1
|
Zimmermann RC, Sardiu ME, Manton CA, Miah MS, Banks CAS, Adams MK, Koestler DC, Hurst DR, Edmonds MD, Washburn MP, Welch DR. Perturbation of BRMS1 interactome reveals pathways that impact metastasis. PLoS One 2021; 16:e0259128. [PMID: 34788285 PMCID: PMC8598058 DOI: 10.1371/journal.pone.0259128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 10/12/2021] [Indexed: 11/25/2022] Open
Abstract
Breast Cancer Metastasis Suppressor 1 (BRMS1) expression is associated with longer patient survival in multiple cancer types. Understanding BRMS1 functionality will provide insights into both mechanism of action and will enhance potential therapeutic development. In this study, we confirmed that the C-terminus of BRMS1 is critical for metastasis suppression and hypothesized that critical protein interactions in this region would explain its function. Phosphorylation status at S237 regulates BRMS1 protein interactions related to a variety of biological processes, phenotypes [cell cycle (e.g., CDKN2A), DNA repair (e.g., BRCA1)], and metastasis [(e.g., TCF2 and POLE2)]. Presence of S237 also directly decreased MDA-MB-231 breast carcinoma migration in vitro and metastases in vivo. The results add significantly to our understanding of how BRMS1 interactions with Sin3/HDAC complexes regulate metastasis and expand insights into BRMS1's molecular role, as they demonstrate BRMS1 C-terminus involvement in distinct protein-protein interactions.
Collapse
Affiliation(s)
- Rosalyn C. Zimmermann
- Department of Cancer Biology, The Kansas University Medical Center, Kansas City, KS, United States of America
| | - Mihaela E. Sardiu
- Stowers Institute for Medical Research, Kansas City, Missouri, United States of America
- Department of Biostatistics and Data Science, The Kansas University Medical Center, Kansas City, KS, United States of America
- The University of Kansas Cancer Center, Kansas City, KS, United States of America
| | - Christa A. Manton
- Department of Cancer Biology, The Kansas University Medical Center, Kansas City, KS, United States of America
- Pathology Department, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
- Department of Biology, Baker University, Baldwin City, KS, United States of America
| | - Md. Sayem Miah
- Stowers Institute for Medical Research, Kansas City, Missouri, United States of America
- Department of Biochemistry and Molecular Biology, University of Arkansas for Health Sciences, Little Rock, AR, United States of America
| | - Charles A. S. Banks
- Stowers Institute for Medical Research, Kansas City, Missouri, United States of America
| | - Mark K. Adams
- Stowers Institute for Medical Research, Kansas City, Missouri, United States of America
| | - Devin C. Koestler
- Department of Biostatistics and Data Science, The Kansas University Medical Center, Kansas City, KS, United States of America
- The University of Kansas Cancer Center, Kansas City, KS, United States of America
| | - Douglas R. Hurst
- Pathology Department, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Mick D. Edmonds
- Department of Genetics, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Michael P. Washburn
- Department of Cancer Biology, The Kansas University Medical Center, Kansas City, KS, United States of America
- Stowers Institute for Medical Research, Kansas City, Missouri, United States of America
- The University of Kansas Cancer Center, Kansas City, KS, United States of America
| | - Danny R. Welch
- Department of Cancer Biology, The Kansas University Medical Center, Kansas City, KS, United States of America
- The University of Kansas Cancer Center, Kansas City, KS, United States of America
| |
Collapse
|
2
|
Kaczmarek JV, Bogan CM, Pierce JM, Tao YK, Chen SC, Liu Q, Liu X, Boyd KL, Calcutt MW, Bridges TM, Lindsley CW, Friedman DL, Richmond A, Daniels AB. Intravitreal HDAC Inhibitor Belinostat Effectively Eradicates Vitreous Seeds Without Retinal Toxicity In Vivo in a Rabbit Retinoblastoma Model. Invest Ophthalmol Vis Sci 2021; 62:8. [PMID: 34757417 PMCID: PMC8590161 DOI: 10.1167/iovs.62.14.8] [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] [Indexed: 11/30/2022] Open
Abstract
Purpose Current melphalan-based regimens for intravitreal chemotherapy for retinoblastoma vitreous seeds are effective but toxic to the retina. Thus, alternative agents are needed. Based on the known biology of histone deacetylases (HDACs) in the retinoblastoma pathway, we systematically studied whether the HDAC inhibitor belinostat is a viable, molecularly targeted alternative agent for intravitreal delivery that might provide comparable efficacy, without toxicity. Methods In vivo pharmacokinetic experiments in rabbits and in vitro cytotoxicity experiments were performed to determine the 90% inhibitory concentration (IC90). Functional toxicity by electroretinography and structural toxicity by optical coherence tomography (OCT), OCT angiography, and histopathology were evaluated in rabbits following three injections of belinostat 350 µg (2× IC90) or 700 µg (4× IC90), compared with melphalan 12.5 µg (rabbit equivalent of the human dose). The relative efficacy of intravitreal belinostat versus melphalan to treat WERI-Rb1 human cell xenografts in rabbit eyes was directly quantified. RNA sequencing was used to assess belinostat-induced changes in RB cell gene expression. Results The maximum nontoxic dose of belinostat was 350 µg, which caused no reductions in electroretinography parameters, retinal microvascular loss on OCT angiography, or retinal degeneration. Melphalan caused severe retinal structural and functional toxicity. Belinostat 350 µg (equivalent to 700 µg in the larger human eye) was equally effective at eradicating vitreous seeds in the rabbit xenograft model compared with melphalan (95.5% reduction for belinostat, P < 0.001; 89.4% reduction for melphalan, P < 0.001; belinostat vs. melphalan, P = 0.10). Even 700 µg belinostat (equivalent to 1400 µg in humans) caused only minimal toxicity. Widespread changes in gene expression resulted. Conclusions Molecularly targeted inhibition of HDACs with intravitreal belinostat was equally effective as standard-of-care melphalan but without retinal toxicity. Belinostat may therefore be an attractive agent to pursue clinically for intravitreal treatment of retinoblastoma.
