Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Daisy Bustos is active.

Publication


Featured researches published by Daisy Bustos.


Molecular Cell | 2010

K11-Linked Polyubiquitination in Cell Cycle Control Revealed by a K11 Linkage-Specific Antibody

Marissa L. Matsumoto; Katherine E. Wickliffe; Ken C. Dong; Christine Yu; Ivan Bosanac; Daisy Bustos; Lilian Phu; Donald S. Kirkpatrick; Sarah G. Hymowitz; Michael Rape; Robert F. Kelley; Vishva M. Dixit

Polyubiquitination is a posttranslational modification where ubiquitin chains containing isopeptide bonds linking one of seven ubiquitin lysines with the C terminus of an adjoining ubiquitin are covalently attached to proteins. While functions of K48- and K63-linked polyubiquitin are understood, the role(s) of noncanonical K11-linked chains is less clear. A crystal structure of K11-linked diubiquitin demonstrates a distinct conformation from K48- or K63-linked diubiquitin. We engineered a K11 linkage-specific antibody and use it to demonstrate that K11 chains are highly upregulated in mitotic human cells precisely when substrates of the ubiquitin ligase anaphase-promoting complex (APC/C) are degraded. These chains increased with proteasomal inhibition, suggesting they act as degradation signals in vivo. Inhibition of the APC/C strongly impeded the formation of K11-linked chains, suggesting that a single ubiquitin ligase is the major source of mitotic K11-linked chains. Our results underscore the importance of K11-linked ubiquitin chains as critical regulators of mitotic protein degradation.


Science Translational Medicine | 2012

Ser1292 Autophosphorylation Is an Indicator of LRRK2 Kinase Activity and Contributes to the Cellular Effects of PD Mutations

Zejuan Sheng; Shuo Zhang; Daisy Bustos; Tracy Kleinheinz; Claire E. Le Pichon; Sara L. Dominguez; Hilda Solanoy; Jason Drummond; Xiaolin Zhang; Xiao Ding; Fang Cai; Qinghua Song; Xianting Li; Zhenyu Yue; Marcel van der Brug; Daniel J. Burdick; Janet Gunzner-Toste; Huifen Chen; Xingrong Liu; Anthony A. Estrada; Zachary Kevin Sweeney; Kimberly Scearce-Levie; John Moffat; Donald S. Kirkpatrick; Haitao Zhu

LRRK2 autophosphorylation on Ser1292 may be a useful indicator of kinase activity, providing a readout for screening candidate LRRK2 inhibitors. LRRK2 Inhibitor Heralds a Happier Song Genetic polymorphisms in the leucine-rich repeat kinase 2 (LRRK2) are the most common causes of familial Parkinson’s disease (PD) and are also linked to idiopathic PD. The most prevalent LRRK2 PD mutation G2019S imbues the kinase with a gain of function, suggesting that blocking LRRK2 activity may be a therapeutic strategy for reversing the pathogenic effects of LRRK2 mutations in PD. However, the mechanistic link between LRRK2 kinase activity and the cellular effects of PD mutations remains elusive, and there has been no reliable way to monitor LRRK2 kinase activity in vivo. Using quantitative mass spectrometry and subsequent phospho-specific antibody approaches, Sheng et al. now report that LRRK2 phosphorylates itself on Ser1292 in vitro and in vivo (Ser1292 autophosphorylation). Five of the six confirmed familial LRRK2 PD mutations increased Ser1292 autophosphorylation when transiently expressed in heterologous cells, suggesting increased Ser1292 autophosphorylation as a common feature of LRRK2 PD mutations. Elimination of the Ser1292 autophosphorylation site abrogated the defects on neurite outgrowth caused by LRRK2 PD mutations in cultured rat embryonic neurons. Using Ser1292 autophosphorylation as the readout of kinase activity, Sheng et al. developed assays to monitor LRRK2 kinase activity in cultured cells and rodents. These assays were used to profile the potencies of hundreds of LRRK2 kinase inhibitors derived from high-throughput compound screening. A potent and selective compound that effectively inhibited LRRK2 kinase activity in mouse brains and reversed cellular effects of LRRK2 PD mutations in cultured primary neurons was identified. Mutations in the leucine-rich repeat kinase 2 (LRRK2) gene are the most common cause of familial Parkinson’s disease (PD). Although biochemical studies have shown that certain PD mutations confer elevated kinase activity in vitro on LRRK2, there are no methods available to directly monitor LRRK2 kinase activity in vivo. We demonstrate that LRRK2 autophosphorylation on Ser1292 occurs in vivo and is enhanced by several familial PD mutations including N1437H, R1441G/C, G2019S, and I2020T. Combining two PD mutations together further increases Ser1292 autophosphorylation. Mutation of Ser1292 to alanine (S1292A) ameliorates the effects of LRRK2 PD mutations on neurite outgrowth in cultured rat embryonic primary neurons. Using cell-based and pharmacodynamic assays with phosphorylated Ser1292 as the readout, we developed a brain-penetrating LRRK2 kinase inhibitor that blocks Ser1292 autophosphorylation in vivo and attenuates the cellular consequences of LRRK2 PD mutations in vitro. These data suggest that Ser1292 autophosphorylation may be a useful indicator of LRRK2 kinase activity in vivo and may contribute to the cellular effects of certain PD mutations.


