with its dimerization partner, ARNT. Here, we explore the feasibility of targeting small molecules to the analogous ARNT PAS-B domain itself, potentially opening a promising route to modulate several ARNT-mediated signaling pathways. Using solution NMR fragment screening, we previously identified several compounds that bind ARNT PAS-B and, in certain cases, antagonize ARNT association with the transforming acidic coiled-coil containing protein 3 transcriptional coactivator. However, these ligands have only modest binding affinities, complicating characterization of their binding sites. We address this challenge by combining NMR, molecular dynamics simulations, and ensemble docking to identify ligand-binding "hotspots" on and within the ARNT PAS-B domain. Our data indicate that the two ARNT/transforming acidic coiled-coil containing protein 3 inhibitors, KG-548 and KG-655, bind to a β-sheet surface implicated in both HIF-2 dimerization and coactivator recruitment. Furthermore, while KG-548 binds exclusively to the β-sheet surface, KG-655 can additionally bind within a water-accessible internal cavity in ARNT PAS-B. Finally, KG-279, while not a coactivator inhibitor, exemplifies ligands that preferentially bind only to the internal cavity. All three ligands promoted ARNT PAS-B homodimerization, albeit to varying degrees. Taken together, our findings provide a comprehensive overview of ARNT PAS-B ligand-binding sites and may guide the development of more potent coactivator inhibitors for cellular and functional studies.
Collapse
Affiliation(s)
- Xingjian Xu
- Structural Biology Initiative, CUNY Advanced Science Research Center, New York, New York, USA; PhD Program in Biochemistry, The Graduate Center, CUNY, New York, New York, USA
| | - Joseph D Closson
- Structural Biology Initiative, CUNY Advanced Science Research Center, New York, New York, USA; PhD Program in Biochemistry, The Graduate Center, CUNY, New York, New York, USA
| | | | - Denize C Favaro
- Structural Biology Initiative, CUNY Advanced Science Research Center, New York, New York, USA
| | - Marion L Silvestrini
- Department of Chemistry, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Riccardo Solazzo
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum-University of Bologna, Bologna, Bologna, Italy
| | - Lillian T Chong
- Department of Chemistry, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Kevin H Gardner
- Structural Biology Initiative, CUNY Advanced Science Research Center, New York, New York, USA; Department of Chemistry and Biochemistry, City College of New York, New York, New York, USA; PhD. Programs in Biochemistry, Chemistry and Biology, The Graduate Center, CUNY, New York, New York, USA.
| |
Collapse
|
2
|
Xu X, Closson J, Marcelino LP, Favaro DC, Silvestrini ML, Solazzo R, Chong LT, Gardner KH. Identification of Small Molecule Ligand Binding Sites On and In the ARNT PAS-B Domain. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.11.03.565595. [PMID: 37961463 PMCID: PMC10635134 DOI: 10.1101/2023.11.03.565595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Transcription factors are generally challenging to target with small molecule inhibitors due to their structural plasticity and lack of catalytic sites. Notable exceptions include several naturally ligand-regulated transcription factors, including our prior work with the heterodimeric HIF-2 transcription factor which showed that small molecule binding within an internal pocket of the HIF-2α PAS-B domain can disrupt its interactions with its dimerization partner, ARNT. Here, we explore the feasibility of similarly targeting small molecules to the analogous ARNT PAS-B domain itself, potentially opening a promising route to simultaneously modulate several ARNT-mediated signaling pathways. Using solution NMR screening of an in-house fragment library, we previously identified several compounds that bind ARNT PAS-B and, in certain cases, antagonize ARNT association with the TACC3 transcriptional coactivator. However, these ligands have only modest binding affinities, complicating characterization of their binding sites. We address this challenge by combining NMR, MD simulations, and ensemble docking to identify ligand-binding 'hotspots' on and within the ARNT PAS-B domain. Our data indicate that the two ARNT/TACC3 inhibitors, KG-548 and KG-655, bind to a β-sheet surface implicated in both HIF-2 dimerization and coactivator recruitment. Furthermore, while KG-548 binds exclusively to the β-sheet surface, KG-655 can additionally bind within a water-accessible internal cavity in ARNT PAS-B. Finally, KG-279, while not a coactivator inhibitor, exemplifies ligands that preferentially bind only to the internal cavity. All three ligands promoted ARNT PAS-B homodimerization, albeit to varying degrees. Taken together, our findings provide a comprehensive overview of ARNT PAS-B ligand-binding sites and may guide the development of more potent coactivator inhibitors for cellular and functional studies.
Collapse
|
3
|
Daffern N, Radhakrishnan I. Per-ARNT-Sim (PAS) Domains in Basic Helix-Loop-Helix (bHLH)-PAS Transcription Factors and Coactivators: Structures and Mechanisms. J Mol Biol 2024; 436:168370. [PMID: 37992889 PMCID: PMC10922228 DOI: 10.1016/j.jmb.2023.168370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 11/10/2023] [Accepted: 11/13/2023] [Indexed: 11/24/2023]
Abstract
PAS domains are ubiquitous in biology. They perform critically important roles in sensing and transducing a wide variety of environmental signals, and through their ability to bind small-molecule ligands, have emerged as targets for therapeutic intervention. Here, we discuss our current understanding of PAS domain structure and function in the context of basic helix-loop-helix (bHLH)-PAS transcription factors and coactivators. Unlike the bHLH-PAS domains of transcription factors, those of the steroid receptor coactivator (SRC) family are poorly characterized. Recent progress for this family and for the broader bHLH-PAS proteins suggest that these domains are ripe for deeper structural and functional studies.
Collapse
Affiliation(s)
- Nicolas Daffern
- Department of Molecular Biosciences, Northwestern University, Evanston, IL 60208, USA
| | - Ishwar Radhakrishnan
- Department of Molecular Biosciences, Northwestern University, Evanston, IL 60208, USA.