Collapse
Affiliation(s)
- Jessica V Kaczmarek
- Division of Ocular Oncology and Pathology, Department of Ophthalmology and Visual Sciences, Vanderbilt University Medical Center, Nashville, Tennessee, United States
| | - Carley M Bogan
- Division of Ocular Oncology and Pathology, Department of Ophthalmology and Visual Sciences, Vanderbilt University Medical Center, Nashville, Tennessee, United States
| | - Janene M Pierce
- Division of Ocular Oncology and Pathology, Department of Ophthalmology and Visual Sciences, Vanderbilt University Medical Center, Nashville, Tennessee, United States
| | - Yuankai K Tao
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee, United States
| | - Sheau-Chiann Chen
- Center for Quantitative Sciences, Vanderbilt University Medical Center, Nashville, Tennessee, United States.,Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee, United States
| | - Qi Liu
- Center for Quantitative Sciences, Vanderbilt University Medical Center, Nashville, Tennessee, United States.,Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee, United States
| | - Xiao Liu
- Center for Quantitative Sciences, Vanderbilt University Medical Center, Nashville, Tennessee, United States.,Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee, United States
| | - Kelli L Boyd
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee, United States.,Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, United States
| | - M Wade Calcutt
- Department of Biochemistry, Vanderbilt University, Nashville, Tennessee, United States
| | - Thomas M Bridges
- Warren Center for Neuroscience Drug Discovery at Vanderbilt, Department of Pharmacology, Vanderbilt University, Nashville, Tennessee, United States
| | - Craig W Lindsley
- Warren Center for Neuroscience Drug Discovery at Vanderbilt, Department of Pharmacology, Vanderbilt University, Nashville, Tennessee, United States
| | - Debra L Friedman
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee, United States.,Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee, United States
| | - Ann Richmond
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee, United States.,Tennessee Valley Healthcare System, Department of Veterans Affairs, Nashville, Tennessee, United States.,Department of Pharmacology, Vanderbilt University, Nashville, Tennessee, United States.,Program in Cancer Biology, Vanderbilt University, Nashville, Tennessee, United States
| | - Anthony B Daniels
- Division of Ocular Oncology and Pathology, Department of Ophthalmology and Visual Sciences, Vanderbilt University Medical Center, Nashville, Tennessee, United States.,Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee, United States.,Program in Cancer Biology, Vanderbilt University, Nashville, Tennessee, United States.,Department of Radiation Oncology, Vanderbilt University Medical Center, Nashville, Tennessee, United States
| |
Collapse
|
3
|
Singh A, Paul MS, Dutta D, Mutsuddi M, Mukherjee A. Regulation of notch signaling by a chromatin modeling protein Hat-trick. Development 2019; 146:dev.170837. [DOI: 10.1242/dev.170837] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2018] [Accepted: 05/16/2019] [Indexed: 01/08/2023]
Abstract
Notch signaling plays pleiotropic role in astounding variety of cellular processes including cell fate determination, differentiation, proliferation and apoptosis. The increasingly complex regulatory mechanisms of Notch signaling account for the multitude of functions exhibited by Notch during development. We identified Hat-trick (Htk), a DNA binding protein, as an interacting partner of Notch-ICD in a yeast two-hybrid screen and their physical interaction was further validated by co-immunoprecipitation experiments. htk genetically interacts with Notch pathway components in trans-heterozygous combinations. Loss of htk function in htk mutant somatic clones showed down-regulation of Notch targets, whereas over-expression of htk caused ectopic expression of Notch target, without affecting the level of Notch protein. Immunocytochemical analysis has demonstrated that Htk co-localizes with over-expressed Notch-ICD in the same nuclear compartment. We have shown here that Htk cooperates with Notch-ICD and Suppressor of Hairless to form activation complex and binds to the regulatory sequences of Notch downstream targets, Enhancer of Split complex genes to direct their expression. Taken together, our results suggest a novel mode of regulation of Notch signaling by a chromatin modeling protein Htk.
Collapse
Affiliation(s)
- Ankita Singh
- Department of Molecular and Human Genetics, Institute of Science, Banaras Hindu University, Varanasi-221005, Uttar Pradesh, India
| | - Maimuna S. Paul
- Department of Molecular and Human Genetics, Institute of Science, Banaras Hindu University, Varanasi-221005, Uttar Pradesh, India
| | - Debdeep Dutta
- Department of Molecular and Human Genetics, Institute of Science, Banaras Hindu University, Varanasi-221005, Uttar Pradesh, India
| | - Mousumi Mutsuddi
- Department of Molecular and Human Genetics, Institute of Science, Banaras Hindu University, Varanasi-221005, Uttar Pradesh, India
| | - Ashim Mukherjee
- Department of Molecular and Human Genetics, Institute of Science, Banaras Hindu University, Varanasi-221005, Uttar Pradesh, India
| |
Collapse
|
4
|
Li Y, Sun W, Han N, Zou Y, Yin D. Curcumin inhibits proliferation, migration, invasion and promotes apoptosis of retinoblastoma cell lines through modulation of miR-99a and JAK/STAT pathway. BMC Cancer 2018; 18:1230. [PMID: 30526546 PMCID: PMC6288931 DOI: 10.1186/s12885-018-5130-y] [Citation(s) in RCA: 82] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Accepted: 11/26/2018] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Curcumin, a primary active ingredient extracted from the Curcuma longa, has been recently identified as a potential anti-tumor agent in multiple kinds of cancers. However, the effect of curcumin on retinoblastoma (Rb) is still unclear. Therefore, we attempted to reveal the functional impacts and the underlying mechanisms of curcumin in Rb cells. METHODS Two Rb cell lines SO-Rb50 and Y79 were pre-treated with various doses of curcumin, and then cell proliferation, apoptosis, migration, and invasion were assessed, respectively. Further, regulatory effects of curcumin on miR-99a expression, as well as the activation of JAK/STAT pathway were studied. RESULTS Data showed that curcumin significantly inhibited the viability, colony formation capacity, migration and invasion, while induced apoptosis of SO-Rb50 and Y79 cells. Up-regulation of miR-99a was observed in curcumin-treated cells. Curcumin suppressed the phosphorylation levels of JAK1, STAT1, and STAT3, while curcumin did not inhibit the activation of JAK/STAT pathway when miR-99a was knocked down. CONCLUSION Curcumin inhibited proliferation, migration, invasion, but promoted apoptosis of Rb cells. The anti-tumor activities of curcumin on Rb cells appeared to be via up-regulation of miR-99a, and thereby inhibition of JAK/STAT pathway.