Molecular & Cellular Proteomics | 2011

Improved Quantitative Mass Spectrometry Methods for Characterizing Complex Ubiquitin Signals

Lilian Phu; Anita Izrael-Tomasevic; Marissa L. Matsumoto; Daisy Bustos; Jasmin N. Dynek; Anna V. Fedorova; Corey E. Bakalarski; David Arnott; Kurt Deshayes; Vishva M. Dixit; Robert F. Kelley; Domagoj Vucic; Donald S. Kirkpatrick

Ubiquitinated substrates can be recruited to macromolecular complexes through interactions between their covalently bound ubiquitin (Ub) signals and Ub receptor proteins. To develop a functional understanding of the Ub system in vivo, methods are needed to determine the composition of Ub signals on individual substrates and in protein mixtures. Mass spectrometry has emerged as an important tool for characterizing the various forms of Ub. In the Ubiquitin-AQUA approach, synthetic isotopically labeled internal standard peptides are used to quantify unbranched peptides and the branched -GG signature peptides generated by trypsin digestion of Ub signals. Here we have built upon existing methods and established a comprehensive platform for the characterization of Ub signals. Digested peptides and isotopically labeled standards are analyzed either by selected reaction monitoring on a QTRAP mass spectrometer or by narrow window extracted ion chromatograms on a high resolution LTQ-Orbitrap. Additional peptides are now monitored to account for the N terminus of ubiquitin, linear polyUb chains, the peptides surrounding K33 and K48, and incomplete digestion products. Using this expanded battery of peptides, the total amount of Ub in a sample can be determined from multiple loci within the protein, minimizing possible confounding effects of complex Ub signals, digestion abnormalities, or use of mutant Ub in experiments. These methods have been useful for the characterization of in vitro, multistage ubiquitination and have now been extended to reactions catalyzed by multiple E2 enzymes. One question arising from in vitro studies is whether individual protein substrates in cells may be modified by multiple forms of polyUb. Here we have taken advantage of recently developed polyubiquitin linkage-specific antibodies recognizing K48- and K63-linked polyUb chains, coupled with these mass spectrometry methods, to further evaluate the abundance of mixed linkage Ub substrates in cultured mammalian cells. By combining these two powerful tools, we show that polyubiquitinated substrates purified from cells can be modified by mixtures of K48, K63, and K11 linkages.


Journal of Cell Biology | 2013

JNK-mediated phosphorylation of DLK suppresses its ubiquitination to promote neuronal apoptosis

Sarah Huntwork-Rodriguez; Bei Wang; Trent Watkins; Arundhati Sengupta Ghosh; Christine D. Pozniak; Daisy Bustos; Kim Newton; Donald S. Kirkpatrick; Joseph W. Lewcock

Neuronal injury induces JNK phosphorylation of DLK, which reduces DLK ubiquitination and creates a positive feedback loop to enhance JNK signaling and increase apoptosis.


Journal of Experimental Medicine | 2013

Dual leucine zipper kinase is required for excitotoxicity-induced neuronal degeneration

Christine D. Pozniak; Arundhati Sengupta Ghosh; Alvin Gogineni; Jesse E. Hanson; Seung-Hye Lee; Jessica L. Larson; Hilda Solanoy; Daisy Bustos; Hong Li; Hai Ngu; Adrian M. Jubb; Gai Ayalon; Jiansheng Wu; Kimberly Scearce-Levie; Qiang Zhou; Robby M. Weimer; Donald S. Kirkpatrick; Joseph W. Lewcock

Loss of dual leucine zipper kinase results in attenuated JNK/c-Jun stress response pathway activation and reduced neuronal degeneration after kainic acid–induced excitotoxic seizures.