| |
Collapse
|
4
|
Hiranuma K, Asami Y, Kato MK, Murakami N, Shimada Y, Matsuda M, Yazaki S, Fujii E, Sudo K, Kuno I, Komatsu M, Hamamoto R, Makinoshima H, Matsumoto K, Ishikawa M, Kohno T, Terao Y, Itakura A, Yoshida H, Shiraishi K, Kato T. Rare FGFR fusion genes in cervical cancer and transcriptome-based subgrouping of patients with a poor prognosis. Cancer Med 2023; 12:17835-17848. [PMID: 37537783 PMCID: PMC10524028 DOI: 10.1002/cam4.6415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 06/25/2023] [Accepted: 07/26/2023] [Indexed: 08/05/2023] Open
Abstract
BACKGROUND Although cervical cancer is often characterized as preventable, its incidence continues to increase in low- and middle-income countries, underscoring the need to develop novel therapeutics for this disease.This study assessed the distribution of fusion genes across cancer types and used an RNA-based classification to divide cervical cancer patients with a poor prognosis into subgroups. MATERIAL AND METHODS RNA sequencing of 116 patients with cervical cancer was conducted. Fusion genes were extracted using StarFusion program. To identify a high-risk group for recurrence, 65 patients who received postoperative adjuvant therapy were subjected to non-negative matrix factorization to identify differentially expressed genes between recurrent and nonrecurrent groups. RESULTS We identified three cases with FGFR3-TACC3 and one with GOPC-ROS1 fusion genes as potential targets. A search of publicly available data from cBioPortal (21,789 cases) and the Center for Cancer Genomics and Advanced Therapeutics (32,608 cases) showed that the FGFR3 fusion is present in 1.5% and 0.6% of patients with cervical cancer, respectively. The frequency of the FGFR3 fusion gene was higher in cervical cancer than in other cancers, regardless of ethnicity. Non-negative matrix factorization identified that the patients were classified into four Basis groups. Pathway enrichment analysis identified more extracellular matrix kinetics dysregulation in Basis 3 and more immune system dysregulation in Basis 4 than in the good prognosis group. CIBERSORT analysis showed that the fraction of M1 macrophages was lower in the poor prognosis group than in the good prognosis group. CONCLUSIONS The distribution of FGFR fusion genes in patients with cervical cancer was determined by RNA-based analysis and used to classify patients into clinically relevant subgroups.
Collapse
Affiliation(s)
- Kengo Hiranuma
- Division of Genome BiologyNational Cancer Center Research InstituteTokyoJapan
- Department of Obstetrics and GynecologyJuntendo University Faculty of MedicineTokyoJapan
| | - Yuka Asami
- Division of Genome BiologyNational Cancer Center Research InstituteTokyoJapan
- Department of Obstetrics and GynecologyShowa University School of MedicineTokyoJapan
| | - Mayumi Kobayashi Kato
- Division of Genome BiologyNational Cancer Center Research InstituteTokyoJapan
- Department of GynecologyNational Cancer Center HospitalTokyoJapan
| | - Naoya Murakami
- Department of Radiation OncologyNational Cancer Center HospitalTokyoJapan
| | - Yoko Shimada
- Division of Genome BiologyNational Cancer Center Research InstituteTokyoJapan
| | - Maiko Matsuda
- Division of Genome BiologyNational Cancer Center Research InstituteTokyoJapan
| | - Shu Yazaki
- Division of Genome BiologyNational Cancer Center Research InstituteTokyoJapan
- Department of Medical OncologyNational Cancer Center HospitalTokyoJapan
| | - Erisa Fujii
- Division of Genome BiologyNational Cancer Center Research InstituteTokyoJapan
- Department of GynecologyNational Cancer Center HospitalTokyoJapan
| | - Kazuki Sudo
- Department of Medical OncologyNational Cancer Center HospitalTokyoJapan
| | - Ikumi Kuno
- Department of GynecologyNational Cancer Center HospitalTokyoJapan
| | - Masaaki Komatsu
- Division of Medical AI Research and DevelopmentNational Cancer Center Research InstituteTokyoJapan
- Cancer Translational Research TeamRIKEN Center for Advanced Intelligence ProjectTokyoJapan
| | - Ryuji Hamamoto
- Division of Medical AI Research and DevelopmentNational Cancer Center Research InstituteTokyoJapan
- Cancer Translational Research TeamRIKEN Center for Advanced Intelligence ProjectTokyoJapan
| | | | - Koji Matsumoto
- Department of Obstetrics and GynecologyShowa University School of MedicineTokyoJapan
| | - Mitsuya Ishikawa
- Department of GynecologyNational Cancer Center HospitalTokyoJapan
| | - Takashi Kohno
- Division of Genome BiologyNational Cancer Center Research InstituteTokyoJapan
| | - Yasuhisa Terao
- Department of Obstetrics and GynecologyJuntendo University Faculty of MedicineTokyoJapan
| | - Atsuo Itakura
- Department of Obstetrics and GynecologyJuntendo University Faculty of MedicineTokyoJapan
| | - Hiroshi Yoshida
- Department of Diagnostic PathologyNational Cancer Center HospitalTokyoJapan
| | - Kouya Shiraishi
- Division of Genome BiologyNational Cancer Center Research InstituteTokyoJapan
- Department of Clinical GenomicsNational Cancer Center Research InstituteTokyoJapan
| | - Tomoyasu Kato
- Department of GynecologyNational Cancer Center HospitalTokyoJapan
| |
Collapse
|
5
|
Cai M, Lin N, Su L, Wu X, Xie X, Xu S, Fu X, Xu L, Huang H. Fetal growth restriction: associated genetic etiology and pregnancy outcomes in a tertiary referral center. J Transl Med 2022; 20:168. [PMID: 35397568 PMCID: PMC8994287 DOI: 10.1186/s12967-022-03373-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 03/29/2022] [Indexed: 11/29/2022] Open
Abstract
Background The etiology of fetal growth restriction (FGR) is complex and currently, there is a paucity of research about the genetic etiology of fetal growth restriction. We investigated the genetic associations and pregnancy outcomes in cases of fetal growth restriction. Methods A retrospective analysis of 210 pregnant women with fetal growth restriction was performed using karyotype analysis and single nucleotide polymorphism arrays (SNP-array). The differences in pathogenic copy number variation (CNV) detected by the two methods were compared. At the same time, the fetuses were divided into three groups: isolated FGR (n = 117), FGR with ultrasonographic soft markers (n = 48), and FGR with ultrasonographic structural anomalies (n = 45). Further, the differences in pathogenic copy number variations were compared among the groups. Results The total detection rate of pathogenic CNVs was 12.4% (26/210). Pathogenic copy number variation was detected in 14 cases (6.7%, 14/210) by karyotype analysis. Furthermore, 25 cases (11.9%, 25/210) with pathogenic CNVs were detected using the SNP-array evaluation method. The difference in the pathogenic CNV detection rate between the two methods was statistically significant. The result of the karyotype analysis and SNP-array evaluation was inconsistent for 13 cases with pathogenic CNV. The rate of detecting pathogenic CNVs in fetuses with isolated FGR, FGR combined with ultrasonographic soft markers, and FGR combined with ultrasonographic structural malformations was 6.0, 10.4, and 31.1%, respectively, with significant differences among the groups. During the follow-up, 35 pregnancies were terminated, two abortions occurred, and 13 cases were lost to follow-up. Of the 160 deliveries, nine fetuses had adverse pregnancy outcomes, and the remaining 151 had normal postnatal growth and developmental assessments. Conclusions Early diagnosis and timely genomic testing for fetal growth restriction can aid in its perinatal prognosis and subsequent intervention. Supplementary Information The online version contains supplementary material available at 10.1186/s12967-022-03373-z.
