67 results on '"Kriwacki, R"'
Search Results
2. Solution structure of a Bcl-xL S62E mutant
- Author
-
Follis, A.V., primary and Kriwacki, R., additional
- Published
- 2017
- Full Text
- View/download PDF
3. The landscape of somatic mutations in infant MLL-rearranged acute lymphoblastic leukemias.
- Author
-
Andersson, AK, Ma, J, Wang, J, Chen, X, Gedman, AL, Dang, J, Nakitandwe, J, Holmfeldt, L, Parker, M, Easton, J, Huether, R, Kriwacki, R, Rusch, M, Wu, G, Li, Y, Mulder, H, Raimondi, S, Pounds, S, Kang, G, Shi, L, Becksfort, J, Gupta, P, Payne-Turner, D, Vadodaria, B, Boggs, K, Yergeau, D, Manne, J, Song, G, Edmonson, M, Nagahawatte, P, Wei, L, Cheng, C, Pei, D, Sutton, R, Venn, NC, Chetcuti, A, Rush, A, Catchpoole, D, Heldrup, J, Fioretos, T, Lu, C, Ding, L, Pui, C-H, Shurtleff, S, Mullighan, CG, Mardis, ER, Wilson, RK, Gruber, TA, Zhang, J, Downing, JR, St. Jude Children's Research Hospital–Washington University Pediatric Cancer Genome Project, Andersson, AK, Ma, J, Wang, J, Chen, X, Gedman, AL, Dang, J, Nakitandwe, J, Holmfeldt, L, Parker, M, Easton, J, Huether, R, Kriwacki, R, Rusch, M, Wu, G, Li, Y, Mulder, H, Raimondi, S, Pounds, S, Kang, G, Shi, L, Becksfort, J, Gupta, P, Payne-Turner, D, Vadodaria, B, Boggs, K, Yergeau, D, Manne, J, Song, G, Edmonson, M, Nagahawatte, P, Wei, L, Cheng, C, Pei, D, Sutton, R, Venn, NC, Chetcuti, A, Rush, A, Catchpoole, D, Heldrup, J, Fioretos, T, Lu, C, Ding, L, Pui, C-H, Shurtleff, S, Mullighan, CG, Mardis, ER, Wilson, RK, Gruber, TA, Zhang, J, Downing, JR, and St. Jude Children's Research Hospital–Washington University Pediatric Cancer Genome Project
- Abstract
Infant acute lymphoblastic leukemia (ALL) with MLL rearrangements (MLL-R) represents a distinct leukemia with a poor prognosis. To define its mutational landscape, we performed whole-genome, exome, RNA and targeted DNA sequencing on 65 infants (47 MLL-R and 18 non-MLL-R cases) and 20 older children (MLL-R cases) with leukemia. Our data show that infant MLL-R ALL has one of the lowest frequencies of somatic mutations of any sequenced cancer, with the predominant leukemic clone carrying a mean of 1.3 non-silent mutations. Despite this paucity of mutations, we detected activating mutations in kinase-PI3K-RAS signaling pathway components in 47% of cases. Surprisingly, these mutations were often subclonal and were frequently lost at relapse. In contrast to infant cases, MLL-R leukemia in older children had more somatic mutations (mean of 6.5 mutations/case versus 1.3 mutations/case, P = 7.15 × 10(-5)) and had frequent mutations (45%) in epigenetic regulators, a category of genes that, with the exception of MLL, was rarely mutated in infant MLL-R ALL.
- Published
- 2015
4. Nuclear ULK1 promotes cell death in response to oxidative stress through PARP1
- Author
-
Joshi, A, primary, Iyengar, R, additional, Joo, J H, additional, Li-Harms, X J, additional, Wright, C, additional, Marino, R, additional, Winborn, B J, additional, Phillips, A, additional, Temirov, J, additional, Sciarretta, S, additional, Kriwacki, R, additional, Peng, J, additional, Shelat, A, additional, and Kundu, M, additional
- Published
- 2015
- Full Text
- View/download PDF
5. Fishing in the Nuclear Pore
- Author
-
Kriwacki, R. W., primary and Yoon, M.-K., additional
- Published
- 2011
- Full Text
- View/download PDF
6. ChemInform Abstract: A Novel Stereoselective Route to (S)-(+)-α-(Fluoromethyl) histidine: α-Halomethylation of (2R,4S)-3-Benzoyl-2-(1,1- dimethylethyl)-1-methyl-4-((N-tritylimidazol-4′-yl )methyl)-1,3- imidazolidin-5-ones. Synthesis and 1H NMR Spectroscop
- Author
-
GROZINGER, K. G., primary, KRIWACKI, R. W., additional, LEONARD, S. F., additional, and PITNER, T. P., additional
- Published
- 2010
- Full Text
- View/download PDF
7. Nuclear ULK1 promotes cell death in response to oxidative stress through PARP1
- Author
-
Joshi, A, Iyengar, R, Joo, J H, Li-Harms, X J, Wright, C, Marino, R, Winborn, B J, Phillips, A, Temirov, J, Sciarretta, S, Kriwacki, R, Peng, J, Shelat, A, and Kundu, M
- Abstract
Reactive oxygen species (ROS) may cause cellular damage and oxidative stress-induced cell death. Autophagy, an evolutionarily conserved intracellular catabolic process, is executed by autophagy (ATG) proteins, including the autophagy initiation kinase Unc-51-like kinase (ULK1)/ATG1. Although autophagy has been implicated to have both cytoprotective and cytotoxic roles in the response to ROS, the role of individual ATG proteins, including ULK1, remains poorly characterized. In this study, we demonstrate that ULK1 sensitizes cells to necrotic cell death induced by hydrogen peroxide (H2O2). Moreover, we demonstrate that ULK1 localizes to the nucleus and regulates the activity of the DNA damage repair protein poly (ADP-ribose) polymerase 1 (PARP1) in a kinase-dependent manner. By enhancing PARP1 activity, ULK1 contributes to ATP depletion and death of H2O2-treated cells. Our study provides the first evidence of an autophagy-independent prodeath role for nuclear ULK1 in response to ROS-induced damage. On the basis of our data, we propose that the subcellular distribution of ULK1 has an important role in deciding whether a cell lives or dies on exposure to adverse environmental or intracellular conditions.
- Published
- 2016
- Full Text
- View/download PDF
8. Structural basis for LFA-1 inhibition upon lovastatin binding to the CD11a I-domain 1 1Edited by F. E. Cohen
- Author
-
Kallen, J., primary, Welzenbach, K., additional, Ramage, P., additional, Geyl, D., additional, Kriwacki, R., additional, Legge, G., additional, Cottens, S., additional, Weitz-Schmidt, G., additional, and Hommel, U., additional
- Published
- 1999
- Full Text
- View/download PDF
9. Structural studies of p21Waf1/Cip1/Sdi1 in the free and Cdk2-bound state: conformational disorder mediates binding diversity.
- Author
-
Kriwacki, R W, primary, Hengst, L, additional, Tennant, L, additional, Reed, S I, additional, and Wright, P E, additional
- Published
- 1996
- Full Text
- View/download PDF
10. Sequence-specific recognition of DNA by zinc-finger peptides derived from the transcription factor Sp1.
- Author
-
Kriwacki, R W, primary, Schultz, S C, additional, Steitz, T A, additional, and Caradonna, J P, additional
- Published
- 1992
- Full Text
- View/download PDF
11. Structural basis for LFA-1 inhibition upon lovastatin binding to the CD11a I-domain11Edited by F. E. Cohen
- Author
-
Kallen, J., Welzenbach, K., Ramage, P., Geyl, D., Kriwacki, R., Legge, G., Cottens, S., Weitz-Schmidt, G., and Hommel, U.
- Abstract
The lymphocyte function-associated antigen (LFA-1) belongs to the family of β2-integrins and plays an important role in T-cell activation and leukocyte migration to sites of inflammation. We report here that lovastatin, a drug clinically used for lowering cholesterol levels, inhibits the interaction of human LFA-1 with its counter-receptor intercellular adhesion molecule-1. Using nuclear magnetic resonance spectroscopy and X-ray crystallography we show that the inhibitor binds to a highly conserved domain of the LFA-1 α-chain called the I-domain. The first three-dimensional structure of an integrin inhibitor bound to its receptor reveals atomic details for a hitherto unknown mode of LFA-1 inhibition. It also sheds light into possible mechanisms of LFA-1 mediated signalling and will support the design of novel anti-adhesive and immunosuppressive drugs.
- Published
- 1999
- Full Text
- View/download PDF
12. The conformational analysis of delta 9- and delta 9,11-tetrahydrocannabinols in solution using high resolution nuclear magnetic resonance spectroscopy.
- Author
-
Kriwacki, R W and Makriyannis, A
- Abstract
The conformation for each of two cannabinoids, delta 9-tetrahydrocannabinol (THC), the principal active constituent of marihuana, and delta 9,11-THC, a biologically inactive structural isomer of delta 9- THC, is analyzed by interpreting 1H-1H and 13C-1H coupling constants and nuclear Overhauser effects determined using one-and two-dimensional 1H and 13C NMR techniques. The interpretation of both vicinal and long range coupling constants was necessary to deduce the conformations of these tricyclic molecules with certainty, with nuclear Overhauser effect enhancements used, when appropriate, to confirm the results. These findings provide insights into the structure-activity requirements for the cannabinoids.
- Published
- 1989
13. Structures of zinc finger domains from transcription factor Sp1. Insights into sequence-specific protein-DNA recognition.