Journal of Biological Chemistry | 2013

HectD1 E3 Ligase Modifies Adenomatous Polyposis Coli (APC) with Polyubiquitin to Promote the APC-Axin Interaction

Hoanh Tran; Daisy Bustos; Ronald Yeh; Bonnee Rubinfeld; Cynthia Lam; Stephanie Shriver; Inna Zilberleyb; Michelle Lee; Lilian Phu; Anjali A. Sarkar; Irene E. Zohn; Ingrid E. Wertz; Donald S. Kirkpatrick; Paul Polakis

Background: APC is modified with Lys-63-linked polyubiquitin when bound to Axin in an assembled β-catenin destruction complex. Results: HectD1 E3 ligase modifies APC with Lys-63-linked ubiquitin chains to facilitate the APC-Axin interaction. Conclusion: HectD1 is a candidate E3 ligase for APC. Significance: The identification of HectD1 could lead to a better understanding of APC function. The adenomatous polyposis coli (APC) protein functions as a negative regulator of the Wnt signaling pathway. In this capacity, APC forms a “destruction complex” with Axin, CK1α, and GSK3β to foster phosphorylation of the Wnt effector β-catenin earmarking it for Lys-48-linked polyubiquitylation and proteasomal degradation. APC is conjugated with Lys-63-linked ubiquitin chains when it is bound to Axin, but it is unclear whether this modification promotes the APC-Axin interaction or confers upon APC an alternative function in the destruction complex. Here we identify HectD1 as a candidate E3 ubiquitin ligase that modifies APC with Lys-63 polyubiquitin. Knockdown of HectD1 diminished APC ubiquitylation, disrupted the APC-Axin interaction, and augmented Wnt3a-induced β-catenin stabilization and signaling. These results indicate that HectD1 promotes the APC-Axin interaction to negatively regulate Wnt signaling.


Molecular & Cellular Proteomics | 2012

Characterizing Ubiquitination Sites by Peptide-based Immunoaffinity Enrichment

Daisy Bustos; Corey E. Bakalarski; Yanling Yang; Junmin Peng; Donald S. Kirkpatrick

Advances in high resolution tandem mass spectrometry and peptide enrichment technologies have transformed the field of protein biochemistry by enabling analysis of end points that have traditionally been inaccessible to molecular and biochemical techniques. One field benefitting from this research has been the study of ubiquitin, a 76-amino acid protein that functions as a covalent modifier of other proteins. Seminal work performed decades ago revealed that trypsin digestion of a branched protein structure known as A24 yielded an enigmatic diglycine signature bound to a lysine residue in histone 2A. With the onset of mass spectrometry proteomics, identification of K-GG-modified peptides has emerged as an effective way to map the position of ubiquitin modifications on a protein of interest and to quantify the extent of substrate ubiquitination. The initial identification of K-GG peptides by mass spectrometry initiated a flurry of work aimed at enriching these post-translationally modified peptides for identification and quantification en masse. Recently, immunoaffinity reagents have been reported that are capable of capturing K-GG peptides from ubiquitin and its thousands of cellular substrates. Here we focus on the history of K-GG peptides, their identification by mass spectrometry, and the utility of immunoaffinity reagents for studying the mechanisms of cellular regulation by ubiquitin.


Proceedings of the National Academy of Sciences of the United States of America | 2013

Phosphoproteomic characterization of DNA damage response in melanoma cells following MEK/PI3K dual inhibition

Donald S. Kirkpatrick; Daisy Bustos; Taner Dogan; Jocelyn Chan; Lilian Phu; Amy E. Young; Lori Friedman; Marcia Belvin; Qinghua Song; Corey E. Bakalarski; Klaus P. Hoeflich

Significance Growing evidence suggests that successful intervention in many human cancers will require combinations of therapeutic agents. Critical to this effort will be a detailed understanding of the crosstalk between signaling networks that modulate proliferation, cell death, drug sensitivity, and acquired resistance. Here we investigated DNA-damage signaling elicited by small-molecule inhibitors against MAP/ERK kinase (MEK) and PI3K in melanoma cells. This work, performed using cutting-edge mass spectrometry proteomics, uncovered a burst of signaling among proteins in the DNA-damage pathway upon initiation of the cell-death program by agents targeting the RAS–RAF–MEK and PI3K–AKT–mTOR pathways. These signals may prove important to the short- and long-term sensitivity of tumor cells to MEK- and PI3K-targeted therapies. Targeted therapeutics that block signal transduction through the RAS–RAF–MEK and PI3K–AKT–mTOR pathways offer significant promise for the treatment of human malignancies. Dual inhibition of MAP/ERK kinase (MEK) and phosphatidylinositol 3-kinase (PI3K) with the potent and selective small-molecule inhibitors GDC-0973 and GDC-0941 has been shown to trigger tumor cell death in preclinical models. Here we have used phosphomotif antibodies and mass spectrometry (MS) to investigate the effects of MEK/PI3K dual inhibition during the period immediately preceding cell death. Upon treatment, melanoma cell lines responded by dramatically increasing phosphorylation on proteins containing a canonical DNA damage-response (DDR) motif, as defined by a phosphorylated serine or threonine residue adjacent to glutamine, [s/t]Q. In total, >2,000 [s/t]Q phosphorylation sites on >850 proteins were identified by LC-MS/MS, including an extensive network of DDR proteins. Linear mixed-effects modeling revealed 101 proteins in which [s/t]Q phosphorylation was altered significantly in response to GDC-0973/GDC-0941. Among the most dramatic changes, we observed rapid and sustained phosphorylation of sites within the ABCDE cluster of DNA-dependent protein kinase. Preincubation of cells with the inhibitors of the DDR kinases DNA-dependent protein kinase or ataxia-telangiectasia mutated enhanced GDC-0973/GDC-0941–mediated cell death. Network analysis revealed specific enrichment of proteins involved in RNA metabolism along with canonical DDR proteins and suggested a prominent role for this pathway in the response to MEK/PI3K dual inhibition.