Collapse
|
6
|
Li F, Cai Y, Deng S, Yang L, Liu N, Chang X, Jing L, Zhou Y, Li H. A peptide CORO1C-47aa encoded by the circular noncoding RNA circ-0000437 functions as a negative regulator in endometrium tumor angiogenesis. J Biol Chem 2021; 297:101182. [PMID: 34534547 PMCID: PMC8573159 DOI: 10.1016/j.jbc.2021.101182] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 08/28/2021] [Accepted: 09/08/2021] [Indexed: 01/16/2023] Open
Abstract
Circular RNAs (circRNAs) are a novel class of widespread noncoding RNAs that regulate gene expression in mammals. Recent studies demonstrate that functional peptides can be encoded by short open reading frames in noncoding RNAs, including circRNAs. However, the role of circRNAs in various physiological and pathological states, such as cancer, is not well understood. In this study, through deep RNA sequencing on human endometrial cancer (EC) samples and their paired adjacent normal tissues, we uncovered that the circRNA hsa-circ-0000437 is significantly reduced in EC compared with matched paracancerous tissue. The hsa-circ-0000437 contains a short open reading frame encoding a functional peptide termed CORO1C-47aa. Overexpression of CORO1C-47aa is capable of inhibiting angiogenesis at the initiation stage by suppressing endothelial cell proliferation, migration, and differentiation through competition with transcription factor TACC3 to bind to ARNT and suppress VEGF. CORO1C-47aa directly bound to ARNT through the PAS-B domain, and blocking the association between ARNT and TACC3, which led to reduced expression of VEGF, ultimately lead to reduced angiogenesis. The antitumor effects of CORO1C-47aa on EC progression suggest that CORO1C-47aa has potential value in anticarcinoma therapies and warrants further investigation.
Collapse
MESH Headings
- Animals
- Endometrial Neoplasms/blood supply
- Endometrial Neoplasms/genetics
- Endometrial Neoplasms/metabolism
- Endometrial Neoplasms/pathology
- Female
- Gene Expression Regulation, Neoplastic
- Humans
- Mice, Inbred BALB C
- Mice, Nude
- Microfilament Proteins/biosynthesis
- Microfilament Proteins/genetics
- Neoplasm Proteins/biosynthesis
- Neoplasm Proteins/genetics
- Neovascularization, Pathologic/genetics
- Neovascularization, Pathologic/metabolism
- Neovascularization, Pathologic/pathology
- Peptides/genetics
- Peptides/metabolism
- RNA, Circular/biosynthesis
- RNA, Circular/genetics
- RNA, Neoplasm/biosynthesis
- RNA, Neoplasm/genetics
- Mice
Collapse
Affiliation(s)
- Fang Li
- Department of Obstetrics and Gynecology, Third Hospital, Peking University, Beijing, China; Department of Genetics, Medical College of Soochow University, Suzhou, China; The State Key Lab of Respiratory Disease, The First Affiliated Hospital, The School of Public Health, Guangzhou Medical University, Guangzhou, China
| | - Yuhan Cai
- Department of Obstetrics and Gynecology, Third Hospital, Peking University, Beijing, China
| | - Sihan Deng
- Xiangya Medical School, Central South University, Changsha, China
| | - Lin Yang
- Department of Obstetrics and Gynecology, Third Hospital, Peking University, Beijing, China
| | - Na Liu
- Department of Obstetrics and Gynecology, No.6 Hospital, Beijing, China
| | - Xiaohan Chang
- Department of Obstetrics and Gynecology, Third Hospital, Peking University, Beijing, China
| | - Lankai Jing
- Department of Obstetrics and Gynecology, Third Hospital, Peking University, Beijing, China
| | - Yifeng Zhou
- Department of Genetics, Medical College of Soochow University, Suzhou, China
| | - Hua Li
- Department of Obstetrics and Gynecology, Third Hospital, Peking University, Beijing, China.
| |
Collapse
|
7
|
Xu X, Dikiy I, Evans MR, Marcelino LP, Gardner KH. Fragile protein folds: Sequence and environmental factors affecting the equilibrium of two interconverting, stably folded protein conformations. MAGNETIC RESONANCE (GOTTINGEN, GERMANY) 2021; 2:63-76. [PMID: 35603043 PMCID: PMC9119131 DOI: 10.5194/mr-2-63-2021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Recent research on fold-switching metamorphic proteins has revealed some notable exceptions to Anfinsen's hypothesis of protein folding. We have previously described how a single point mutation can enable a well-folded protein domain, one of the two PAS (Per-ARNT-Sim) domains of the human ARNT (aryl hydrocarbon receptor nuclear translocator) protein, to interconvert between two conformers related by a slip of an internal β-strand. Using this protein as a test case, we advance the concept of a "fragile fold," a protein fold that can reversibly rearrange into another fold that differs by a substantial number of hydrogen bonds, entailing reorganization of single secondary structure elements to more drastic changes seen in metamorphic proteins. Here we use a battery of biophysical tests to examine several factors affecting the equilibrium between the two conformations of the switching ARNT PAS-B Y456T protein. Of note, we find that factors which impact the HI loop preceding the shifted Iβ-strand affect both the equilibrium levels of the two conformers and the denatured state which links them in the interconversion process. Finally, we describe small molecules that selectively bind to and stabilize the wildtype conformation of ARNT PAS-B. These studies form a toolkit for studying fragile protein folds and could enable ways to modulate the biological functions of such fragile folds, both in natural and engineered proteins.