- Author
-
Narayan, V A, Kriwacki, R W, and Caradonna, J P
- Abstract
The carboxyl terminus of transcription factor Sp1 contains three contiguous Cys2-His2 zinc finger domains with the consensus sequence Cys-X2-4-Cys-X12-His-X3-His. We have used standard homonuclear two-dimensional NMR techniques to solve the solution structures of synthetic peptides corresponding to the last two zinc finger domains (Sp1f2 and Sp1f3, respectively) of Sp1. Our studies indicate a classical Cys2-His2 type fold for both the domains differing from each other primarily in the conformation of Cys-X2-Cys (beta-type I turn) and Cys-X4-Cys (beta-type II turn) elements. There are, however, no significant differences in the metal binding properties between the Cys-X4-Cys (Sp1f2) and Cys-X2-Cys (Sp1f3) subclasses of zinc fingers. The free solution structures of Sp1f2 and Sp1f3 are very similar to those of the analogous fingers of Zif268 bound to DNA. There is NMR spectral evidence suggesting that the Arg-Asp buttressing interaction observed in the Zif-268.DNA complex is also preserved in unbound Sp1f2 and Sp1f3. Modeling Sp1-DNA complex by overlaying the Sp1f2 and Sp1f3 structures on Zif268 fingers 1 and 2, respectively, predicts the role of key amino acid residues, the interference/protection data, and supports the model of Sp1-DNA interaction proposed earlier.
- Published
- 1997
14. Probing protein structure using biochemical and biophysical methods. Proteolysis, matrix-assisted laser desorption/ionization mass spectrometry, high-performance liquid chromatography and size-exclusion chromatography of p21^W^a^t^1^/^C^i^p^1^/^S^d^i^1
- Author
-
Kriwacki, R. W., Wu, J., Tenant, L., Wright, P. E., and Siuzdak, G.
- Published
- 1997
- Full Text
- View/download PDF
15. ChemInform Abstract: Amidomethylation of Indoles and Cyclizations to Spiro(pyrrolo-(4,3,2-de)isoquinoline-3,4′-piperidines).
- Author
-
CARTER, C., primary, FRETER, K. R., additional, KRIWACKI, R. W., additional, LEONARD, S. F., additional, and PITNER, T. P., additional
- Published
- 1987
- Full Text
- View/download PDF
16. ChemInform Abstract: Amidomethylation of Phenanthridines: Synthesis and Structure Determination by 1H NMR.
- Author
-
FRETER, K. R., primary, GROEZINGER, K. G., additional, and KRIWACKI, R. W., additional
- Published
- 1989
- Full Text
- View/download PDF
17. Amidomethylation of phenanthridines: Synthesis and structure determination by 1H NMR
- Author
-
Freter, K. R., primary, Grözinger, K. G., additional, and Kriwacki, R. W., additional
- Published
- 1988
- Full Text
- View/download PDF
18. ChemInform Abstract: A Novel Stereoselective Route to (S)-(+)-α-(Fluoromethyl) histidine: α-Halomethylation of (2R,4S)-3-Benzoyl-2-(1,1- dimethylethyl)-1-methyl-4-((N-tritylimidazol-4′-yl )methyl)-1,3- imidazolidin-5-ones. Synthesis and 1H NMR Spectroscopy.
- Author
-
GROZINGER, K. G., KRIWACKI, R. W., LEONARD, S. F., and PITNER, T. P.
- Published
- 1993
- Full Text
- View/download PDF
19. p14 ARF forms meso-scale assemblies upon phase separation with NPM1.
- Author
-
Gibbs E, Miao Q, Ferrolino M, Bajpai R, Hassan A, Phillips AH, Pitre A, Kümmerle R, Miller S, Nagy G, Leite W, Heller W, Stanley C, Perrone B, and Kriwacki R
- Subjects
- Humans, Hydrophobic and Hydrophilic Interactions, Cell Proliferation, Tumor Suppressor Protein p53 metabolism, Protein Binding, Phase Separation, Nucleophosmin, Nuclear Proteins metabolism, Nuclear Proteins genetics, Nuclear Proteins chemistry, Tumor Suppressor Protein p14ARF metabolism, Tumor Suppressor Protein p14ARF genetics, Cell Nucleolus metabolism
- Abstract
NPM1 is an abundant nucleolar chaperone that, in addition to facilitating ribosome biogenesis, contributes to nucleolar stress responses and tumor suppression through its regulation of the p14 Alternative Reading Frame tumor suppressor protein (p14
ARF ). Oncogenic stress induces p14ARF to inhibit MDM2, stabilize p53 and arrest the cell cycle. Under non-stress conditions, NPM1 stabilizes p14ARF in nucleoli, preventing its degradation and blocking p53 activation. However, the mechanisms underlying the regulation of p14ARF by NPM1 are unclear because the structural features of the p14ARF -NPM1 complex were elusive. Here we show that p14ARF assembles into a gel-like meso-scale network upon phase separation with NPM1. This assembly is mediated by intermolecular contacts formed by hydrophobic residues in an α-helix and β-strands within a partially folded N-terminal portion of p14ARF . These hydrophobic interactions promote phase separation with NPM1, enhance p14ARF nucleolar partitioning, restrict NPM1 diffusion within condensates and nucleoli, and reduce cellular proliferation. Our structural analysis provides insights into the multifaceted chaperone function of NPM1 in nucleoli by mechanistically linking the nucleolar localization of p14ARF to its partial folding and meso-scale assembly upon phase separation with NPM1., Competing Interests: Competing interests The authors declare no competing interests., (© 2024. The Author(s).)- Published
- 2024
- Full Text
- View/download PDF
20. Properties governing small-molecule partitioning into biomolecular condensates.
- Author
-
Chandra B, Tripathi S, and Kriwacki R
- Published
- 2024
- Full Text
- View/download PDF
21. Disruption of cotranscriptional splicing suggests that RBM39 is a therapeutic target in acute lymphoblastic leukemia.
- Author
-
Jin Q, Harris E, Myers JA, Mehmood R, Cotton A, Shirnekhi HK, Baggett DW, Wen JQ, Schild AB, Bhansali RS, Klein J, Narina S, Pieters T, Yoshimi A, Pruett-Miller SM, Kriwacki R, Abdel-Wahab O, Malinge S, Ntziachristos P, Obeng EA, and Crispino JD
- Abstract
There are few options for patients with relapse/refractory B-cell acute lymphoblastic leukemia (B-ALL), thus this is a major area of unmet medical need. Here, we reveal that inclusion of a poison exon in RBM39, which could be induced both by CDK9 or CDK9 independent CMGC (cyclin-dependent kinases, mitogen-activated protein kinases, glycogen synthase kinases, CDC-like kinases) kinase inhibition, is recognized by the nonsense-mediated mRNA decay (NMD) pathway for degradation. Targeting this poison exon in RBM39 with CMGC inhibitors lead to protein downregulation and inhibition of ALL growth, particularly in relapse/refractory B-ALL. Mechanistically, disruption of co-transcriptional splicing by inhibition of CMGC kinases including DYRK1A, or inhibition of CDK9, which phosphorylate the C-terminal domain of RNA polymerase II (Pol II), results in alteration of SF3B1 and Pol II association. Disruption of SF3B1 and transcriptional elongation complex alters Pol II pausing, which promotes the inclusion of a poison exon in RBM39. Moreover, RBM39 ablation suppresses the growth of human B-ALL, and targeting RBM39 with sulfonamides, which degrade RBM39 protein, showed strong anti-tumor activity in preclinical models. Our data reveal that relapse/refractory B-ALL is susceptible to pharmacologic and genetic inhibition of RBM39 and provide two potential strategies to target this axis., (Copyright © 2024 American Society of Hematology.)
- Published
- 2024
- Full Text
- View/download PDF
22. Dissecting the biophysics and biology of intrinsically disordered proteins.
- Author
-
Banerjee PR, Holehouse AS, Kriwacki R, Robustelli P, Jiang H, Sobolevsky AI, Hurley JM, and Mendell JT
- Subjects
- Humans, Biophysics, Biology, Intrinsically Disordered Proteins metabolism
- Abstract
Intrinsically disordered regions (IDRs) within human proteins play critical roles in cellular information processing, including signaling, transcription, stress response, DNA repair, genome organization, and RNA processing. Here, we summarize current challenges in the field and propose cutting-edge approaches to address them in physiology and disease processes, with a focus on cancer., Competing Interests: Declaration of interests The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2023 Elsevier Ltd. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
23. p14 ARF forms meso-scale assemblies upon phase separation with NPM1.