Collapse
Affiliation(s)
- Yaping Li
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun, Jilin, 130041 China
| | - Weixuan Sun
- Department of Gastroenterological Surgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin, 130033 China
| | - Ning Han
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun, Jilin, 130041 China
| | - Ying Zou
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun, Jilin, 130041 China
| | - Dexin Yin
- Department of Vascular Surgery, China-Japan Union Hospital of Jilin University, No.126, Xiantai Street, Changchun, Jilin, 130033 China
| |
Collapse
|
5
|
Roesley SNA, La Marca JE, Deans AJ, Mckenzie L, Suryadinata R, Burke P, Portela M, Wang H, Bernard O, Sarcevic B, Richardson HE. Phosphorylation of Drosophila Brahma on CDK-phosphorylation sites is important for cell cycle regulation and differentiation. Cell Cycle 2018; 17:1559-1578. [PMID: 29963966 DOI: 10.1080/15384101.2018.1493414] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
The SWI/SNF ATP-dependent chromatin-remodeling complex is an important evolutionarily conserved regulator of cell cycle progression. It associates with the Retinoblastoma (pRb)/HDAC/E2F/DP transcription complex to modulate cell cycle-dependent gene expression. The key catalytic component of the SWI/SNF complex in mammals is the ATPase subunit, Brahma (BRM) or BRG1. BRG1 was previously shown to be phosphorylated by the G1-S phase cell cycle regulatory kinase Cyclin E/CDK2 in vitro, which was associated with the bypass of G1 arrest conferred by BRG1 expression. However, it is unknown whether direct Cyclin E/CDK2-mediated phosphorylation of BRM/BRG1 is important for G1-S phase cell cycle progression and proliferation in vivo. Herein, we demonstrate for the first time the importance of CDK-mediated phosphorylation of Brm in cell proliferation and differentiation in vivo using the Drosophila melanogaster model organism. Expression of a CDK-site phospho-mimic mutant of Brm, brm-ASP (all the potential CDK sites are mutated from Ser/Thr to Asp), which acts genetically as a brm loss-of-function allele, dominantly accelerates progression into the S phase, and bypasses a Retinoblastoma-induced developmental G1 phase arrest in the wing epithelium. Conversely, expression of a CDK-site phospho-blocking mutation of Brm, brm-ALA, acts genetically as a brm gain-of-function mutation, and in a Brm complex compromised background reduces S phase cells. Expression of the brm phospho-mutants also affected differentiation and Decapentaplegic (BMP/TGFβ) signaling in the wing epithelium. Altogether our results show that CDK-mediated phosphorylation of Brm is important in G1-S phase regulation and differentiation in vivo. ABBREVIATIONS A-P: Anterior-Posterior; BAF: BRG1-associated factor; BMP: Bone Morphogenetic Protein; Brg1: Brahma-Related Gene 1; Brm: Brahma; BSA: Bovine Serum Albumin; CDK: Cyclin dependent kinase dpp: decapentaplegic; EdU: 5-Ethynyl 2'-DeoxyUridine; EGFR: Epidermal Growth Factor Receptor; en: engrailed; GFP: Green Fluorescent Protein; GST: Glutathione-S-Transferase; HDAC: Histone DeACetylase; JNK: c-Jun N-terminal Kinase; Mad: Mothers Against Dpp; MAPK: Mitogen Activated Protein Kinase; MB:: Myelin Basic Protein; nub: nubbin; pH3: phosphorylated Histone H3; PBS: Phosphate Buffered Saline; PBT: PBS Triton; PFA: ParaFormAldehydep; Rb: Retinoblastoma protein; PCV: Posterior Cross-Vein; Snr1: Snf5-Related 1; SWI/SNF: SWitch/Sucrose Non-Fermentable; TGFβ: Transforming Growth Factor β; TUNEL: TdT-mediated dUTP Nick End Labelling; Wg: Wingless; ZNC: Zone of Non-Proliferating Cells.