Collapse
Affiliation(s)
- Xingjian Xu
- Structural Biology Initiative, CUNY Advanced Science Research Center, New York, NY, USA
- Ph.D. Program in Biochemistry, The Graduate Center, CUNY, New York, NY, USA
| | - Igor Dikiy
- Structural Biology Initiative, CUNY Advanced Science Research Center, New York, NY, USA
- Current address: Regeneron Pharmaceuticals, Tarrytown, NY, USA
| | - Matthew R Evans
- Current address: Acclaim Physician Group, Inc. Fort Worth, TX, USA
| | - Leandro P Marcelino
- Structural Biology Initiative, CUNY Advanced Science Research Center, New York, NY, USA
- Department of Chemistry and Biochemistry, City College of New York, New York, NY, USA
| | - Kevin H Gardner
- Structural Biology Initiative, CUNY Advanced Science Research Center, New York, NY, USA
- Department of Chemistry and Biochemistry, City College of New York, New York, NY, USA
- Biochemistry, Chemistry and Biology Ph.D. Programs, The Graduate Center, CUNY, New York, NY, USA
| |
Collapse
|
8
|
Xu X, Gagné D, Aramini JM, Gardner KH. Volume and compressibility differences between protein conformations revealed by high-pressure NMR. Biophys J 2021; 120:924-935. [PMID: 33524371 DOI: 10.1016/j.bpj.2020.12.034] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 11/25/2020] [Accepted: 12/21/2020] [Indexed: 12/12/2022] Open
Abstract
Proteins often interconvert between different conformations in ways critical to their function. Although manipulating such equilibria for biophysical study is often challenging, the application of pressure is a potential route to achieve such control by favoring the population of lower volume states. Here, we use this feature to study the interconversion of ARNT PAS-B Y456T, which undergoes a dramatic +3 slip in the β-strand register as it switches between two stably folded conformations. Using high-pressure biomolecular NMR approaches, we obtained the first, to our knowledge, quantitative data testing two key hypotheses of this process: the slipped conformation is both smaller and less compressible than the wild-type equivalent, and the interconversion proceeds through a chiefly unfolded intermediate state. Data collected in steady-state pressure and time-resolved pressure-jump modes, including observed pressure-dependent changes in the populations of the two conformers and increased rate of interconversion between conformers, support both hypotheses. Our work exemplifies how these approaches, which can be generally applied to protein conformational switches, can provide unique information that is not easily accessible through other techniques.
Collapse
Affiliation(s)
- Xingjian Xu
- Structural Biology Initiative, CUNY Advanced Science Research Center, New York, New York; Ph.D Program in Biochemistry, The Graduate Center, CUNY, New York, New York
| | - Donald Gagné
- Structural Biology Initiative, CUNY Advanced Science Research Center, New York, New York
| | - James M Aramini
- Structural Biology Initiative, CUNY Advanced Science Research Center, New York, New York
| | - Kevin H Gardner
- Structural Biology Initiative, CUNY Advanced Science Research Center, New York, New York; Department of Chemistry and Biochemistry, City College of New York, New York, New York; Ph.D. Programs in Biochemistry, Chemistry, and Biology, The Graduate Center, CUNY, New York, New York.
| |
Collapse
|
9
|
Lu B, Jin H, Fu J. Molecular convergent and parallel evolution among four high-elevation anuran species from the Tibetan region. BMC Genomics 2020; 21:839. [PMID: 33246413 PMCID: PMC7694343 DOI: 10.1186/s12864-020-07269-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Accepted: 11/23/2020] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND To date, evidence for the relative prevalence or rarity of molecular convergent and parallel evolution is conflicting, and understanding of how these processes contribute to adaptation is limited. We compared four high-elevation anuran species (Bufo tibetanus, Nanorana parkeri, Rana kukunoris and Scutiger boulengeri) from the Tibetan region, and examined convergent and parallel amino acid substitutions between them and how they may have contributed to high-elevation adaptation. RESULTS Genomic data of the four high-elevation species and eight of their low-elevation close relatives were gathered. A total of 1098 orthologs shared by all species were identified. We first conducted pairwise comparisons using Zhang and Kumar's test. Then, the Rconv index was calculated and convergence/divergence correlation plotting was conducted. Furthermore, genes under positive selection and with elevated evolutionary rate were examined. We detected a large number of amino acid sites with convergent or parallel substitutions. Several pairs of high-elevation species, in particular, R. kukunoris vs N. parkeri and B. tibetanus vs S. boulengeri, had excessive amounts of convergent substitutions compared to neutral expectation. Nevertheless, these sites were mostly concentrated in a small number of genes (3-32), and no genome-wide convergence was detected. Furthermore, the majority of these convergent genes were neither under detectable positive selection nor had elevated evolutionary rates, although functional prediction analysis suggested some of the convergent genes could potentially contribute to high-elevation adaptation. CONCLUSIONS There is a substantial amount of convergent evolution at the amino-acid level among high-elevation amphibians, although these sites are concentrated in a few genes, not widespread across the genomes. This may attribute to the fact that all the target species are from the same environment. The relative prevalence of convergent substitutions among high-elevation amphibians provides an excellent opportunity for further study of molecular convergent evolution.
Collapse
Affiliation(s)
- Bin Lu
- Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu, China
| | - Hong Jin
- Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu, China.,University of the Chinese Academy of Sciences, Beijing, China
| | - Jinzhong Fu
- Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu, China. .,Department of Integrative Biology, University of Guelph, Guelph, Canada.
| |
Collapse
|
10
|
Wood SH, Hindle MM, Mizoro Y, Cheng Y, Saer BRC, Miedzinska K, Christian HC, Begley N, McNeilly J, McNeilly AS, Meddle SL, Burt DW, Loudon ASI. Circadian clock mechanism driving mammalian photoperiodism. Nat Commun 2020; 11:4291. [PMID: 32855407 PMCID: PMC7453030 DOI: 10.1038/s41467-020-18061-z] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Accepted: 07/27/2020] [Indexed: 12/19/2022] Open
Abstract
The annual photoperiod cycle provides the critical environmental cue synchronizing rhythms of life in seasonal habitats. In 1936, Bünning proposed a circadian-based coincidence timer for photoperiodic synchronization in plants. Formal studies support the universality of this so-called coincidence timer, but we lack understanding of the mechanisms involved. Here we show in mammals that long photoperiods induce the circadian transcription factor BMAL2, in the pars tuberalis of the pituitary, and triggers summer biology through the eyes absent/thyrotrophin (EYA3/TSH) pathway. Conversely, long-duration melatonin signals on short photoperiods induce circadian repressors including DEC1, suppressing BMAL2 and the EYA3/TSH pathway, triggering winter biology. These actions are associated with progressive genome-wide changes in chromatin state, elaborating the effect of the circadian coincidence timer. Hence, circadian clock-pituitary epigenetic pathway interactions form the basis of the mammalian coincidence timer mechanism. Our results constitute a blueprint for circadian-based seasonal timekeeping in vertebrates.