- Author
-
Gibbs E, Miao Q, Ferrolino M, Bajpai R, Hassan A, Phillips AH, Pitre A, Kümmerle R, Miller S, Heller W, Stanley C, Perrone B, and Kriwacki R
- Abstract
NPM1 is an abundant nucleolar chaperone that, in addition to facilitating ribosome biogenesis, contributes to nucleolar stress responses and tumor suppression through its regulation of the p14 Alternative Reading Frame tumor suppressor protein (p14
ARF ). Oncogenic stress induces p14ARF to inhibit MDM2, stabilize p53 and arrest the cell cycle. Under non-stress conditions, NPM1 stabilizes p14ARF in nucleoli, preventing its degradation and blocking p53 activation. However, the mechanisms underlying the regulation of p14ARF by NPM1 are unclear because the structural features of the p14ARF -NPM1 complex remain elusive. Here we show that NPM1 sequesters p14ARF within phase-separated condensates, facilitating the assembly of p14ARF into a gel-like meso-scale network. This assembly is mediated by intermolecular contacts formed by hydrophobic residues in an α-helix and β-strands within a partially folded N-terminal domain of p14ARF . Those hydrophobic interactions promote phase separation with NPM1, enhance nucleolar partitioning of p14ARF , restrict p14ARF and NPM1 diffusion within condensates and in nucleoli, and reduce cell viability. Our structural model provides novel insights into the multifaceted chaperone function of NPM1 in nucleoli by mechanistically linking the nucleolar localization of p14ARF to its partial folding and meso-scale assembly upon phase separation with NPM1., Competing Interests: Additional Declarations: There is NO Competing Interest.- Published
- 2023
- Full Text
- View/download PDF
24. Viscoelasticity and advective flow of RNA underlies nucleolar form and function.
- Author
-
Riback JA, Eeftens JM, Lee DSW, Quinodoz SA, Donlic A, Orlovsky N, Wiesner L, Beckers L, Becker LA, Strom AR, Rana U, Tolbert M, Purse BW, Kleiner R, Kriwacki R, and Brangwynne CP
- Subjects
- Biomolecular Condensates, Cell Nucleolus genetics, Nuclear Proteins genetics, RNA genetics, RNA, Ribosomal genetics
- Abstract
The nucleolus is the largest biomolecular condensate and facilitates transcription, processing, and assembly of ribosomal RNA (rRNA). Although nucleolar function is thought to require multiphase liquid-like properties, nucleolar fluidity and its connection to the highly coordinated transport and biogenesis of ribosomal subunits are poorly understood. Here, we use quantitative imaging, mathematical modeling, and pulse-chase nucleotide labeling to examine nucleolar material properties and rRNA dynamics. The mobility of rRNA is several orders of magnitude slower than that of nucleolar proteins, with rRNA steadily moving away from the transcriptional sites in a slow (∼1 Å/s), radially directed fashion. This constrained but directional mobility, together with polymer physics-based calculations, suggests that nascent rRNA forms an entangled gel, whose constant production drives outward flow. We propose a model in which progressive maturation of nascent rRNA reduces its initial entanglement, fluidizing the nucleolar periphery to facilitate the release of assembled pre-ribosomal particles., Competing Interests: Declaration of interests C.P.B. is a founder and consultant for Nereid Therapeutics., (Copyright © 2023 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
25. Phase Separation in Biology and Disease; Current Perspectives and Open Questions.
- Author
-
Boeynaems S, Chong S, Gsponer J, Holt L, Milovanovic D, Mitrea DM, Mueller-Cajar O, Portz B, Reilly JF, Reinkemeier CD, Sabari BR, Sanulli S, Shorter J, Sontag E, Strader L, Stachowiak J, Weber SC, White M, Zhang H, Zweckstetter M, Elbaum-Garfinkle S, and Kriwacki R
- Subjects
- Humans, Biomolecular Condensates, Disease, Phase Transition
- Abstract
In the past almost 15 years, we witnessed the birth of a new scientific field focused on the existence, formation, biological functions, and disease associations of membraneless bodies in cells, now referred to as biomolecular condensates. Pioneering studies from several laboratories [reviewed in
1-3 ] supported a model wherein biomolecular condensates associated with diverse biological processes form through the process of phase separation. These and other findings that followed have revolutionized our understanding of how biomolecules are organized in space and time within cells to perform myriad biological functions, including cell fate determination, signal transduction, endocytosis, regulation of gene expression and protein translation, and regulation of RNA metabolism. Further, condensates formed through aberrant phase transitions have been associated with numerous human diseases, prominently including neurodegeneration and cancer. While in some cases, rigorous evidence supports links between formation of biomolecular condensates through phase separation and biological functions, in many others such links are less robustly supported, which has led to rightful scrutiny of the generality of the roles of phase separation in biology and disease.4-7 During a week-long workshop in March 2022 at the Telluride Science Research Center (TSRC) in Telluride, Colorado, ∼25 scientists addressed key questions surrounding the biomolecular condensates field. Herein, we present insights gained through these discussions, addressing topics including, roles of condensates in diverse biological processes and systems, and normal and disease cell states, their applications to synthetic biology, and the potential for therapeutically targeting biomolecular condensates., Competing Interests: Declaration of Competing Interest The authors declare the following financial interests/personal relationships which may be considered as potential competing interests: D.M.M. is an employee and shareholder of Dewpoint Therapeutics; B.P. is an employee and shareholder of Dewpoint Therapeutics; J.F.R. is an an officer and shareholder of Nereid Therapeutics; J.S. is a consultant for Dewpoint Therapeutics, ADRx, and Neumora, and a shareholder and advisor for Confluence Therapeutics; L.C.S. is on the Prose Foods Scientific Advisory Board; M.W. was an employee and shareholder of Faze Medicines when this article was conceived and initially written, and currently is an employee of IDEXX Laboratories; R.K. reports personal fees from Dewpoint Therapeutics, GLG Consulting, and New Equilibrium Biosciences outside the submitted work., (Copyright © 2023 The Authors. Published by Elsevier Ltd.. All rights reserved.)- Published
- 2023
- Full Text
- View/download PDF
26. Charting the human disease condensate dysregulome.
- Author
-
Chandra B and Kriwacki R
- Subjects
- Humans, Proteins
- Abstract
Although it is understood that myriad proteins function in cells within biomolecular condensates, it is unclear how protein condensation is altered in human disease. In this issue of Developmental Cell, Banani et al. show that mutations in disease-associated proteins may map to phase-separation-prone regions and thereby alter condensate formation., Competing Interests: Declaration of interests R.K. has received compensation unrelated to this work for advisory board membership of Dewpoint Therapeutics, for consulting with GLG Consulting, and through patent licensing to New Equilibrium Biosciences., (Copyright © 2022 Elsevier Inc. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
27. An Image Analysis Pipeline for Quantifying the Features of Fluorescently-Labeled Biomolecular Condensates in Cells.
- Author
-
Baggett DW, Medyukhina A, Tripathi S, Shirnekhi HK, Wu H, Pounds SB, Khairy K, and Kriwacki R
- Abstract
Biomolecular condensates are cellular organelles formed through liquid-liquid phase separation (LLPS) that play critical roles in cellular functions including signaling, transcription, translation, and stress response. Importantly, condensate misregulation is associated with human diseases, including neurodegeneration and cancer among others. When condensate-forming biomolecules are fluorescently-labeled and examined with fluorescence microscopy they appear as illuminated foci, or puncta, in cells. Puncta features such as number, volume, shape, location, and concentration of biomolecular species within them are influenced by the thermodynamics of biomolecular interactions that underlie LLPS. Quantification of puncta features enables evaluation of the thermodynamic driving force for LLPS and facilitates quantitative comparisons of puncta formed under different cellular conditions or by different biomolecules. Our work on nucleoporin 98 (NUP98) fusion oncoproteins (FOs) associated with pediatric leukemia inspired us to develop an objective and reliable computational approach for such analyses. The NUP98-HOXA9 FO forms hundreds of punctate transcriptional condensates in cells, leading to hematopoietic cell transformation and leukemogenesis. To quantify the features of these puncta and derive the associated thermodynamic parameters, we developed a live-cell fluorescence microscopy image processing pipeline based on existing methodologies and open-source tools. The pipeline quantifies the numbers and volumes of puncta and fluorescence intensities of the fluorescently-labeled biomolecule(s) within them and generates reports of their features for hundreds of cells. Using a standard curve of fluorescence intensity versus protein concentration, the pipeline determines the apparent molar concentration of fluorescently-labeled biomolecules within and outside of puncta and calculates the partition coefficient (K
p ) and Gibbs free energy of transfer (ΔGTr ), which quantify the favorability of a labeled biomolecule partitioning into puncta. In addition, we provide a library of R functions for statistical analysis of the extracted measurements for certain experimental designs. The source code, analysis notebooks, and test data for the Punctatools pipeline are available on GitHub: https://github.com/stjude/punctatools. Here, we provide a protocol for applying our Punctatools pipeline to extract puncta features from fluorescence microscopy images of cells., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2022 Baggett, Medyukhina, Tripathi, Shirnekhi, Wu, Pounds, Khairy and Kriwacki.)- Published
- 2022
- Full Text
- View/download PDF
28. LAG3 associates with TCR-CD3 complexes and suppresses signaling by driving co-receptor-Lck dissociation.
- Author
-
Guy C, Mitrea DM, Chou PC, Temirov J, Vignali KM, Liu X, Zhang H, Kriwacki R, Bruchez MP, Watkins SC, Workman CJ, and Vignali DAA
- Subjects
- Antigens, CD immunology, CD3 Complex immunology, CD8 Antigens metabolism, Histocompatibility Antigens Class II, Receptors, Antigen, T-Cell metabolism, Signal Transduction, Lymphocyte Activation Gene 3 Protein, CD8-Positive T-Lymphocytes, Lymphocyte Specific Protein Tyrosine Kinase p56(lck) metabolism
- Abstract
LAG3 is an inhibitory receptor that is highly expressed on exhausted T cells. Although LAG3-targeting immunotherapeutics are currently in clinical trials, how LAG3 inhibits T cell function remains unclear. Here, we show that LAG3 moved to the immunological synapse and associated with the T cell receptor (TCR)-CD3 complex in CD4
+ and CD8+ T cells, in the absence of binding to major histocompatibility complex class II-its canonical ligand. Mechanistically, a phylogenetically conserved, acidic, tandem glutamic acid-proline repeat in the LAG3 cytoplasmic tail lowered the pH at the immune synapse and caused dissociation of the tyrosine kinase Lck from the CD4 or CD8 co-receptor, which resulted in a loss of co-receptor-TCR signaling and limited T cell activation. These observations indicated that LAG3 functioned as a signal disruptor in a major histocompatibility complex class II-independent manner, and provide insight into the mechanism of action of LAG3-targeting immunotherapies., (© 2022. The Author(s), under exclusive licence to Springer Nature America, Inc.)- Published