Collapse
Affiliation(s)
- Siti Nur Ain Roesley
- a Cell Cycle and Cancer Unit , St Vincent's Medical Institute , Melbourne , Australia.,b Department of Medicine , University of Melbourne , Melbourne , Australia.,c Cell Cycle & Development Laboratory , Peter MacCallum Cancer Centre , Melbourne , Australia
| | - John E La Marca
- d Department of Biochemistry & Genetics , La Trobe Institute for Molecular Science, La Trobe University , Melbourne , Australia
| | - Andrew J Deans
- b Department of Medicine , University of Melbourne , Melbourne , Australia.,e Genome Stability Unit , St Vincent's Medical Institute , Melbourne , Australia
| | - Lisa Mckenzie
- c Cell Cycle & Development Laboratory , Peter MacCallum Cancer Centre , Melbourne , Australia
| | - Randy Suryadinata
- a Cell Cycle and Cancer Unit , St Vincent's Medical Institute , Melbourne , Australia.,b Department of Medicine , University of Melbourne , Melbourne , Australia
| | - Peter Burke
- c Cell Cycle & Development Laboratory , Peter MacCallum Cancer Centre , Melbourne , Australia.,d Department of Biochemistry & Genetics , La Trobe Institute for Molecular Science, La Trobe University , Melbourne , Australia
| | - Marta Portela
- c Cell Cycle & Development Laboratory , Peter MacCallum Cancer Centre , Melbourne , Australia.,d Department of Biochemistry & Genetics , La Trobe Institute for Molecular Science, La Trobe University , Melbourne , Australia
| | - Hongyan Wang
- f Neuroscience and Behavioral Disorders Program , Duke-National University of Singapore Graduate Medical School , Singapore.,g National University of Singapore Graduate School for Integrative Sciences and Engineering , National University of Singapore , Singapore.,h Department of Physiology , Yong Loo Lin School of Medicine, National University of Singapore , Singapore
| | - Ora Bernard
- b Department of Medicine , University of Melbourne , Melbourne , Australia
| | - Boris Sarcevic
- a Cell Cycle and Cancer Unit , St Vincent's Medical Institute , Melbourne , Australia.,b Department of Medicine , University of Melbourne , Melbourne , Australia
| | - Helena E Richardson
- d Department of Biochemistry & Genetics , La Trobe Institute for Molecular Science, La Trobe University , Melbourne , Australia.,e Genome Stability Unit , St Vincent's Medical Institute , Melbourne , Australia.,i Peter MacCallum Department of Oncology, Department of Biochemistry & Molecular Biology, Department of Anatomy & Neuroscience , University of Melbourne , Melbourne , Australia
| |
Collapse
|
6
|
The Temporal Regulation of S Phase Proteins During G 1. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1042:335-369. [PMID: 29357066 DOI: 10.1007/978-981-10-6955-0_16] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Successful DNA replication requires intimate coordination with cell-cycle progression. Prior to DNA replication initiation in S phase, a series of essential preparatory events in G1 phase ensures timely, complete, and precise genome duplication. Among the essential molecular processes are regulated transcriptional upregulation of genes that encode replication proteins, appropriate post-transcriptional control of replication factor abundance and activity, and assembly of DNA-loaded protein complexes to license replication origins. In this chapter we describe these critical G1 events necessary for DNA replication and their regulation in the context of both cell-cycle entry and cell-cycle progression.
Collapse
|
7
|
Snezhkina AV, Nyushko KM, Zaretsky AR, Shagin DA, Sadritdinova AF, Fedorova MS, Guvatova ZG, Abramov IS, Pudova EA, Alekseev BY, Dmitriev AA, Kudryavtseva AV. Transcription Factor SAP30 Is Involved in the Activation of NETO2 Gene Expression in Clear Cell Renal Cell Carcinoma. Mol Biol 2018. [DOI: 10.1134/s0026893318020152] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
8
|
Berrak O, Arisan ED, Obakan-Yerlikaya P, Coker-Gürkan A, Palavan-Unsal N. mTOR is a fine tuning molecule in CDK inhibitors-induced distinct cell death mechanisms via PI3K/AKT/mTOR signaling axis in prostate cancer cells. Apoptosis 2018; 21:1158-78. [PMID: 27484210 DOI: 10.1007/s10495-016-1275-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Purvalanol and roscovitine are cyclin dependent kinase (CDK) inhibitors that induce cell cycle arrest and apoptosis in various cancer cells. We further hypothesized that co-treatment of CDK inhibitors with rapamycin, an mTOR inhibitor, would be an effective combinatory strategy for the inhibition of prostate cancer regard to androgen receptor (AR) status due to inhibition of proliferative pathway, PI3K/AKT/mTOR, and induction of cell death mechanisms. Androgen responsive (AR+), PTEN(-/-) LNCaP and androgen independent (AR-), PTEN(+/-) DU145 prostate cancer cells were exposed to purvalanol (20 µM) and roscovitine (30 µM) with or without rapamycin for 24 h. Cell viability assay, immunoblotting, flow cytometry and fluorescence microscopy was used to define the effect of CDK inhibitors with or without rapamycin on proliferative pathway and cell death mechanisms in LNCaP and DU145 prostate cancer cells. Co-treatment of rapamycin modulated CDK inhibitors-induced cytotoxicity and apoptosis that CDK inhibitors were more potent to induce cell death in AR (+) LNCaP cells than AR (-) DU145 cells. CDK inhibitors in the presence or absence of rapamycin induced cell death via modulating upstream PI3K/AKT/mTOR signaling pathway in LNCaP cells, exclusively only treatment of purvalanol have strong potential to inhibit both upstream and downstream targets of mTOR in LNCaP and DU145 cells. However, co-treatment of rapamycin with CDK inhibitors protects DU145 cells from apoptosis via induction of autophagy mechanism. We confirmed that purvalanol and roscovitine were strong apoptotic and autophagy inducers that based on regulation of PI3K/AKT/mTOR signaling pathway. Co-treatment of rapamycin with purvalanol and roscovitine exerted different effects on cell survival and death mechanisms in LNCaP and DU145 cell due to their AR receptor status. Our studies show that co-treatment of rapamycin with CDK inhibitors inhibit prostate cancer cell viability more effectively than either agent alone, in part, by targeting the mTOR signaling cascade in AR (+) LNCaP cells. In this point, mTOR is a fine-tuning player in purvalanol and roscovitine-induced apoptosis and autophagy via regulation of PI3K/AKT and the downstream targets, which related with cell proliferation.