Collapse
Affiliation(s)
- S H Wood
- Centre for Biological Timing, Faculty of Life Sciences, University of Manchester, Manchester, M13 9PT, UK
- Arctic Chronobiology and Physiology Research Group, Department of Arctic and Marine Biology, UiT - The Arctic University of Norway, Tromsø, 9037, Norway
| | - M M Hindle
- The Roslin Institute, and Royal (Dick) School of Veterinary Studies University of Edinburgh, Roslin, Midlothian, EH25 9PRG, UK
| | - Y Mizoro
- Centre for Biological Timing, Faculty of Life Sciences, University of Manchester, Manchester, M13 9PT, UK
| | - Y Cheng
- UQ Genomics Initiative, The University of Queensland, Brisbane, QLD, 4072, Australia
- School of Life and Environmental Sciences, Faculty of Science, The University of Sydney, Camperdown, NSW, Australia
| | - B R C Saer
- Centre for Biological Timing, Faculty of Life Sciences, University of Manchester, Manchester, M13 9PT, UK
| | - K Miedzinska
- The Roslin Institute, and Royal (Dick) School of Veterinary Studies University of Edinburgh, Roslin, Midlothian, EH25 9PRG, UK
| | - H C Christian
- University of Oxford, Department of Physiology, Anatomy and Genetics, Le Gros Clark Building, South Parks Road, Oxford, OX1 3QX, UK
| | - N Begley
- Centre for Biological Timing, Faculty of Life Sciences, University of Manchester, Manchester, M13 9PT, UK
| | - J McNeilly
- MRC Centre for Reproductive Health, Queen's Medical Research Institute, Edinburgh, EH16 4TJ, UK
| | - A S McNeilly
- MRC Centre for Reproductive Health, Queen's Medical Research Institute, Edinburgh, EH16 4TJ, UK
| | - S L Meddle
- The Roslin Institute, and Royal (Dick) School of Veterinary Studies University of Edinburgh, Roslin, Midlothian, EH25 9PRG, UK
| | - D W Burt
- The Roslin Institute, and Royal (Dick) School of Veterinary Studies University of Edinburgh, Roslin, Midlothian, EH25 9PRG, UK
- UQ Genomics Initiative, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - A S I Loudon
- Centre for Biological Timing, Faculty of Life Sciences, University of Manchester, Manchester, M13 9PT, UK.
| |
Collapse
|
11
|
Chong AS, Anderson PC. Molecular Dynamics Simulations of the Hypoxia-Inducible Factor PAS-B Domain Confirm That Internally Bound Water Molecules Function To Stabilize the Protein Core for Ligand Binding. Biochemistry 2019; 59:450-459. [DOI: 10.1021/acs.biochem.9b00872] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Andrew S. Chong
- Department of Physical Sciences, University of Washington Bothell, Bothell, Washington 98011, United States
| | - Peter C. Anderson
- Department of Physical Sciences, University of Washington Bothell, Bothell, Washington 98011, United States
| |
Collapse
|
12
|
de Souza JV, Reznikov S, Zhu R, Bronowska AK. Druggability assessment of mammalian Per-Arnt-Sim [PAS] domains using computational approaches. MEDCHEMCOMM 2019; 10:1126-1137. [PMID: 31391885 PMCID: PMC6640724 DOI: 10.1039/c9md00148d] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/09/2019] [Accepted: 05/01/2019] [Indexed: 01/10/2023]
Abstract
Per-Arnt-Sim (PAS) domains are key regions that occur in different regulatory proteins from all kingdoms of life. PAS domains show a remarkably conserved structural scaffold, despite a highly variable primary sequence. In this study we have attempted to address some of the gaps in knowledge regarding the druggability of PAS-A domains, differences in structure and dynamics within the PAS domain family and how this affects the druggability potential, as well as give insight into the druggability of steroid receptor coactivators and putative binding modes of the NCOA1. Investigations were performed through a range of computational methods including molecular docking studies, atomistic molecular dynamics simulations, and hotspot mapping. Atomistic molecular dynamics simulations show that the function of the AhR PAS-B domain is regulated by the dynamics of the highly conserved tyrosine Y322 residue, which acts as a "gatekeeper" controlling the access to the binding cavity and finely tuning the binding affinity. Furthermore, the transition between the partially unfolded and helical conformation of the loop1 segment within PAS-B domains was shown to be essential for the generation of "druggable" sites, especially for the NCOA1 PAS-B domain. Finally, our simulations indicated the undruggability of PAS-A domains, caused by the inherent characteristics of their putative binding sites. In conclusion, this work emphasises the role of intrinsic dynamics in tuning the druggability of PAS-B domains and shows that PAS-B domains of steroid receptor coactivators, such as NCOA1, can be targeted by small molecule ligands, which highlights the potential of developing new therapeutics designed to target these coactivators using structure-based approaches.
Collapse
Affiliation(s)
- João V de Souza
- School of Natural and Environmental Sciences , Newcastle University , NE1 7RU Newcastle , UK .
| | - Sylvia Reznikov
- School of Natural and Environmental Sciences , Newcastle University , NE1 7RU Newcastle , UK .
| | - Ruidi Zhu
- School of Natural and Environmental Sciences , Newcastle University , NE1 7RU Newcastle , UK .
| | - Agnieszka K Bronowska
- School of Natural and Environmental Sciences , Newcastle University , NE1 7RU Newcastle , UK .
| |
Collapse
|
13
|
Modulation of HIF-2α PAS-B domain contributes to physiological responses. Proc Natl Acad Sci U S A 2018; 115:13240-13245. [PMID: 30523118 DOI: 10.1073/pnas.1810897115] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Hypoxia-inducible factors (HIFs) are transcription factors in the basic helix-loop-helix PER-ARNT-SIM (bHLH-PAS) protein family that contain internal hydrophobic cavities within their PAS-A and PAS-B domains. Among HIFs, the HIF-2α PAS-B domain contains a relatively large cavity exploited for the development of specific artificial ligands such as PT2399. Administration of PT2399 could suppress HIF-2α target gene expression without affecting HIF-1 activity in mice under hypoxia conditions. A single mutation (S305M) within the HIF-2α PAS-B domain suppressed HIF-2α activity while conferring resistance to PT2399 in vivo, indicating the vital role of PAS-B domain in HIF-2α hypoxia response. In contrast, the mutant mice did not phenocopy PT2399 intervention in wild-type mice under metabolic stress. Under a high-fat diet (HFD), the mutant mice exert enhanced adipogenesis and obtain larger adipose mass and body weight gain compared to wild type. However, administration of PT2399 along with HFD feeding sufficiently suppressed HFD-induced body weight and adipose mass increase through suppression of adipogenesis and lipogenesis. The accompanying decreased lipid accumulation in the liver and improved glucose tolerance in wild-type mice were not observed in the mutant mice indicating negative regulation of HIF-2α on obesity and a complex role for the PAS-B domain in metabolic regulation. Notably, short-term administration of PT2399 to obese mice decreased adipose mass and improved metabolic condition. These results indicate a regulatory role for HIF-2α in obesity progression and suggest a therapeutic opportunity for PT2399 in obesity and associated metabolic disorders.