- 2022
- Full Text
- View/download PDF
29. Clinical and Functional Significance of TP53 Exon 4-Intron 4 Splice Junction Variants.
- Author
-
Pinto EM, Maxwell KN, Halalsheh H, Phillips A, Powers J, MacFarland S, Walsh MF, Breen K, Formiga MN, Kriwacki R, Nichols KE, Mostafavi R, Wang J, Clay MR, Rodriguez-Galindo C, Ribeiro RC, and Zambetti GP
- Subjects
- Humans, Exons genetics, Genetic Variation genetics, Introns genetics, Tumor Suppressor Protein p53 genetics
- Abstract
Germline TP53 splicing variants are uncommon, and their clinical relevance is unknown. However, splice-altering variants at exon 4-intron 4 junctions are relatively enriched in pediatric adrenocortical tumors (ACT). Nevertheless, family histories of cancer compatible with classic Li-Fraumeni syndrome are rarely seen in these patients. We used conventional and in silico assays to determine protein stability, splicing, and transcriptional activity of 10 TP53 variants at exon 4-intron 4 junctions and analyzed their clinical correlates. We reviewed public databases that report the impact of TP53 variants in human cancer and examined individual reports, focusing on family history of cancer. TP53 exon 4-intron 4 junction germline variants were identified in 9 of 75 pediatric ACTs enrolled in the International Pediatric Adrenocortical Tumor Registry and Children's Oncology Group ARAR0332 study. An additional eight independent TP53 variants involving exon 4 splicing were identified in the Pediatric Cancer Genome Project ( n = 5,213). These variants resulted in improper expression due to ineffective splicing, protein instability, altered subcellular localization, and loss of function. Clinical case review of carriers of TP53 exon 4-intron 4 junction variants revealed a high incidence of pediatric ACTs and atypical tumor types not consistent with classic Li-Fraumeni syndrome. Germline variants involving TP53 exon 4-intron 4 junctions are frequent in ACT and rare in other pediatric tumors. The collective impact of these germline TP53 variants on the fidelity of splicing, protein structure, and function must be considered in evaluating cancer susceptibility. IMPLICATIONS: Taken together, the data indicate that splice variants at TP53 codon 125 and surrounding bases differentially impacted p53 gene expression and function., (©2021 American Association for Cancer Research.)
- Published
- 2022
- Full Text
- View/download PDF
30. NPM1 exhibits structural and dynamic heterogeneity upon phase separation with the p14ARF tumor suppressor.
- Author
-
Gibbs E, Perrone B, Hassan A, Kümmerle R, and Kriwacki R
- Subjects
- Amino Acid Sequence, Cell Nucleolus chemistry, Cloning, Molecular, Humans, Models, Molecular, Molecular Structure, Nuclear Magnetic Resonance, Biomolecular, Nucleophosmin, Open Reading Frames, Protein Structure, Secondary, Nuclear Proteins chemistry, Tumor Suppressor Protein p14ARF chemistry
- Abstract
Nucleophosmin (NPM1) is an abundant nucleolar protein that aids in the maturation of pre-ribosomal particles and participates in oncogenic stress responses through its interaction with the Alternative Reading Frame tumor suppressor (p14ARF). NPM1 mediates multiple mechanisms of phase separation which contribute to the liquid-like properties of nucleoli. However, the effects of phase separation on the structure and dynamics of NPM1 are poorly understood. Here we show that NPM1 undergoes phase separation with p14ARF in vitro, forming condensates that immobilize both proteins. We probed the structure and dynamics of NPM1 within the condensed phase using solid-state NMR spectroscopy. Our results demonstrate that within the condensed phase, the NPM1 oligomerization domain forms an immobile scaffold, while the central intrinsically disordered region and the C-terminal nucleic acid binding domain exhibit relative mobility., (Copyright © 2019 Elsevier Inc. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
31. Dynamic anticipation by Cdk2/Cyclin A-bound p27 mediates signal integration in cell cycle regulation.
- Author
-
Tsytlonok M, Sanabria H, Wang Y, Felekyan S, Hemmen K, Phillips AH, Yun MK, Waddell MB, Park CG, Vaithiyalingam S, Iconaru L, White SW, Tompa P, Seidel CAM, and Kriwacki R
- Subjects
- Crystallography, X-Ray, Cyclin A isolation & purification, Cyclin-Dependent Kinase 2 isolation & purification, Cyclin-Dependent Kinase Inhibitor p27 genetics, Cyclin-Dependent Kinase Inhibitor p27 isolation & purification, Fusion Proteins, bcr-abl metabolism, Molecular Dynamics Simulation, Mutagenesis, Site-Directed, Phosphorylation physiology, Protein Binding physiology, Protein Processing, Post-Translational physiology, Protein Structure, Tertiary physiology, Proteolysis, Recombinant Proteins genetics, Recombinant Proteins isolation & purification, Recombinant Proteins metabolism, Signal Transduction physiology, Threonine metabolism, Tyrosine metabolism, src-Family Kinases isolation & purification, src-Family Kinases metabolism, Cell Division physiology, Cyclin A metabolism, Cyclin-Dependent Kinase 2 metabolism, Cyclin-Dependent Kinase Inhibitor p27 metabolism
- Abstract
p27
Kip1 is an intrinsically disordered protein (IDP) that inhibits cyclin-dependent kinase (Cdk)/cyclin complexes (e.g., Cdk2/cyclin A), causing cell cycle arrest. Cell division progresses when stably Cdk2/cyclin A-bound p27 is phosphorylated on one or two structurally occluded tyrosine residues and a distal threonine residue (T187), triggering degradation of p27. Here, using an integrated biophysical approach, we show that Cdk2/cyclin A-bound p27 samples lowly-populated conformations that provide access to the non-receptor tyrosine kinases, BCR-ABL and Src, which phosphorylate Y88 or Y88 and Y74, respectively, thereby promoting intra-assembly phosphorylation (of p27) on distal T187. Even when tightly bound to Cdk2/cyclin A, intrinsic flexibility enables p27 to integrate and process signaling inputs, and generate outputs including altered Cdk2 activity, p27 stability, and, ultimately, cell cycle progression. Intrinsic dynamics within multi-component assemblies may be a general mechanism of signaling by regulatory IDPs, which can be subverted in human disease.- Published
- 2019
- Full Text
- View/download PDF
32. CDK2 inhibitors as candidate therapeutics for cisplatin- and noise-induced hearing loss.
- Author
-
Teitz T, Fang J, Goktug AN, Bonga JD, Diao S, Hazlitt RA, Iconaru L, Morfouace M, Currier D, Zhou Y, Umans RA, Taylor MR, Cheng C, Min J, Freeman B, Peng J, Roussel MF, Kriwacki R, Guy RK, Chen T, and Zuo J
- Subjects
- Animals, Benzazepines pharmacology, Benzazepines therapeutic use, Cell Death drug effects, Cell Line, Tumor, Cyclin-Dependent Kinase 2 metabolism, Cytoprotection drug effects, Drug Resistance, Germ Cells metabolism, Hair Cells, Auditory drug effects, Hair Cells, Auditory pathology, Indoles pharmacology, Indoles therapeutic use, Lateral Line System drug effects, Lateral Line System pathology, Mice, Inbred C57BL, Mice, Knockout, Mitochondria metabolism, Rats, Reactive Oxygen Species metabolism, Small Molecule Libraries analysis, Zebrafish, Cisplatin adverse effects, Cyclin-Dependent Kinase 2 antagonists & inhibitors, Hearing Loss, Noise-Induced chemically induced, Hearing Loss, Noise-Induced drug therapy, Protein Kinase Inhibitors therapeutic use
- Abstract
Hearing loss caused by aging, noise, cisplatin toxicity, or other insults affects 360 million people worldwide, but there are no Food and Drug Administration-approved drugs to prevent or treat it. We screened 4,385 small molecules in a cochlear cell line and identified 10 compounds that protected against cisplatin toxicity in mouse cochlear explants. Among them, kenpaullone, an inhibitor of multiple kinases, including cyclin-dependent kinase 2 (CDK2), protected zebrafish lateral-line neuromasts from cisplatin toxicity and, when delivered locally, protected adult mice and rats against cisplatin- and noise-induced hearing loss. CDK2-deficient mice displayed enhanced resistance to cisplatin toxicity in cochlear explants and to cisplatin- and noise-induced hearing loss in vivo. Mechanistically, we showed that kenpaullone directly inhibits CDK2 kinase activity and reduces cisplatin-induced mitochondrial production of reactive oxygen species, thereby enhancing cell survival. Our experiments have revealed the proapoptotic function of CDK2 in postmitotic cochlear cells and have identified promising therapeutics for preventing hearing loss., (© 2018 Teitz et al.)
- Published
- 2018
- Full Text
- View/download PDF
33. Direct detection of carbon and nitrogen nuclei for high-resolution analysis of intrinsically disordered proteins using NMR spectroscopy.