Collapse
Affiliation(s)
- Ozge Berrak
- Molecular Biology and Genetics Department, Science and Literature Faculty, Istanbul Kultur University, Atakoy Campus, 34156, Istanbul, Turkey
| | - Elif Damla Arisan
- Molecular Biology and Genetics Department, Science and Literature Faculty, Istanbul Kultur University, Atakoy Campus, 34156, Istanbul, Turkey.
| | - Pinar Obakan-Yerlikaya
- Molecular Biology and Genetics Department, Science and Literature Faculty, Istanbul Kultur University, Atakoy Campus, 34156, Istanbul, Turkey
| | - Ajda Coker-Gürkan
- Molecular Biology and Genetics Department, Science and Literature Faculty, Istanbul Kultur University, Atakoy Campus, 34156, Istanbul, Turkey
| | - Narçin Palavan-Unsal
- Molecular Biology and Genetics Department, Science and Literature Faculty, Istanbul Kultur University, Atakoy Campus, 34156, Istanbul, Turkey
| |
Collapse
|
9
|
Cantor DJ, David G. The potential of targeting Sin3B and its associated complexes for cancer therapy. Expert Opin Ther Targets 2017; 21:1051-1061. [PMID: 28956957 DOI: 10.1080/14728222.2017.1386655] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
INTRODUCTION Sin3B serves as a scaffold for chromatin-modifying complexes that repress gene transcription to regulate distinct biological processes. Sin3B-containing complexes are critical for cell cycle withdrawal, and abrogation of Sin3B-dependent cell cycle exit impacts tumor progression. Areas covered: In this review, we discuss the biochemical characteristics of Sin3B-containing complexes and explore how these complexes regulate gene transcription. We focus on how Sin3B-containing complexes, through the association of the Rb family of proteins, repress the expression of E2F target genes during quiescence, differentiation, and senescence. Finally, we speculate on the potential benefits of the inhibition of Sin3B-containing complexes for the treatment of cancer. Expert opinion: Further identification and characterization of specific Sin3B-containing complexes provide a unique opportunity to prevent the pro-tumorigenic effects of the senescence-associated secretory phenotype, and to abrogate cancer stem cell quiescence and the associated resistance to therapy.
Collapse
Affiliation(s)
- David J Cantor
- a Department of Biochemistry and Molecular Pharmacology , New York University School of Medicine , New York , NY , USA
| | - Gregory David
- a Department of Biochemistry and Molecular Pharmacology , New York University School of Medicine , New York , NY , USA.,b Department of Urology.,c NYU Cancer Institute , New York University School of Medicine , New York , NY , USA
| |
Collapse
|
10
|
Li J, Vervoorts J, Carloni P, Rossetti G, Lüscher B. Structural prediction of the interaction of the tumor suppressor p27 KIP1 with cyclin A/CDK2 identifies a novel catalytically relevant determinant. BMC Bioinformatics 2017; 18:15. [PMID: 28056778 PMCID: PMC5217639 DOI: 10.1186/s12859-016-1411-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Accepted: 12/07/2016] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The cyclin-dependent kinase 2 (CDK2) together with its cyclin E and A partners is a central regulator of cell growth and division. Deregulation of CDK2 activity is associated with diseases such as cancer. The analysis of substrates identified S/T-P-X-R/K/H as the CDK2 consensus sequence. The crystal structure of cyclin A/CDK2 with a short model peptide supports this sequence and identifies key interactions. However, CDKs use additional determinants to recognize substrates, including the RXL motif that is read by the cyclin subunits. We were interested to determine whether additional amino acids beyond the minimal consensus sequence of the well-studied substrate and tumor suppressor p27KIP1 were relevant for catalysis. RESULTS To address whether additional amino acids, close to the minimal consensus sequence, play a role in binding, we investigate the interaction of cyclin A/CDK2 with an in vivo cellular partner and CDK inhibitor p27KIP1. This protein is an intrinsically unfolded protein and, in particular, the C-terminal half of the protein has not been accessible to structural analysis. This part harbors the CDK2 phosphorylation site. We used bioinformatics tools, including MODELLER, iTASSER and HADDOCK, along with partial structural information to build a model of the C-terminal region of p27KIP1 with cyclin A/CDK2. This revealed novel interactions beyond the consensus sequence with a proline and a basic amino acid at the P + 1 and the P + 3 sites, respectively. We suggest that the lysine at P + 2 might regulate the reversible association of the second counter ion in the active site of CDK2. The arginine at P + 7 interacts with both cyclin A and CDK2 and is important for the catalytic turnover rate. CONCLUSION Our modeling identifies additional amino acids in p27KIP1 beyond the consensus sequence that contribute to the efficiency of substrate phosphorylation.
Collapse
Affiliation(s)
- Jinyu Li
- College of Chemistry, Fuzhou University, Fuzhou, 350002, China.,Institute of Biochemistry and Molecular Biology, Medical School, RWTH Aachen University, 52057, Aachen, Germany.,Computational Biomedicine, Institute for Advanced Simulation IAS-5 and Institute of Neuroscience and Medicine INM-9, Forschungszentrum Jülich, 52425, Jülich, Germany
| | - Jörg Vervoorts
- Institute of Biochemistry and Molecular Biology, Medical School, RWTH Aachen University, 52057, Aachen, Germany
| | - Paolo Carloni
- Computational Biomedicine, Institute for Advanced Simulation IAS-5 and Institute of Neuroscience and Medicine INM-9, Forschungszentrum Jülich, 52425, Jülich, Germany
| | - Giulia Rossetti
- Computational Biomedicine, Institute for Advanced Simulation IAS-5 and Institute of Neuroscience and Medicine INM-9, Forschungszentrum Jülich, 52425, Jülich, Germany. .,Department of Oncology, Hematology and Stem Cell Transplantation, Medical School, RWTH Aachen University, Aachen, Germany. .,Jülich Supercomputing Centre (JSC), Forschungszentrum Jülich, 52425, Jülich, Germany.
| | - Bernhard Lüscher
- Institute of Biochemistry and Molecular Biology, Medical School, RWTH Aachen University, 52057, Aachen, Germany.
| |
Collapse
|
11
|
Roesley SNA, Suryadinata R, Morrish E, Tan AR, Issa SMA, Oakhill JS, Bernard O, Welch DR, Šarčević B. Cyclin-dependent kinase-mediated phosphorylation of breast cancer metastasis suppressor 1 (BRMS1) affects cell migration. Cell Cycle 2016; 15:137-51. [PMID: 26771717 DOI: 10.1080/15384101.2015.1121328] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Expression of Breast Cancer Metastasis Suppressor 1 (BRMS1) reduces the incidence of metastasis in many human cancers, without affecting tumorigenesis. BRMS1 carries out this function through several mechanisms, including regulation of gene expression by binding to the mSin3/histone deacetylase (HDAC) transcriptional repressor complex. In the present study, we show that BRMS1 is a novel substrate of Cyclin-Dependent Kinase 2 (CDK2) that is phosphorylated on serine 237 (S237). Although CDKs are known to regulate cell cycle progression, the mutation of BRMS1 on serine 237 did not affect cell cycle progression and proliferation of MDA-MB-231 breast cancer cells; however, their migration was affected. Phosphorylation of BRMS1 does not affect its association with the mSin3/HDAC transcriptional repressor complex or its transcriptional repressor activity. The serine 237 phosphorylation site is immediately proximal to a C-terminal nuclear localization sequence that plays an important role in BRMS1-mediated metastasis suppression but phosphorylation does not control BRMS1 subcellular localization. Our studies demonstrate that CDK-mediated phosphorylation of BRMS1 regulates the migration of tumor cells.