Collapse
|
14
|
Luoma LM, Berry FB. Molecular analysis of NPAS3 functional domains and variants. BMC Mol Biol 2018; 19:14. [PMID: 30509165 PMCID: PMC6276216 DOI: 10.1186/s12867-018-0117-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Accepted: 11/26/2018] [Indexed: 12/14/2022] Open
Abstract
Background NPAS3 encodes a transcription factor which has been associated with multiple human psychiatric and neurodevelopmental disorders. In mice, deletion of Npas3 was found to cause alterations in neurodevelopment, as well as a marked reduction in neurogenesis in the adult mouse hippocampus. This neurogenic deficit, alongside the reduction in cortical interneuron number, likely contributes to the behavioral and cognitive alterations observed in Npas3 knockout mice. Although loss of Npas3 has been found to affect proliferation and apoptosis, the molecular function of NPAS3 is largely uncharacterized outside of predictions based on its high homology to bHLH–PAS transcription factors. Here we set out to characterize NPAS3 as a transcription factor, and to confirm whether NPAS3 acts as predicted for a Class 1 bHLH–PAS family member. Results Through these studies we have experimentally demonstrated that NPAS3 behaves as a true transcription factor, capable of gene regulation through direct association with DNA. NPAS3 and ARNT are confirmed to directly interact in human cells through both bHLH and PAS dimerization domains. The C-terminus of NPAS3 was found to contain a functional transactivation domain. Further, the NPAS3::ARNT heterodimer was shown to directly regulate the expression of VGF and TXNIP through binding of their proximal promoters. Finally, we assessed the effects of three human variants of NPAS3 on gene regulatory function and do not observe significant deficits. Conclusions NPAS3 is a true transcription factor capable of regulating expression of target genes through their promoters by directly cooperating with ARNT. The tested human variants of NPAS3 require further characterization to identify their effects on NPAS3 expression and function in the individuals that carry them. These data enhance our understanding of the molecular function of NPAS3 and the mechanism by which it contributes to normal and abnormal neurodevelopment and neural function. Electronic supplementary material The online version of this article (10.1186/s12867-018-0117-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Leiah M Luoma
- Department of Medical Genetics, University of Alberta, Edmonton, AB, Canada
| | - Fred B Berry
- Department of Medical Genetics, University of Alberta, Edmonton, AB, Canada. .,Department of Surgery, 3002D Li Ka Shing Centre, University of Alberta, Edmonton, AB, T6G 2E1, Canada.
| |
Collapse
|
15
|
TACC3 transcriptionally upregulates E2F1 to promote cell growth and confer sensitivity to cisplatin in bladder cancer. Cell Death Dis 2018; 9:72. [PMID: 29358577 PMCID: PMC5833822 DOI: 10.1038/s41419-017-0112-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Revised: 08/28/2017] [Accepted: 09/18/2017] [Indexed: 12/11/2022]
Abstract
Accumulating evidence has shown that transforming acidic coiled-coil 3 (TACC3) is deregulated in a broad spectrum of cancers. In the present study, we reported that TACC3 was markedly elevated in bladder cancer, especially in muscle-invasive bladder cancers (MIBCs). The upregulation of TACC3 was positively associated with tumor invasiveness, grade, T stage, and progression in patients with bladder cancer. Furthermore, a Kaplan-Meier survival analysis showed that patients with bladder cancer whose tumors had high TACC3 expression experienced a dismal prognosis compared with patients whose tumors had low TACC3 expression. Functional studies have found that TACC3 is a prerequisite for the development of malignant characteristics of bladder cancer cells, including cell proliferation and invasion. Moreover, TACC3 promoted G1/S transition, which was mediated via activation of the transcription of E2F1, eventually enhancing cell proliferation. Notably, the overexpression of TACC3 or E2F1 indicates a high sensitivity to cisplatin. Taken together, these findings define a tumor-supportive role for TACC3, which may also serve as a prognostic and therapeutic indicator in bladder cancers.
Collapse
|
16
|
Sarkar S, Ryan EL, Royle SJ. FGFR3-TACC3 cancer gene fusions cause mitotic defects by removal of endogenous TACC3 from the mitotic spindle. Open Biol 2017; 7:170080. [PMID: 28855393 PMCID: PMC5577446 DOI: 10.1098/rsob.170080] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Accepted: 07/22/2017] [Indexed: 12/31/2022] Open
Abstract
Fibroblast growth factor receptor 3-transforming acidic coiled-coil containing protein 3 (FGFR3-TACC3; FT3) is a gene fusion resulting from rearrangement of chromosome 4 that has been identified in many cancers including those of the urinary bladder. Altered FGFR3 signalling in FT3-positive cells is thought to contribute to cancer progression. However, potential changes in TACC3 function in these cells have not been explored. TACC3 is a mitotic spindle protein required for accurate chromosome segregation. Errors in segregation lead to aneuploidy, which can contribute to cancer progression. Here we show that FT3-positive bladder cancer cells have lower levels of endogenous TACC3 on the mitotic spindle, and that this is sufficient to cause mitotic defects. FT3 is not localized to the mitotic spindle, and by virtue of its TACC domain, recruits endogenous TACC3 away from the spindle. Knockdown of the fusion gene or low-level overexpression of TACC3 partially rescues the chromosome segregation defects in FT3-positive bladder cancer cells. This function of FT3 is specific to TACC3 as inhibition of FGFR3 signalling does not rescue the TACC3 level on the spindle in these cancer cells. Models of FT3-mediated carcinogenesis should, therefore, include altered mitotic functions of TACC3 as well as altered FGFR3 signalling.
Collapse
Affiliation(s)
- Sourav Sarkar
- Centre for Mechanochemical Cell Biology, Warwick Medical School, University of Warwick, Gibbet Hill Road, Coventry, CV4 7AL, UK
| | - Ellis L Ryan
- Centre for Mechanochemical Cell Biology, Warwick Medical School, University of Warwick, Gibbet Hill Road, Coventry, CV4 7AL, UK
| | - Stephen J Royle
- Centre for Mechanochemical Cell Biology, Warwick Medical School, University of Warwick, Gibbet Hill Road, Coventry, CV4 7AL, UK
| |
Collapse
|
17
|
The myosin mesa and the basis of hypercontractility caused by hypertrophic cardiomyopathy mutations. Nat Struct Mol Biol 2017; 24:525-533. [PMID: 28481356 DOI: 10.1038/nsmb.3408] [Citation(s) in RCA: 158] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Accepted: 04/05/2017] [Indexed: 12/12/2022]
Abstract
Hypertrophic cardiomyopathy (HCM) is primarily caused by mutations in β-cardiac myosin and myosin-binding protein-C (MyBP-C). Changes in the contractile parameters of myosin measured so far do not explain the clinical hypercontractility caused by such mutations. We propose that hypercontractility is due to an increase in the number of myosin heads (S1) that are accessible for force production. In support of this hypothesis, we demonstrate myosin tail (S2)-dependent functional regulation of actin-activated human β-cardiac myosin ATPase. In addition, we show that both S2 and MyBP-C bind to S1 and that phosphorylation of either S1 or MyBP-C weakens these interactions. Importantly, the S1-S2 interaction is also weakened by four myosin HCM-causing mutations but not by two other mutations. To explain these experimental results, we propose a working structural model involving multiple interactions, including those with myosin's own S2 and MyBP-C, that hold myosin in a sequestered state.