- Author
-
Gibbs EB and Kriwacki RW
- Subjects
- Humans, Intrinsically Disordered Proteins analysis, Intrinsically Disordered Proteins metabolism, Nuclear Proteins analysis, Nuclear Proteins chemistry, Nuclear Proteins metabolism, Nucleophosmin, Protein Conformation, Proto-Oncogene Proteins c-mdm2 analysis, Proto-Oncogene Proteins c-mdm2 chemistry, Proto-Oncogene Proteins c-mdm2 metabolism, Carbon analysis, Intrinsically Disordered Proteins chemistry, Magnetic Resonance Spectroscopy methods, Nitrogen analysis
- Abstract
Nuclear magnetic resonance spectroscopy (NMR) is a powerful technique for characterizing the structural and dynamic properties of intrinsically disordered proteins and protein regions (IDPs & IDRs). However, the application of NMR to IDPs has been limited by poor chemical shift dispersion in two-dimensional (2D)
1 H-15 N heteronuclear correlation spectra. Among the various detection schemes available for heteronuclear correlation spectroscopy,13 C direct-detection has become a mainstay for investigations of IDPs owing to the favorable chemical shift dispersion in 2D13 C'-15 N correlation spectra. Recent advances in cryoprobe technology have enhanced the sensitivity for direct detection of both13 C and15 N resonances at high magnetic field strengths, thus prompting the development of15 N direct-detect experiments to complement established13 C-detection experiments. However, the application of15 N-detection has not been widely explored for IDPs. Here we compare1 H,13 C, and15 N detection schemes for a variety of 2D heteronuclear correlation spectra and evaluate their performance on the basis of resolution, chemical shift dispersion, and sensitivity. We performed experiments with a variety of disordered systems ranging in size and complexity; from a small IDR (99 amino acids), to a large low complexity IDR (185 amino acids), and finally a ∼73 kDa folded homopentameric protein that also contains disordered regions (133 amino acids/monomer). We conclude that, while requiring high sample concentration and long acquisition times,15 N-detection often offers enhanced resolution over other detection schemes in studies of disordered protein regions with low complexity sequences., (Copyright © 2018 Elsevier Inc. All rights reserved.)- Published
- 2018
- Full Text
- View/download PDF
34. An unexpected protein interaction promotes drug resistance in leukemia.
- Author
-
Pitre A, Ge Y, Lin W, Wang Y, Fukuda Y, Temirov J, Phillips AH, Peters JL, Fan Y, Ma J, Nourse A, Sinha C, Lin H, Kriwacki R, Downing JR, Gruber TA, Centonze VE, Naren AP, Chen T, and Schuetz JD
- Subjects
- Acute Disease, Animals, Antimycin A pharmacology, Blood Proteins genetics, Cell Line, Tumor, Female, HEK293 Cells, Hematopoietic Stem Cells metabolism, Humans, Kaplan-Meier Estimate, Leukemia, Myeloid genetics, Leukemia, Myeloid pathology, Membrane Proteins genetics, Mice, Multidrug Resistance-Associated Proteins genetics, Protein Binding drug effects, Blood Proteins metabolism, Drug Resistance, Neoplasm, Leukemia, Myeloid metabolism, Membrane Proteins metabolism, Multidrug Resistance-Associated Proteins metabolism
- Abstract
The overall survival of patients with acute myeloid leukemia (AML) is poor and identification of new disease-related therapeutic targets remains a major goal for this disease. Here we show that expression of MPP1, a PDZ-domain-containing protein, highly correlated with ABCC4 in AML, is associated with worse overall survival in AML. Murine hematopoietic progenitor cells overexpressing MPP1 acquired the ability to serially replate in methylcellulose culture, a property crucially dependent upon ABCC4. The highly conserved PDZ-binding motif of ABCC4 is required for ABCC4 and MPP1 to form a protein complex, which increased ABCC4 membrane localization and retention, to enhance drug resistance. Specific disruption of this protein complex, either genetically or chemically, removed ABCC4 from the plasma membrane, increased drug sensitivity, and abrogated MPP1-dependent hematopoietic progenitor cell replating in methylcellulose. High-throughput screening identified Antimycin A as a small molecule that disrupted the ABCC4-MPP1 protein complex and reversed drug resistance in AML cell lines and in primary patient AML cells. In all, targeting the ABCC4-MPP1 protein complex can lead to new therapies to improve treatment outcome of AML, a disease where the long-term prognosis is poor.
- Published
- 2017
- Full Text
- View/download PDF
35. The whole-genome landscape of medulloblastoma subtypes.
- Author
-
Northcott PA, Buchhalter I, Morrissy AS, Hovestadt V, Weischenfeldt J, Ehrenberger T, Gröbner S, Segura-Wang M, Zichner T, Rudneva VA, Warnatz HJ, Sidiropoulos N, Phillips AH, Schumacher S, Kleinheinz K, Waszak SM, Erkek S, Jones DTW, Worst BC, Kool M, Zapatka M, Jäger N, Chavez L, Hutter B, Bieg M, Paramasivam N, Heinold M, Gu Z, Ishaque N, Jäger-Schmidt C, Imbusch CD, Jugold A, Hübschmann D, Risch T, Amstislavskiy V, Gonzalez FGR, Weber UD, Wolf S, Robinson GW, Zhou X, Wu G, Finkelstein D, Liu Y, Cavalli FMG, Luu B, Ramaswamy V, Wu X, Koster J, Ryzhova M, Cho YJ, Pomeroy SL, Herold-Mende C, Schuhmann M, Ebinger M, Liau LM, Mora J, McLendon RE, Jabado N, Kumabe T, Chuah E, Ma Y, Moore RA, Mungall AJ, Mungall KL, Thiessen N, Tse K, Wong T, Jones SJM, Witt O, Milde T, Von Deimling A, Capper D, Korshunov A, Yaspo ML, Kriwacki R, Gajjar A, Zhang J, Beroukhim R, Fraenkel E, Korbel JO, Brors B, Schlesner M, Eils R, Marra MA, Pfister SM, Taylor MD, and Lichter P
- Subjects
- Carcinogenesis genetics, Carrier Proteins genetics, Cohort Studies, DNA Methylation, Datasets as Topic, Epistasis, Genetic, Genomics, Humans, Molecular Targeted Therapy, Muscle Proteins genetics, Mutation, Oncogenes genetics, Transcription Factors genetics, Wnt Proteins genetics, DNA Mutational Analysis, Genome, Human genetics, Medulloblastoma classification, Medulloblastoma genetics, Whole Genome Sequencing
- Abstract
Current therapies for medulloblastoma, a highly malignant childhood brain tumour, impose debilitating effects on the developing child, and highlight the need for molecularly targeted treatments with reduced toxicity. Previous studies have been unable to identify the full spectrum of driver genes and molecular processes that operate in medulloblastoma subgroups. Here we analyse the somatic landscape across 491 sequenced medulloblastoma samples and the molecular heterogeneity among 1,256 epigenetically analysed cases, and identify subgroup-specific driver alterations that include previously undiscovered actionable targets. Driver mutations were confidently assigned to most patients belonging to Group 3 and Group 4 medulloblastoma subgroups, greatly enhancing previous knowledge. New molecular subtypes were differentially enriched for specific driver events, including hotspot in-frame insertions that target KBTBD4 and 'enhancer hijacking' events that activate PRDM6. Thus, the application of integrative genomics to an extensive cohort of clinical samples derived from a single childhood cancer entity revealed a series of cancer genes and biologically relevant subtype diversity that represent attractive therapeutic targets for the treatment of patients with medulloblastoma.
- Published
- 2017
- Full Text
- View/download PDF
36. The landscape of somatic mutations in infant MLL-rearranged acute lymphoblastic leukemias.
- Author
-
Andersson AK, Ma J, Wang J, Chen X, Gedman AL, Dang J, Nakitandwe J, Holmfeldt L, Parker M, Easton J, Huether R, Kriwacki R, Rusch M, Wu G, Li Y, Mulder H, Raimondi S, Pounds S, Kang G, Shi L, Becksfort J, Gupta P, Payne-Turner D, Vadodaria B, Boggs K, Yergeau D, Manne J, Song G, Edmonson M, Nagahawatte P, Wei L, Cheng C, Pei D, Sutton R, Venn NC, Chetcuti A, Rush A, Catchpoole D, Heldrup J, Fioretos T, Lu C, Ding L, Pui CH, Shurtleff S, Mullighan CG, Mardis ER, Wilson RK, Gruber TA, Zhang J, and Downing JR
- Subjects
- Allelic Imbalance genetics, Cohort Studies, DNA Mutational Analysis, Gene Frequency, Histone-Lysine N-Methyltransferase, Humans, Infant, Oncogene Proteins, Fusion genetics, Phosphatidylinositol 3-Kinases genetics, Phosphatidylinositol 3-Kinases metabolism, Precursor Cell Lymphoblastic Leukemia-Lymphoma epidemiology, Protein-Tyrosine Kinases genetics, Protein-Tyrosine Kinases metabolism, Signal Transduction genetics, ras Proteins genetics, ras Proteins metabolism, Mutation, Myeloid-Lymphoid Leukemia Protein genetics, Precursor Cell Lymphoblastic Leukemia-Lymphoma genetics
- Abstract
Infant acute lymphoblastic leukemia (ALL) with MLL rearrangements (MLL-R) represents a distinct leukemia with a poor prognosis. To define its mutational landscape, we performed whole-genome, exome, RNA and targeted DNA sequencing on 65 infants (47 MLL-R and 18 non-MLL-R cases) and 20 older children (MLL-R cases) with leukemia. Our data show that infant MLL-R ALL has one of the lowest frequencies of somatic mutations of any sequenced cancer, with the predominant leukemic clone carrying a mean of 1.3 non-silent mutations. Despite this paucity of mutations, we detected activating mutations in kinase-PI3K-RAS signaling pathway components in 47% of cases. Surprisingly, these mutations were often subclonal and were frequently lost at relapse. In contrast to infant cases, MLL-R leukemia in older children had more somatic mutations (mean of 6.5 mutations/case versus 1.3 mutations/case, P = 7.15 × 10(-5)) and had frequent mutations (45%) in epigenetic regulators, a category of genes that, with the exception of MLL, was rarely mutated in infant MLL-R ALL.