Journal of Proteome Research | 2012

Complementary Proteomic Tools for the Dissection of Apoptotic Proteolysis Events

Victoria Pham; Robert M. Pitti; Veronica G. Anania; Corey E. Bakalarski; Daisy Bustos; Suchit Jhunjhunwala; Qui T. Phung; Kebing Yu; William F. Forrest; Donald S. Kirkpatrick; Avi Ashkenazi; Jennie R. Lill

Proteolysis is a key regulatory event that controls intracellular and extracellular signaling through irreversible changes in a proteins structure that greatly alters its function. Here we describe a platform for profiling caspase substrates which encompasses two highly complementary proteomic techniques--the first is a differential gel based approach termed Global Analyzer of SILAC-derived Substrates of Proteolysis (GASSP) and the second involves affinity enrichment of peptides containing a C-terminal aspartic acid residue. In combination, these techniques have enabled the profiling of a large cellular pool of apoptotic-mediated proteolytic events across a wide dynamic range. By applying this integrated proteomic work flow to analyze proteolytic events resulting from the induction of intrinsic apoptosis in Jurkat cells via etoposide treatment, 3346 proteins were quantified, of which 360 proteins were identified as etoposide-induced proteolytic substrates, including 160 previously assigned caspase substrates. In addition to global profiling, a targeted approach using BAX HCT116 isogenic cell lines was utilized to dissect pre- and post-mitochondrial extrinsic apoptotic cleavage events. By employing apoptotic activation with a pro-apoptotic receptor agonist (PARA), a limited set of apoptotic substrates including known caspase substrates such as BH3 interacting-domain death agonist (BID) and Poly (ADP-ribose) polymerase (PARP)-1, and novel substrates such as Basic Transcription Factor 3, TRK-fused gene protein (TFG), and p62/Sequestosome were also identified.


Journal of Molecular Biology | 2015

Immunoaffinity Enrichment Coupled to Quantitative Mass Spectrometry Reveals Ubiquitin-Mediated Signaling Events

Kebing Yu; Lilian Phu; Eugene Varfolomeev; Daisy Bustos; Domagoj Vucic; Donald S. Kirkpatrick

Ubiquitination is one of the most prevalent posttranslational modifications in eukaryotic cells, with functional importance in protein degradation, subcellular localization and signal transduction pathways. Immunoaffinity enrichment coupled with quantitative mass spectrometry enables the in-depth characterization of protein ubiquitination events at the site-specific level. We have applied this strategy to investigate cellular response triggered by two distinct type agents: small molecule inhibitors of the tumor-associated kinases MEK and PI3K or the pro-inflammatory cytokine IL-17. Temporal profiling of protein ubiquitination events across a series of time points covering the biological response permits interrogation of signaling through thousands of quantified proteins, of which only a subset display significant and physiologically meaningful regulation. Distinctive clusters of residues within proteins can display distinct temporal patterns attributable to diverse molecular functions, although the majority of differential ubiquitination appears as a coordinated response across the modifiable residues present within an individual substrate. In cells treated with a combination of MEK and PI3K inhibitors, we found differential ubiquitination of MEK within the first hour after treatment and a series of mitochondria proteins at later time points. In the IL-17 signaling pathway, ubiquitination events on several signaling proteins including HOIL-1 and Tollip were observed. The functional relevance of these putative IL-17 mediators was subsequently validated by knockdown of HOIL-1, HOIP and TOLIP, each of which decreased IL-17-stimulated cytokine production. Together, these data validate proteomic profiling of protein ubiquitination as a viable approach for identifying dynamic signaling components in response to intracellular and extracellular perturbations.

Collaboration


Dive into the Daisy Bustos's collaboration.

Researchain Logo
Decentralizing Knowledge