Collapse
|
18
|
Burslem GM, Kyle HF, Nelson A, Edwards TA, Wilson AJ. Hypoxia inducible factor (HIF) as a model for studying inhibition of protein-protein interactions. Chem Sci 2017; 8:4188-4202. [PMID: 28878873 PMCID: PMC5576430 DOI: 10.1039/c7sc00388a] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Accepted: 03/31/2017] [Indexed: 12/21/2022] Open
Abstract
The state of the art in identifying protein–protein interaction inhibitors of hypoxia inducible factor – a promising target for anticancer drug design – is described.
The modulation of protein–protein interactions (PPIs) represents a major challenge in modern chemical biology. Current approaches (e.g. high-throughput screening, computer aided ligand design) are recognised as having limitations in terms of identification of hit matter. Considerable success has been achieved in terms of developing new approaches to PPI modulator discovery using the p53/hDM2 and Bcl-2 family of PPIs. However these important targets in oncology might be considered as “low-hanging-fruit”. Hypoxia inducible factor (HIF) is an emerging, but not yet fully validated target for cancer chemotherapy. Its role is to regulate the hypoxic response and it does so through a plethora of protein–protein interactions of varying topology, topography and complexity: its modulation represents an attractive approach to prevent development of new vasculature by hypoxic tumours.
Collapse
Affiliation(s)
- George M Burslem
- School of Chemistry , University of Leeds , Woodhouse Lane , Leeds LS2 9JT , UK . .,Astbury Centre for Structural Molecular Biology , University of Leeds , Woodhouse Lane , Leeds LS2 9JT , UK
| | - Hannah F Kyle
- Astbury Centre for Structural Molecular Biology , University of Leeds , Woodhouse Lane , Leeds LS2 9JT , UK.,School of Molecular and Cellular Biology , Faculty of Biological Sciences , University of Leeds , Woodhouse Lane , Leeds LS2 9JT , UK
| | - Adam Nelson
- School of Chemistry , University of Leeds , Woodhouse Lane , Leeds LS2 9JT , UK . .,Astbury Centre for Structural Molecular Biology , University of Leeds , Woodhouse Lane , Leeds LS2 9JT , UK
| | - Thomas A Edwards
- Astbury Centre for Structural Molecular Biology , University of Leeds , Woodhouse Lane , Leeds LS2 9JT , UK.,School of Molecular and Cellular Biology , Faculty of Biological Sciences , University of Leeds , Woodhouse Lane , Leeds LS2 9JT , UK
| | - Andrew J Wilson
- School of Chemistry , University of Leeds , Woodhouse Lane , Leeds LS2 9JT , UK . .,Astbury Centre for Structural Molecular Biology , University of Leeds , Woodhouse Lane , Leeds LS2 9JT , UK
| |
Collapse
|
19
|
Lasorella A, Sanson M, Iavarone A. FGFR-TACC gene fusions in human glioma. Neuro Oncol 2017; 19:475-483. [PMID: 27852792 PMCID: PMC5464372 DOI: 10.1093/neuonc/now240] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Accepted: 08/12/2016] [Indexed: 12/30/2022] Open
Abstract
Chromosomal translocations joining in-frame members of the fibroblast growth factor receptor-transforming acidic coiled-coil gene families (the FGFR-TACC gene fusions) were first discovered in human glioblastoma multiforme (GBM) and later in many other cancer types. Here, we review this rapidly expanding field of research and discuss the unique biological and clinical features conferred to isocitrate dehydrogenase wild-type glioma cells by FGFR-TACC fusions. FGFR-TACC fusions generate powerful oncogenes that combine growth-promoting effects with aneuploidy through the activation of as yet unclear intracellular signaling mechanisms. FGFR-TACC fusions appear to be clonal tumor-initiating events that confer strong sensitivity to FGFR tyrosine kinase inhibitors. Screening assays have recently been reported for the accurate identification of FGFR-TACC fusion variants in human cancer, and early clinical data have shown promising effects in cancer patients harboring FGFR-TACC fusions and treated with FGFR inhibitors. Thus, FGFR-TACC gene fusions provide a "low-hanging fruit" model for the validation of precision medicine paradigms in human GBM.
Collapse
Affiliation(s)
- Anna Lasorella
- Institute for Cancer Genetics, Department of Pediatrics and Pathology, Columbia University Medical Center, New York, New York, USA
| | - Marc Sanson
- Sorbonne Universités UPMC Univ Paris 06, INSERM CNRS, U1127, UMR 7225, ICM, F-75013,Groupe Hospitalier Pitié-Salpêtrière, Service de Neurologie 2, Paris, France
| | - Antonio Iavarone
- Institute for Cancer Genetics, Department of Neurology and Pathology, Columbia University Medical Center, New York, New York, USA
| |
Collapse
|
20
|
Sharma AK, Birrane G, Anklin C, Rigby AC, Alper SL. NMR insight into myosin-binding subunit coiled-coil structure reveals binding interface with protein kinase G-Iα leucine zipper in vascular function. J Biol Chem 2017; 292:7052-7065. [PMID: 28280239 DOI: 10.1074/jbc.m117.781260] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Revised: 03/08/2017] [Indexed: 11/06/2022] Open
Abstract
Nitrovasodilators relax vascular smooth-muscle cells in part by modulating the interaction of the C-terminal coiled-coil domain (CC) and/or the leucine zipper (LZ) domain of the myosin light-chain phosphatase component, myosin-binding subunit (MBS), with the N-terminal LZ domain of protein kinase G (PKG)-Iα. Despite the importance of vasodilation in cardiovascular homeostasis and therapy, our structural understanding of the MBS CC interaction with LZ PKG-1α has remained limited. Here, we report the 3D NMR solution structure of homodimeric CC MBS in which amino acids 932-967 form a coiled-coil of two monomeric α-helices in parallel orientation. We found that the structure is stabilized by non-covalent interactions, with dominant contributions from hydrophobic residues at a and d heptad positions. Using NMR chemical-shift perturbation (CSP) analysis, we identified a subset of hydrophobic and charged residues of CC MBS (localized within and adjacent to the C-terminal region) contributing to the dimer-dimer interaction interface between homodimeric CC MBS and homodimeric LZ PKG-Iα. 15N backbone relaxation NMR revealed the dynamic features of the CC MBS interface residues identified by NMR CSP. Paramagnetic relaxation enhancement- and CSP-NMR-guided HADDOCK modeling of the dimer-dimer interface of the heterotetrameric complex exhibits the involvement of non-covalent intermolecular interactions that are localized within and adjacent to the C-terminal regions of each homodimer. These results deepen our understanding of the binding restraints of this CC MBS·LZ PKG-Iα low-affinity heterotetrameric complex and allow reevaluation of the role(s) of myosin light-chain phosphatase partner polypeptides in regulation of vascular smooth-muscle cell contractility.