- Published
- 2015
- Full Text
- View/download PDF
37. Conditionally and transiently disordered proteins: awakening cryptic disorder to regulate protein function.
- Author
-
Jakob U, Kriwacki R, and Uversky VN
- Subjects
- Humans, Protein Processing, Post-Translational, Protein Unfolding, Structure-Activity Relationship, Intrinsically Disordered Proteins chemistry, Intrinsically Disordered Proteins physiology
- Published
- 2014
- Full Text
- View/download PDF
38. The genomic landscape of diffuse intrinsic pontine glioma and pediatric non-brainstem high-grade glioma.
- Author
-
Wu G, Diaz AK, Paugh BS, Rankin SL, Ju B, Li Y, Zhu X, Qu C, Chen X, Zhang J, Easton J, Edmonson M, Ma X, Lu C, Nagahawatte P, Hedlund E, Rusch M, Pounds S, Lin T, Onar-Thomas A, Huether R, Kriwacki R, Parker M, Gupta P, Becksfort J, Wei L, Mulder HL, Boggs K, Vadodaria B, Yergeau D, Russell JC, Ochoa K, Fulton RS, Fulton LL, Jones C, Boop FA, Broniscer A, Wetmore C, Gajjar A, Ding L, Mardis ER, Wilson RK, Taylor MR, Downing JR, Ellison DW, Zhang J, and Baker SJ
- Subjects
- Animals, Child, Cohort Studies, Computational Biology, Gene Expression Profiling, Gene Fusion genetics, Humans, Immunoblotting, Immunohistochemistry, Microarray Analysis, Receptor, trkA genetics, Receptor, trkB genetics, Receptor, trkC genetics, Reverse Transcriptase Polymerase Chain Reaction, Sequence Analysis, DNA, Statistics, Nonparametric, Zebrafish, Activin Receptors, Type I genetics, Brain Stem Neoplasms genetics, Glioma genetics, Signal Transduction genetics
- Abstract
Pediatric high-grade glioma (HGG) is a devastating disease with a less than 20% survival rate 2 years after diagnosis. We analyzed 127 pediatric HGGs, including diffuse intrinsic pontine gliomas (DIPGs) and non-brainstem HGGs (NBS-HGGs), by whole-genome, whole-exome and/or transcriptome sequencing. We identified recurrent somatic mutations in ACVR1 exclusively in DIPGs (32%), in addition to previously reported frequent somatic mutations in histone H3 genes, TP53 and ATRX, in both DIPGs and NBS-HGGs. Structural variants generating fusion genes were found in 47% of DIPGs and NBS-HGGs, with recurrent fusions involving the neurotrophin receptor genes NTRK1, NTRK2 and NTRK3 in 40% of NBS-HGGs in infants. Mutations targeting receptor tyrosine kinase-RAS-PI3K signaling, histone modification or chromatin remodeling, and cell cycle regulation were found in 68%, 73% and 59% of pediatric HGGs, respectively, including in DIPGs and NBS-HGGs. This comprehensive analysis provides insights into the unique and shared pathways driving pediatric HGG within and outside the brainstem.
- Published
- 2014
- Full Text
- View/download PDF
39. Broadly protective protein-based pneumococcal vaccine composed of pneumolysin toxoid-CbpA peptide recombinant fusion protein.
- Author
-
Mann B, Thornton J, Heath R, Wade KR, Tweten RK, Gao G, El Kasmi K, Jordan JB, Mitrea DM, Kriwacki R, Maisonneuve J, Alderson M, and Tuomanen EI
- Subjects
- Animals, Antibodies, Bacterial blood, Bacterial Proteins genetics, Cross Protection, Disease Models, Animal, Female, Haemophilus influenzae immunology, Mice, Mice, Inbred BALB C, Neisseria meningitidis immunology, Pneumococcal Vaccines administration & dosage, Pneumococcal Vaccines genetics, Recombinant Fusion Proteins genetics, Recombinant Fusion Proteins immunology, Streptococcus pneumoniae immunology, Streptolysins genetics, Toxoids genetics, Vaccines, Synthetic administration & dosage, Vaccines, Synthetic genetics, Vaccines, Synthetic immunology, Bacterial Proteins immunology, Carrier State prevention & control, Pneumococcal Infections prevention & control, Pneumococcal Vaccines immunology, Streptolysins immunology, Toxoids immunology
- Abstract
Background: Pneumococcus, meningococcus, and Haemophilus influenzae cause a similar spectrum of infections in the ear, lung, blood, and brain. They share cross-reactive antigens that bind to the laminin receptor of the blood-brain barrier as a molecular basis for neurotropism, and this step in pathogenesis was addressed in vaccine design., Methods: Biologically active peptides derived from choline-binding protein A (CbpA) of pneumococcus were identified and then genetically fused to L460D pneumolysoid. The fusion construct was tested for vaccine efficacy in mouse models of nasopharyngeal carriage, otitis media, pneumonia, sepsis, and meningitis., Results: The CbpA peptide-L460D pneumolysoid fusion protein was more broadly immunogenic than pneumolysoid alone, and antibodies were active in vitro against Streptococcus pneumoniae, Neisseria meningitidis, and H. influenzae. Passive and active immunization protected mice from pneumococcal carriage, otitis media, pneumonia, bacteremia, meningitis, and meningococcal sepsis., Conclusions: The CbpA peptide-L460D pneumolysoid fusion protein was broadly protective against pneumococcal infection, with the potential for additional protection against other meningeal pathogens.
- Published
- 2014
- Full Text
- View/download PDF
40. Whole-genome sequencing identifies genetic alterations in pediatric low-grade gliomas.
- Author
-
Zhang J, Wu G, Miller CP, Tatevossian RG, Dalton JD, Tang B, Orisme W, Punchihewa C, Parker M, Qaddoumi I, Boop FA, Lu C, Kandoth C, Ding L, Lee R, Huether R, Chen X, Hedlund E, Nagahawatte P, Rusch M, Boggs K, Cheng J, Becksfort J, Ma J, Song G, Li Y, Wei L, Wang J, Shurtleff S, Easton J, Zhao D, Fulton RS, Fulton LL, Dooling DJ, Vadodaria B, Mulder HL, Tang C, Ochoa K, Mullighan CG, Gajjar A, Kriwacki R, Sheer D, Gilbertson RJ, Mardis ER, Wilson RK, Downing JR, Baker SJ, and Ellison DW
- Subjects
- Adolescent, Animals, Base Sequence, Brain Neoplasms pathology, Child, Child, Preschool, Female, Gene Duplication, Gene Rearrangement, Genes, myb, Genome-Wide Association Study, Glioma pathology, Humans, Infant, Male, Mice, Mice, Nude, Molecular Sequence Data, Mutation, Neoplasm Grading, Neoplasm Transplantation, Proto-Oncogene Proteins genetics, Proto-Oncogene Proteins B-raf, Receptor, Fibroblast Growth Factor, Type 1 genetics, Sequence Analysis, DNA, Signal Transduction, Trans-Activators genetics, Transcriptome, Brain Neoplasms genetics, Glioma genetics
- Abstract
The most common pediatric brain tumors are low-grade gliomas (LGGs). We used whole-genome sequencing to identify multiple new genetic alterations involving BRAF, RAF1, FGFR1, MYB, MYBL1 and genes with histone-related functions, including H3F3A and ATRX, in 39 LGGs and low-grade glioneuronal tumors (LGGNTs). Only a single non-silent somatic alteration was detected in 24 of 39 (62%) tumors. Intragenic duplications of the portion of FGFR1 encoding the tyrosine kinase domain (TKD) and rearrangements of MYB were recurrent and mutually exclusive in 53% of grade II diffuse LGGs. Transplantation of Trp53-null neonatal astrocytes expressing FGFR1 with the duplication involving the TKD into the brains of nude mice generated high-grade astrocytomas with short latency and 100% penetrance. FGFR1 with the duplication induced FGFR1 autophosphorylation and upregulation of the MAPK/ERK and PI3K pathways, which could be blocked by specific inhibitors. Focusing on the therapeutically challenging diffuse LGGs, our study of 151 tumors has discovered genetic alterations and potential therapeutic targets across the entire range of pediatric LGGs and LGGNTs.