Collapse
Affiliation(s)
- Siti Nur Ain Roesley
- a Cell Cycle and Cancer Unit , St Vincent's Institute of Medical Research , Victoria , Australia.,b Department of Medicine , University of Melbourne , Victoria , Australia
| | | | - Emma Morrish
- a Cell Cycle and Cancer Unit , St Vincent's Institute of Medical Research , Victoria , Australia
| | | | - Samah M A Issa
- e Metabolic Signalling Laboratory, St Vincent's Institute of Medical Research , Victoria , Australia
| | - Jonathan S Oakhill
- e Metabolic Signalling Laboratory, St Vincent's Institute of Medical Research , Victoria , Australia
| | - Ora Bernard
- b Department of Medicine , University of Melbourne , Victoria , Australia
| | - Danny R Welch
- f Department of Cancer Biology and The University of Kansas Cancer Center , University of Kansas Medical Center , Kansas City , KS , USA
| | - Boris Šarčević
- a Cell Cycle and Cancer Unit , St Vincent's Institute of Medical Research , Victoria , Australia.,b Department of Medicine , University of Melbourne , Victoria , Australia
| |
Collapse
|
12
|
Liu YR, Jiang YZ, Xu XE, Hu X, Yu KD, Shao ZM. Comprehensive Transcriptome Profiling Reveals Multigene Signatures in Triple-Negative Breast Cancer. Clin Cancer Res 2016; 22:1653-62. [DOI: 10.1158/1078-0432.ccr-15-1555] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Accepted: 11/12/2015] [Indexed: 11/16/2022]
|
13
|
Welch D, Manton C, Hurst D. Breast Cancer Metastasis Suppressor 1 (BRMS1): Robust Biological and Pathological Data, But Still Enigmatic Mechanism of Action. Adv Cancer Res 2016; 132:111-37. [PMID: 27613131 DOI: 10.1016/bs.acr.2016.05.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Metastasis requires coordinated expression of multiple genetic cassettes, often via epigenetic regulation of gene transcription. BRMS1 blocks metastasis, but not orthotopic tumor growth in multiple tumor types, presumably via SIN3 chromatin remodeling complexes. Although there is an abundance of strong data supporting BRMS1 as a metastasis suppressor, the mechanistic data directly connecting molecular pathways with inhibition of particular steps in metastasis are not well defined. In this review, the data for BRMS1-mediated metastasis suppression in multiple tumor types are discussed along with the steps in metastasis that are inhibited.
Collapse
|
14
|
Zhao LW, Zhong XH, Yang SY, Zhang YZ, Yang NJ. Inotodiol Inhabits Proliferation and Induces Apoptosis through Modulating Expression of cyclinE, p27, bcl-2, and bax in Human Cervical Cancer HeLa Cells. Asian Pac J Cancer Prev 2014; 15:3195-9. [DOI: 10.7314/apjcp.2014.15.7.3195] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
|
15
|
Lehalle D, Sanlaville D, Guimier A, Plouvier E, Leblanc T, Galmiche L, Radford I, Romana S, Colleaux L, de Pontual L, Lyonnet S, Amiel J. Multiple congenital anomalies-intellectual disability (MCA-ID) and neuroblastoma in a patient harboring a de novo 14q23.1q23.3 deletion. Am J Med Genet A 2014; 164A:1310-7. [DOI: 10.1002/ajmg.a.36452] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2013] [Accepted: 12/15/2013] [Indexed: 11/06/2022]
Affiliation(s)
- Daphné Lehalle
- Département de Génétique Histologie-Embryologie-Cytogénétique; Hôpital Necker-Enfants Malades; Paris France
- INSERM U781; Université Sorbonne Paris Cité, Institut IMAGINE; Paris France
| | - Damien Sanlaville
- Hospices Civils de Lyon; Service de Génétique and CRNL; CNRS UMR 5292; INSERM U1028, Université Claude Bernard Lyon I; Lyon France
| | - Anne Guimier
- Département de Génétique Histologie-Embryologie-Cytogénétique; Hôpital Necker-Enfants Malades; Paris France
- INSERM U781; Université Sorbonne Paris Cité, Institut IMAGINE; Paris France
| | - Emmanuel Plouvier
- Service d'Onco-Hématologie Pédiatrique; Centre Hospitalo-Universitaire de Besançon; Paris France
| | - Thierry Leblanc
- Département d'Hématologie Pédiatrique; Hôpitaux Robert Debré et Université Paris Diderot; Paris France
| | - Louise Galmiche
- Département d'Anatomo-Pathologie; Hôpital Necker-Enfants Malades; Paris France
| | - Isabelle Radford
- Département de Génétique Histologie-Embryologie-Cytogénétique; Hôpital Necker-Enfants Malades; Paris France
| | - Serge Romana
- Département de Génétique Histologie-Embryologie-Cytogénétique; Hôpital Necker-Enfants Malades; Paris France
| | - Laurence Colleaux
- INSERM U781; Université Sorbonne Paris Cité, Institut IMAGINE; Paris France
| | - Loïc de Pontual
- INSERM U781; Université Sorbonne Paris Cité, Institut IMAGINE; Paris France
| | - Stanislas Lyonnet
- Département de Génétique Histologie-Embryologie-Cytogénétique; Hôpital Necker-Enfants Malades; Paris France
- INSERM U781; Université Sorbonne Paris Cité, Institut IMAGINE; Paris France
| | - Jeanne Amiel
- Département de Génétique Histologie-Embryologie-Cytogénétique; Hôpital Necker-Enfants Malades; Paris France
- INSERM U781; Université Sorbonne Paris Cité, Institut IMAGINE; Paris France
| |
Collapse
|
16
|
Hasan MM, Brocca S, Sacco E, Spinelli M, Papaleo E, Lambrughi M, Alberghina L, Vanoni M. A comparative study of Whi5 and retinoblastoma proteins: from sequence and structure analysis to intracellular networks. Front Physiol 2014; 4:315. [PMID: 24478706 PMCID: PMC3897220 DOI: 10.3389/fphys.2013.00315] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2013] [Accepted: 10/13/2013] [Indexed: 11/18/2022] Open
Abstract