Collapse
Affiliation(s)
- Alok K Sharma
- From the Division of Nephrology and Center for Vascular Biology Research, .,the Department of Medicine, Harvard Medical School, Boston, Massachusetts 02215
| | - Gabriel Birrane
- the Department of Medicine, Harvard Medical School, Boston, Massachusetts 02215.,Division of Experimental Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - Clemens Anklin
- Bruker Biospin Corp., Billerica, Massachusetts 01821, and
| | - Alan C Rigby
- Warp Drive Bio, Inc., Cambridge, Massachusetts 02139
| | - Seth L Alper
- From the Division of Nephrology and Center for Vascular Biology Research, .,the Department of Medicine, Harvard Medical School, Boston, Massachusetts 02215
| |
Collapse
|
21
|
Rutherford EL, Lowery LA. Exploring the developmental mechanisms underlying Wolf-Hirschhorn Syndrome: Evidence for defects in neural crest cell migration. Dev Biol 2016; 420:1-10. [PMID: 27777068 PMCID: PMC5193094 DOI: 10.1016/j.ydbio.2016.10.012] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Revised: 10/03/2016] [Accepted: 10/18/2016] [Indexed: 01/20/2023]
Abstract
Wolf-Hirschhorn Syndrome (WHS) is a neurodevelopmental disorder characterized by mental retardation, craniofacial malformation, and defects in skeletal and heart development. The syndrome is associated with irregularities on the short arm of chromosome 4, including deletions of varying sizes and microduplications. Many of these genotypic aberrations in humans have been correlated with the classic WHS phenotype, and animal models have provided a context for mapping these genetic irregularities to specific phenotypes; however, there remains a significant knowledge gap concerning the cell biological mechanisms underlying these phenotypes. This review summarizes literature that has made recent contributions to this topic, drawing from the vast body of knowledge detailing the genetic particularities of the disorder and the more limited pool of information on its cell biology. Finally, we propose a novel characterization for WHS as a pathophysiology owing in part to defects in neural crest cell motility and migration during development.
Collapse
Affiliation(s)
- Erin L Rutherford
- Boston College, Department of Biology, 140 Commonwealth Avenue, Chestnut Hill, MA 02467, United States
| | - Laura Anne Lowery
- Boston College, Department of Biology, 140 Commonwealth Avenue, Chestnut Hill, MA 02467, United States.
| |
Collapse
|
22
|
Structural integration in hypoxia-inducible factors. Nature 2015; 524:303-8. [PMID: 26245371 DOI: 10.1038/nature14883] [Citation(s) in RCA: 244] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2015] [Accepted: 07/07/2015] [Indexed: 12/27/2022]
Abstract
The hypoxia-inducible factors (HIFs) coordinate cellular adaptations to low oxygen stress by regulating transcriptional programs in erythropoiesis, angiogenesis and metabolism. These programs promote the growth and progression of many tumours, making HIFs attractive anticancer targets. Transcriptionally active HIFs consist of HIF-α and ARNT (also called HIF-1β) subunits. Here we describe crystal structures for each of mouse HIF-2α-ARNT and HIF-1α-ARNT heterodimers in states that include bound small molecules and their hypoxia response element. A highly integrated quaternary architecture is shared by HIF-2α-ARNT and HIF-1α-ARNT, wherein ARNT spirals around the outside of each HIF-α subunit. Five distinct pockets are observed that permit small-molecule binding, including PAS domain encapsulated sites and an interfacial cavity formed through subunit heterodimerization. The DNA-reading head rotates, extends and cooperates with a distal PAS domain to bind hypoxia response elements. HIF-α mutations linked to human cancers map to sensitive sites that establish DNA binding and the stability of PAS domains and pockets.
Collapse
|
23
|
Scheuermann TH, Stroud D, Sleet CE, Bayeh L, Shokri C, Wang H, Caldwell CG, Longgood J, MacMillan JB, Bruick RK, Gardner KH, Tambar UK. Isoform-Selective and Stereoselective Inhibition of Hypoxia Inducible Factor-2. J Med Chem 2015. [DOI: 10.1021/acs.jmedchem.5b00529] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Thomas H. Scheuermann
- Department
of Biophysics, University of Texas Southwestern Medical Center, 5323
Harry Hines Boulevard, Dallas, Texas 75390, United States
| | - Daniel Stroud
- Department
of Biochemistry, University of Texas Southwestern Medical Center, 5323
Harry Hines Boulevard, Dallas, Texas 75390, United States
| | - Christopher E. Sleet
- Department
of Biochemistry, University of Texas Southwestern Medical Center, 5323
Harry Hines Boulevard, Dallas, Texas 75390, United States
| | - Liela Bayeh
- Department
of Biochemistry, University of Texas Southwestern Medical Center, 5323
Harry Hines Boulevard, Dallas, Texas 75390, United States
| | - Cameron Shokri
- Department
of Biochemistry, University of Texas Southwestern Medical Center, 5323
Harry Hines Boulevard, Dallas, Texas 75390, United States
| | - Hanzhi Wang
- Department
of Biochemistry, University of Texas Southwestern Medical Center, 5323
Harry Hines Boulevard, Dallas, Texas 75390, United States
| | - Charles G. Caldwell
- Department
of Biochemistry, University of Texas Southwestern Medical Center, 5323
Harry Hines Boulevard, Dallas, Texas 75390, United States
| | - Jamie Longgood
- Department
of Biochemistry, University of Texas Southwestern Medical Center, 5323
Harry Hines Boulevard, Dallas, Texas 75390, United States
| | - John B. MacMillan
- Department
of Biochemistry, University of Texas Southwestern Medical Center, 5323
Harry Hines Boulevard, Dallas, Texas 75390, United States
| | - Richard K. Bruick
- Department
of Biochemistry, University of Texas Southwestern Medical Center, 5323
Harry Hines Boulevard, Dallas, Texas 75390, United States
| | - Kevin H. Gardner
- Department
of Biophysics, University of Texas Southwestern Medical Center, 5323
Harry Hines Boulevard, Dallas, Texas 75390, United States
- Structural
Biology Initiative, CUNY Advanced Science Research Center, New York, New York 10031, United States
- Department
of Chemistry and Biochemistry, City College of New York, New York, New York 10031, United States
| | - Uttam K. Tambar
- Department
of Biochemistry, University of Texas Southwestern Medical Center, 5323
Harry Hines Boulevard, Dallas, Texas 75390, United States
| |
Collapse
|