- Published
- 2013
- Full Text
- View/download PDF
41. Novel mutations target distinct subgroups of medulloblastoma.
- Author
-
Robinson G, Parker M, Kranenburg TA, Lu C, Chen X, Ding L, Phoenix TN, Hedlund E, Wei L, Zhu X, Chalhoub N, Baker SJ, Huether R, Kriwacki R, Curley N, Thiruvenkatam R, Wang J, Wu G, Rusch M, Hong X, Becksfort J, Gupta P, Ma J, Easton J, Vadodaria B, Onar-Thomas A, Lin T, Li S, Pounds S, Paugh S, Zhao D, Kawauchi D, Roussel MF, Finkelstein D, Ellison DW, Lau CC, Bouffet E, Hassall T, Gururangan S, Cohn R, Fulton RS, Fulton LL, Dooling DJ, Ochoa K, Gajjar A, Mardis ER, Wilson RK, Downing JR, Zhang J, and Gilbertson RJ
- Subjects
- Animals, Antigens, CD, CREB-Binding Protein genetics, Cadherins genetics, Cdh1 Proteins, Cell Cycle Proteins deficiency, Cell Cycle Proteins genetics, Cell Lineage, Cerebellar Neoplasms pathology, Child, Class I Phosphatidylinositol 3-Kinases, DEAD-box RNA Helicases genetics, DNA Copy Number Variations, DNA Helicases genetics, DNA Mutational Analysis, Disease Models, Animal, Genome, Human genetics, Genomics, Hedgehog Proteins metabolism, Histone Demethylases genetics, Histones metabolism, Humans, Medulloblastoma pathology, Methylation, Mice, Nuclear Proteins genetics, Phosphatidylinositol 3-Kinases genetics, Transcription Factors genetics, Wnt Proteins metabolism, beta Catenin genetics, Cerebellar Neoplasms classification, Cerebellar Neoplasms genetics, Medulloblastoma classification, Medulloblastoma genetics, Mutation genetics
- Abstract
Medulloblastoma is a malignant childhood brain tumour comprising four discrete subgroups. Here, to identify mutations that drive medulloblastoma, we sequenced the entire genomes of 37 tumours and matched normal blood. One-hundred and thirty-six genes harbouring somatic mutations in this discovery set were sequenced in an additional 56 medulloblastomas. Recurrent mutations were detected in 41 genes not yet implicated in medulloblastoma; several target distinct components of the epigenetic machinery in different disease subgroups, such as regulators of H3K27 and H3K4 trimethylation in subgroups 3 and 4 (for example, KDM6A and ZMYM3), and CTNNB1-associated chromatin re-modellers in WNT-subgroup tumours (for example, SMARCA4 and CREBBP). Modelling of mutations in mouse lower rhombic lip progenitors that generate WNT-subgroup tumours identified genes that maintain this cell lineage (DDX3X), as well as mutated genes that initiate (CDH1) or cooperate (PIK3CA) in tumorigenesis. These data provide important new insights into the pathogenesis of medulloblastoma subgroups and highlight targets for therapeutic development.
- Published
- 2012
- Full Text
- View/download PDF
42. Probing the role of nascent helicity in p27 function as a cell cycle regulator.
- Author
-
Otieno S and Kriwacki R
- Subjects
- Amino Acid Sequence, Animals, Binding Sites, Cyclin A chemistry, Cyclin A metabolism, Cyclin-Dependent Kinase Inhibitor p21 chemistry, Cyclin-Dependent Kinase Inhibitor p21 metabolism, Cyclin-Dependent Kinase Inhibitor p27 physiology, Humans, Mice, Molecular Sequence Data, NIH 3T3 Cells, Protein Structure, Tertiary, Sequence Alignment, Structure-Activity Relationship, Thermodynamics, Cyclin-Dependent Kinase Inhibitor p27 chemistry
- Abstract
p27 regulates the activity of Cdk complexes which are the principal governors of phase transitions during cell division. Members of the p27 family of proteins, which also includes p21 and p57, are called the Cip/Kip cyclin-dependent kinase regulators (CKRs). Interestingly, the Cip/Kip CKRs play critical roles in cell cycle regulation by being intrinsically unstructured, a characteristic contrary to the classical structure-function paradigm. They exhibit nascent helicity which has been localized to a segment referred to as sub-domain LH. The nascent helicity of this sub-domain is conserved and we hypothesize that it is an important determinant of their functional properties. To test this hypothesis, we successfully designed and prepared p27 variants in which domain LH was either more or less helical with respect to the wild-type protein. Thermal denaturation experiments showed that the ternary complexes of the p27 variants bound to Cdk2/Cyclin A were less stable compared to the wild-type complex. Isothermal titration calorimetry experiments showed a decrease in the enthalpy of binding for all the mutants with respect to p27. The free energies of binding varied within a much narrower range. In vitro Cdk2 inhibition assays showed that the p27 variants exhibited disparate inhibitory potencies. Furthermore, when over-expressed in NIH 3T3 mouse fibroblast cells, the less helical p27 variants were less effective in causing cell cycle arrest relative to the wild-type p27. Our results indicate that the nascent helicity of sub-domain LH plays a key role mediating the biological function of p27.
- Published
- 2012
- Full Text
- View/download PDF
43. Crystallographic and NMR analyses of UvsW and UvsW.1 from bacteriophage T4.
- Author
-
Kerr ID, Sivakolundu S, Li Z, Buchsbaum JC, Knox LA, Kriwacki R, and White SW
- Subjects
- Crystallography, X-Ray, DNA Helicases metabolism, DNA Repair physiology, DNA, Single-Stranded chemistry, DNA, Single-Stranded metabolism, Escherichia coli Proteins chemistry, Escherichia coli Proteins metabolism, Eukaryotic Cells enzymology, Genome, Viral physiology, Nuclear Magnetic Resonance, Biomolecular, Protein Structure, Secondary physiology, Protein Structure, Tertiary physiology, Rec A Recombinases chemistry, Rec A Recombinases metabolism, RecQ Helicases chemistry, RecQ Helicases metabolism, Recombination, Genetic physiology, Structural Homology, Protein, Viral Proteins metabolism, Virus Replication physiology, Bacteriophage T4 enzymology, DNA Helicases chemistry, Viral Proteins chemistry
- Abstract
The uvsWXY system is implicated in the replication and repair of the bacteriophage T4 genome. Whereas the roles of the recombinase (UvsX) and the recombination mediator protein (UvsY) are known, the precise role of UvsW is unclear. Sequence analysis identifies UvsW as a member of the monomeric SF2 helicase superfamily that translocates nucleic acid substrates via the action of two RecA-like motor domains. Functional homologies to Escherichia coli RecG and biochemical analyses have shown that UvsW interacts with branched nucleic acid substrates, suggesting roles in recombination and the rescue of stalled replication forks. A sequencing error at the 3'-end of the uvsW gene has revealed a second, short open reading frame that encodes a protein of unknown function called UvsW.1. We have determined the crystal structure of UvsW to 2.7A and the NMR solution structure of UvsW.1. UvsW has a four-domain architecture with structural homology to the eukaryotic SF2 helicase, Rad54. A model of the UvsW-ssDNA complex identifies structural elements and conserved residues that may interact with nucleic acid substrates. The NMR solution structure of UvsW.1 reveals a dynamic four-helix bundle with homology to the structure-specific nucleic acid binding module of RecQ helicases.
- Published
- 2007
- Full Text
- View/download PDF
44. Germline TP53 R337H mutation is not sufficient to establish Li-Fraumeni or Li-Fraumeni-like syndrome.
- Author
-
Ribeiro RC, Rodriguez-Galindo C, Figueiredo BC, Mastellaro MJ, West AN, Kriwacki R, and Zambetti GP
- Subjects
- Humans, Genes, p53, Li-Fraumeni Syndrome genetics, Mutation
- Published
- 2007
- Full Text
- View/download PDF
45. Identification of a novel germ line variant hotspot mutant p53-R175L in pediatric adrenal cortical carcinoma.
- Author
-
West AN, Ribeiro RC, Jenkins J, Rodriguez-Galindo C, Figueiredo BC, Kriwacki R, and Zambetti GP
- Subjects
- Adrenal Cortex Neoplasms pathology, Adult, Apoptosis genetics, Cell Growth Processes genetics, Child, Child, Preschool, Female, Humans, Male, Models, Molecular, Pedigree, Adrenal Cortex Neoplasms genetics, Genes, p53 genetics, Germ-Line Mutation
- Abstract
Hotspot mutations in the p53 tumor suppressor gene result in the disruption of DNA contact points or alter the overall structure of the protein to prevent DNA binding. When inherited, hotspot mutants are associated with Li-Fraumeni syndrome (LFS), a familial cancer predisposition. One of the most common hotspot mutations occurs at codon 175, resulting in an arginine to histidine substitution. We have identified a novel germ line variant of the 175 mutant (Arg to Leu; R175L) in a pediatric patient who developed adrenal cortical carcinoma. Surprisingly, the family is not tumor prone or associated with LFS. In vitro, the R175L mutant displayed an attenuated tumor suppressor activity in the regulation of transcription, colony formation, and apoptosis when compared with wild-type p53 and the R175H mutant. These findings suggest that p53-R175L retains sufficient activity to suppress LFS, but not adrenal cortical carcinoma. Therefore, not all hotspot mutants are functionally equivalent and the biochemical nature of the mutant may significantly influence clinical outcome. The implications of these results for genetic counseling are discussed.