Cell growth and proliferation require a complex series of tight-regulated and well-orchestrated events. Accordingly, proteins governing such events are evolutionary conserved, even among distant organisms. By contrast, it is more singular the case of “core functions” exerted by functional analogous proteins that are not homologous and do not share any kind of structural similarity. This is the case of proteins regulating the G1/S transition in higher eukaryotes–i.e., the retinoblastoma (Rb) tumor suppressor Rb—and budding yeast, i.e., Whi5. The interaction landscape of Rb and Whi5 is quite large, with more than one hundred proteins interacting either genetically or physically with each protein. The Whi5 interactome has been used to construct a concept map of Whi5 function and regulation. Comparison of physical and genetic interactors of Rb and Whi5 allows highlighting a significant core of conserved, common functionalities associated with the interactors indicating that structure and function of the network—rather than individual proteins—are conserved during evolution. A combined bioinformatics and biochemical approach has shown that the whole Whi5 protein is highly disordered, except for a small region containing the protein family signature. The comparison with Whi5 homologs from Saccharomycetales has prompted the hypothesis of a modular organization of structural disorder, with most evolutionary conserved regions alternating with highly variable ones. The finding of a consensus sequence points to the conservation of a specific phosphorylation rhythm along with two disordered sequence motifs, probably acting as phosphorylation-dependent seeds in Whi5 folding/unfolding. Thus, the widely disordered Whi5 appears to act as a hierarchical, “date hub” that has evolutionary assayed an original way of modular organization before being supplanted by the globular, multi-domain structured Rb, more suitable to cover the role of a “party hub”.
Collapse
Affiliation(s)
- Md Mehedi Hasan
- SYSBIO Centre for Systems Biology Milano, Italy ; Department of Biotechnology and Biosciences, University of Milano-Bicocca Milano, Italy
| | - Stefania Brocca
- SYSBIO Centre for Systems Biology Milano, Italy ; Department of Biotechnology and Biosciences, University of Milano-Bicocca Milano, Italy
| | - Elena Sacco
- SYSBIO Centre for Systems Biology Milano, Italy ; Department of Biotechnology and Biosciences, University of Milano-Bicocca Milano, Italy
| | - Michela Spinelli
- SYSBIO Centre for Systems Biology Milano, Italy ; Department of Biotechnology and Biosciences, University of Milano-Bicocca Milano, Italy
| | - Elena Papaleo
- Department of Biotechnology and Biosciences, University of Milano-Bicocca Milano, Italy
| | - Matteo Lambrughi
- Department of Biotechnology and Biosciences, University of Milano-Bicocca Milano, Italy
| | - Lilia Alberghina
- SYSBIO Centre for Systems Biology Milano, Italy ; Department of Biotechnology and Biosciences, University of Milano-Bicocca Milano, Italy
| | - Marco Vanoni
- SYSBIO Centre for Systems Biology Milano, Italy ; Department of Biotechnology and Biosciences, University of Milano-Bicocca Milano, Italy
| |
Collapse
|
17
|
Metastasis suppression by BRMS1 associated with SIN3 chromatin remodeling complexes. Cancer Metastasis Rev 2013; 31:641-51. [PMID: 22678236 DOI: 10.1007/s10555-012-9363-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Epigenetic regulation of gene transcription by histone modification and chromatin remodeling has been linked to many biological and pathological events including cancer metastasis. Breast cancer metastasis suppressor 1 (BRMS1) interacts with SIN3 chromatin remodeling complexes, and, upon forced expression in metastatic cells, a nearly complete suppression of metastasis is noted without preventing primary tumor growth. The data for BRMS1-mediated metastasis suppression and SIN3 interaction are clear; however, connecting the inhibition directly to the association of BRMS1 with SIN3 complexes is currently not well defined. Considering the recent advancements in developing epigenetic drugs for cancer therapy, an improved understanding of how the interactions between BRMS1 and SIN3 regulate the process of metastasis should lead to novel therapies specifically targeting the most deadly aspect of tumor progression. In this article, the data for BRMS1-mediated metastasis suppression are reviewed with a focus on how the SIN3 chromatin remodeling complexes may be functionally involved.
Collapse
|
18
|
Vidal-Laliena M, Gallastegui E, Mateo F, Martínez-Balbás M, Pujol MJ, Bachs O. Histone deacetylase 3 regulates cyclin A stability. J Biol Chem 2013; 288:21096-21104. [PMID: 23760262 DOI: 10.1074/jbc.m113.458323] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
PCAF and GCN5 acetylate cyclin A at specific lysine residues targeting it for degradation at mitosis. We report here that histone deacetylase 3 (HDAC3) directly interacts with and deacetylates cyclin A. HDAC3 interacts with a domain included in the first 171 aa of cyclin A, a region involved in the regulation of its stability. In cells, overexpression of HDAC3 reduced cyclin A acetylation whereas the knocking down of HDAC3 increased its acetylation. Moreover, reduction of HDAC3 levels induced a decrease of cyclin A that can be reversed by proteasome inhibitors. These results indicate that HDAC3 is able to regulate cyclin A degradation during mitosis via proteasome. Interestingly, HDAC3 is abruptly degraded at mitosis also via proteasome thus facilitating cyclin A acetylation by PCAF/GCN5, which will target cyclin A for degradation. Because cyclin A is crucial for S phase progression and mitosis entry, the knock down of HDAC3 affects cell cycle progression specifically at both, S phase and G2/M transition. In summary we propose here that HDAC3 regulates cyclin A stability by counteracting the action of the acetylases PCAF/GCN5.