- Published
- 2006
- Full Text
- View/download PDF
46. Defining the molecular basis of Arf and Hdm2 interactions.
- Author
-
Bothner B, Lewis WS, DiGiammarino EL, Weber JD, Bothner SJ, and Kriwacki RW
- Subjects
- 3T3 Cells, Amino Acid Motifs, Amino Acid Sequence, Animals, Binding Sites, Cell Nucleolus chemistry, Cell Nucleolus metabolism, Circular Dichroism, Cyclin-Dependent Kinase Inhibitor p16, Gene Deletion, Humans, Magnetic Resonance Spectroscopy, Mice, Molecular Sequence Data, Peptide Fragments chemistry, Peptide Fragments metabolism, Protein Binding, Protein Interaction Mapping, Protein Structure, Secondary, Protein Structure, Tertiary, Proto-Oncogene Proteins c-mdm2, Sequence Alignment, Surface Plasmon Resonance, Tumor Suppressor Protein p14ARF genetics, Tumor Suppressor Protein p53 metabolism, Nuclear Proteins, Proto-Oncogene Proteins chemistry, Proto-Oncogene Proteins metabolism, Tumor Suppressor Protein p14ARF chemistry, Tumor Suppressor Protein p14ARF metabolism
- Abstract
Understanding the interaction of Arf and Hdm2 has recently become a central issue in cancer biology. In response to hyperproliferative signals, p14(Arf) stabilizes p53 by binding to Hdm2 and inhibits the ubiquitination and subsequent proteosome-dependent degradation of p53. The medical importance of the Arf-Hdm2-p53 regulatory system is highlighted by the finding that either p53 or p14(Arf) are lost or modified in virtually all human cancers. Isolated Arf and Hdm2 domains are dynamically disordered in solution, yet they retain the ability to interact in vitro and in cellular assays. Upon binding, domains of both Arf and Hdm2 undergo a dramatic transition from disordered conformations to extended structures comprised of beta-strands. The presence of domains from both proteins are necessary and sufficient for the formation of the highly stable extended beta structures. We have mapped sites within Arf and Hdm2 that interact at a resolution of five amino acid residues using surface plasmon resonance. Surface plasmon resonance and circular dichroism spectropolarimetry confirm the presence of multiple interaction domains within each protein. Both p14(Arf) (human) and p19(Arf) (mouse) interact with Hdm2 through two short motifs present in their N termini. The Arf interacting region of Hdm2 is also composed of two short sequences located in the central acidic domain, between residues 235-264 and 270-289. The binding-induced structural transition is also induced by short peptides, 15 amino acids in length, that contain the binding motifs. Micro-injection and live cell imaging of proteins tagged with fluorescent labels was used to confirm the in vivo function of the interaction domains. Arf and Hdm2 thus appear to interact through a novel mechanism that exerts control over the cell division cycle. The novel molecular mechanism of interaction and the limited size of the protein domains involved provide opportunities for the development of anticancer therapeutics., (Copyright 2001 Academic Press.)
- Published
- 2001
- Full Text
- View/download PDF
47. Solution structure of the transcriptional activation domain of the bacteriophage T4 protein, MotA.
- Author
-
Li N, Zhang W, White SW, and Kriwacki RW
- Subjects
- Amino Acid Sequence, Chromatography, Gel, Crystallography, X-Ray, Dimerization, Light, Molecular Sequence Data, Nuclear Magnetic Resonance, Biomolecular, Protein Structure, Secondary, Protein Structure, Tertiary, Scattering, Radiation, Solutions, Bacteriophage T4 chemistry, DNA-Binding Proteins chemistry, Peptide Fragments chemistry, Trans-Activators chemistry, Transcription Factors chemistry, Viral Proteins chemistry
- Abstract
Bacteriophage T4 encodes a transcription factor, MotA, that binds to the -30 region of middle-mode promoters and activates transcription by host RNA polymerase. The crystal structure of the N-terminal domain of MotA (MotNF) revealed a six-helix domain in which the two C-terminal alpha-helices mediate the formation of a dimer via a coiled-coil motif and hydrophobic interactions. This structure suggested that full-length MotA binds DNA as a dimer, but subsequent biochemical results have shown that a monomeric form of MotA binds DNA. In this study, gel filtration chromatography, dynamic light scattering, and NMR-based diffusion measurements show conclusively that MotNF is a monomer, and not a dimer, in solution. In addition, we have determined the monomeric solution structure of MotNF using NMR spectroscopy, and have compared this with the dimer structure observed in crystals. The core of the protein assumes the same helical conformation in solution and in crystals, but important differences are observed at the extreme C-terminus. In solution, helix alpha5 is followed by five disordered residues that probably link the N-terminal and C-terminal domains of MotA. In crystals, helix alpha5 forms the dimer interface and is followed by a short sixth helix that further stabilizes the dimer configuration. The solution structure of MotNF supports the conclusion that MotA functions as a monomer, and suggests that the existence of the sixth helix in crystals is a consequence of crystal packing. Our work highlights the importance of investigating protein structures in both crystals and solution to fully understand biomolecular structure and to accurately deduce relationships between structure and function.
- Published
- 2001
- Full Text
- View/download PDF
48. Solution structure of the p53 regulatory domain of the p19Arf tumor suppressor protein.
- Author
-
DiGiammarino EL, Filippov I, Weber JD, Bothner B, and Kriwacki RW
- Subjects
- 3T3 Cells, Amino Acid Sequence, Animals, Cell Line, Circular Dichroism, Genes, Tumor Suppressor, Mice, Mice, Knockout, Molecular Sequence Data, Nuclear Magnetic Resonance, Biomolecular, Peptide Fragments genetics, Protein Structure, Tertiary genetics, Proteins genetics, Recombinant Proteins biosynthesis, Recombinant Proteins chemistry, Solutions, Solvents, Trifluoroethanol, Tumor Suppressor Protein p14ARF, Peptide Fragments chemistry, Peptide Fragments physiology, Proteins chemistry, Proteins physiology, Tumor Suppressor Protein p53 metabolism
- Abstract
Arf is a tumor suppressor that regulates p53 function and is a frequent target for loss in human cancers. Through two novel mechanisms, Arf inhibits the oncoprotein Hdm2, a negative regulator of p53. (1) Arf inhibits the E3 ubiquitin ligase activity of Hdm2 that leads to p53 degradation, and (2) Arf sequesters Hdm2 within nucleoli. These activities of Arf promote p53-mediated cell cycle arrest and apoptosis. Fundamental to these processes are interactions between Arf and Hdm2. Here we show that a peptide containing the 37 N-terminal amino acids of mouse Arf (mArfN37) localizes to nucleoli, sequesters Hdm2 within nucleoli, and causes cell cycle arrest. Circular dichroism and NMR spectroscopy show that mArfN37 is largely unstructured under aqueous conditions; however, the peptide adopts two alpha-helices (helix 1, residues 4-14; and helix 2, residues 20-29) in 2,2,2-trifluoroethanol (TFE). Each helix contains an amino acid motif that is repeated twice in mArfN37, once in each helix. The two helices, however, do not interact but are connected by an apparently flexible linker. The repeated motif contains Arg residues spaced by a hydrophobic segment that may be involved in Hdm2 recognition and binding. The RRPR nucleolar localization signal, contained within residues 31-34, appears to be disordered under all conditions. The identification of two Arf structural modules suggests that short peptides containing the repeated motif may function as Arf mimics and may allow the design of small molecule Arf mimics in the future.
- Published
- 2001
- Full Text
- View/download PDF
49. Cooperative signals governing ARF-mdm2 interaction and nucleolar localization of the complex.
- Author
-
Weber JD, Kuo ML, Bothner B, DiGiammarino EL, Kriwacki RW, Roussel MF, and Sherr CJ
- Subjects
- 3T3 Cells, Amino Acid Sequence, Animals, Cell Cycle genetics, Cell Division genetics, Fluorescent Antibody Technique, Humans, Mice, Molecular Sequence Data, Mutation, Peptide Fragments metabolism, Protein Binding genetics, Protein Conformation, Proteins genetics, Proto-Oncogene Proteins c-mdm2, Recombinant Proteins, Retroviridae genetics, Transfection, Tumor Suppressor Protein p14ARF, Tumor Suppressor Protein p53 metabolism, Cell Nucleolus metabolism, Nuclear Proteins, Proteins metabolism, Proto-Oncogene Proteins metabolism
- Abstract
The ARF tumor suppressor protein stabilizes p53 by antagonizing its negative regulator, Mdm2 (Hdm2 in humans). Both mouse p19(ARF) and human p14(ARF) bind to the central region of Mdm2 (residues 210 to 304), a segment that does not overlap with its N-terminal p53-binding domain, nuclear import or export signals, or C-terminal RING domain required for Mdm2 E3 ubiquitin ligase activity. The N-terminal 37 amino acids of mouse p19(ARF) are necessary and sufficient for binding to Mdm2, localization of Mdm2 to nucleoli, and p53-dependent cell cycle arrest. Although a nucleolar localization signal (NrLS) maps within a different segment (residues 82 to 101) of the human p14(ARF) protein, binding to Mdm2 and nucleolar import of ARF-Mdm2 complexes are both required for cell cycle arrest induced by either the mouse or human ARF proteins. Because many codons of mouse ARF mRNA are not recognized by the most abundant bacterial tRNAs, we synthesized ARF minigenes containing preferred bacterial codons. Using bacterially produced ARF polypeptides and chemically synthesized peptides conjugated to Sepharose, residues 1 to 14 and 26 to 37 of mouse p19(ARF) were found to interact independently and cooperatively with Mdm2, while residues 15 to 25 were dispensable for binding. Paradoxically, residues 26 to 37 of mouse p19(ARF) are also essential for ARF nucleolar localization in the absence of Mdm2. However, the mobilization of the p19(ARF)-Mdm2 complex into nucleoli also requires a cryptic NrLS within the Mdm2 C-terminal RING domain. The Mdm2 NrLS is unmasked upon ARF binding, and its deletion prevents import of the ARF-Mdm2 complex into nucleoli. Collectively, the results suggest that ARF binding to Mdm2 induces a conformational change that facilitates nucleolar import of the ARF-Mdm2 complex and p53-dependent cell cycle arrest. Hence, the ARF-Mdm2 interaction can be viewed as bidirectional, with each protein being capable of regulating the subnuclear localization of the other.
- Published
- 2000
- Full Text
- View/download PDF
50. Assignment of 1H, 13C and 15N resonances of the I-domain of human leukocyte function associated antigen-1.
- Author
-
Kriwacki RW, Legge GB, Hommel U, Ramage P, Chung J, Tennant LL, Wright PE, and Dyson HJ
- Subjects
- Humans, Intercellular Adhesion Molecule-1 chemistry, Nuclear Magnetic Resonance, Biomolecular, Protein Structure, Tertiary, Recombinant Proteins chemistry, Lymphocyte Function-Associated Antigen-1 chemistry
- Published
- 2000
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.