Collapse
Affiliation(s)
- Miriam Vidal-Laliena
- From the Department of Cell Biology, Immunology and Neurosciences, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, 08036 Barcelona, Spain and
| | - Edurne Gallastegui
- From the Department of Cell Biology, Immunology and Neurosciences, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, 08036 Barcelona, Spain and
| | | | - Marian Martínez-Balbás
- Molecular Biology, Barcelona Institute of Molecular Biology, Consejo Superior de Investigaciones Científicas (CSIC), 08028 Barcelona, Spain
| | - Maria Jesús Pujol
- From the Department of Cell Biology, Immunology and Neurosciences, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, 08036 Barcelona, Spain and
| | - Oriol Bachs
- From the Department of Cell Biology, Immunology and Neurosciences, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, 08036 Barcelona, Spain and.
| |
Collapse
|
19
|
Suryadinata R, Holien JK, Yang G, Parker MW, Papaleo E, Šarčević B. Molecular and structural insight into lysine selection on substrate and ubiquitin lysine 48 by the ubiquitin-conjugating enzyme Cdc34. Cell Cycle 2013; 12:1732-44. [PMID: 23656784 DOI: 10.4161/cc.24818] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
The attachment of ubiquitin (Ub) to lysines on substrates or itself by ubiquitin-conjugating (E2) and ubiquitin ligase (E3) enzymes results in protein ubiquitination. Lysine selection is important for generating diverse substrate-Ub structures and targeting proteins to different fates; however, the mechanisms of lysine selection are not clearly understood. The positioning of lysine(s) toward the E2/E3 active site and residues proximal to lysines are critical in their selection. We investigated determinants of lysine specificity of the ubiquitin-conjugating enzyme Cdc34, toward substrate and Ub lysines. Evaluation of the relative importance of different residues positioned -2, -1, +1 and +2 toward ubiquitination of its substrate, Sic1, on lysine 50 showed that charged residues in the -1 and -2 positions negatively impact on ubiquitination. Modeling suggests that charged residues at these positions alter the native salt-bridge interactions in Ub and Cdc34, resulting in misplacement of Sic1 lysine 50 in the Cdc34 catalytic cleft. During polyubiquitination, Cdc34 showed a strong preference for Ub lysine 48 (K48), with lower activity towards lysine 11 (K11) and lysine 63 (K63). Mutating the -2, -1, +1 and +2 sites surrounding K11 and K63 to mimic those surrounding K48 did not improve their ubiquitination, indicating that further determinants are important for Ub K48 specificity. Modeling the ternary structure of acceptor Ub with the Cdc34~Ub complex as well as in vitro ubiquitination assays unveiled the importance of K6 and Q62 of acceptor Ub for Ub K48 polyubiquitination. These findings provide molecular and structural insight into substrate lysine and Ub K48 specificity by Cdc34.
Collapse
Affiliation(s)
- Randy Suryadinata
- Cell Cycle and Cancer Unit, St. Vincent's Institute of Medical Research, Melbourne, VIC Australia
| | | | | | | | | | | |
Collapse
|
20
|
Smith KT, Sardiu ME, Martin-Brown SA, Seidel C, Mushegian A, Egidy R, Florens L, Washburn MP, Workman JL. Human family with sequence similarity 60 member A (FAM60A) protein: a new subunit of the Sin3 deacetylase complex. Mol Cell Proteomics 2012; 11:1815-28. [PMID: 22984288 DOI: 10.1074/mcp.m112.020255] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Here we describe the function of a previously uncharacterized protein, named family with sequence similarity 60 member A (FAM60A) that maps to chromosome 12p11 in humans. We use quantitative proteomics to determine that the main biochemical partners of FAM60A are subunits of the Sin3 deacetylase complex and show that FAM60A resides in active HDAC complexes. In addition, we conduct gene expression pathway analysis and find that FAM60A regulates expression of genes that encode components of the TGF-beta signaling pathway. Moreover, our studies reveal that loss of FAM60A or another component of the Sin3 complex, SDS3, leads to a change in cell morphology and an increase in cell migration. These studies reveal the function of a previously uncharacterized protein and implicate the Sin3 complex in suppressing cell migration.
Collapse
Affiliation(s)
- Karen T Smith
- Stowers Institute for Medical Research, Kansas City, Missouri 64110, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Unraveling the enigmatic complexities of BRMS1-mediated metastasis suppression. FEBS Lett 2011; 585:3185-90. [PMID: 21827753 DOI: 10.1016/j.febslet.2011.07.045] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2011] [Revised: 07/28/2011] [Accepted: 07/29/2011] [Indexed: 01/15/2023]
Abstract
Expression of BRMS1 causes dramatic suppression of metastasis in multiple in vivo model systems. As we gain further insight into the biochemical mechanisms of BRMS1, we appreciate the importance of both molecular and cellular context for functional metastasis suppression. BRMS1 associates with large chromatin remodeling complexes including SIN3:HDAC which are powerful epigenetic regulators of gene expression. Additionally, BRMS1 inhibits the activity of NFκB, a well-known transcription factor that plays significant roles in tumor progression. Moreover, BRMS1 coordinately regulates the expression of metastasis-associated microRNA known as metastamir. How these biochemical mechanisms and biological pathways are linked, either directly or indirectly, and the influence of molecular and cellular context, are critical considerations for the discovery of novel therapeutic targets for the most deadly aspect of tumor progression-metastasis.
Collapse
|