95 results on '"Nicholas T. Woods"'
Search Results
2. Ctdp1 deficiency leads to early embryonic lethality in mice and defects in cell cycle progression in MEFs
- Author
-
Fangfang Qiao, Henry C.-H. Law, Kimiko L. Krieger, Emalie J. Clement, Yi Xiao, Shannon M. Buckley, and Nicholas T. Woods
- Subjects
ctdp1 ,knockout ,ccfdn ,embryonic lethality ,mefs ,cell death ,cell cycle arrest ,Science ,Biology (General) ,QH301-705.5 - Abstract
RNA polymerase II subunit A Carboxy-Terminal Domain Phosphatase 1 (CTDP1), a member of the haloacid dehalogenase superfamily phosphatases, has a defined role in transcriptional regulation, but emerging evidence suggests an expanded functional repertoire in the cell cycle and DNA damage response. In humans, a splice site mutation in CTDP1 gives rise to the rare Congenital Cataracts Facial Dysmorphism and Neuropathy syndrome, and recent evidence from our lab indicates CTDP1 is required for breast cancer growth and proliferation. To explore the physiological function of CTDP1 in a mammalian system, we generated a conditional Ctdp1 knockout mouse model by insertion of loxP sites upstream of exon 3 and downstream of exon 4. Biallelic deletion of Ctdp1 results in lethality before embryonic day 7.5, with morphological features indicating embryo cell death and resorption. However, Ctdp1+/− mice are haplosufficient for phenotypic traits including body weight, hematological parameters, exploratory and locomotive functions. To investigate the potential mechanisms of the embryonic death caused by biallelic Ctdp1 knockout, mouse embryonic fibroblasts (MEFs) were established from Ctdp1+/+ and Ctdp1flox/flox mice. Lentivirus delivered Cre-mediated biallelic deletion of Ctdp1 in MEFs results in cell death preceded by impaired proliferation characterized by an increase in G1- and G2-phase populations and a reduction in the S-phase population. These cell cycle alterations caused by deletion of Ctdp1 are associated with an increase in p27 protein expression and a decrease in phosphorylated RB, phosphorylated Histone H3, and Cyclin B expression. Together, these results reveal that Ctdp1 plays an essential role in early mouse embryo development and cell growth and survival in part by regulating the cell cycle.
- Published
- 2021
- Full Text
- View/download PDF
3. Lipid droplet membrane proteome remodeling parallels ethanol-induced hepatic steatosis and its resolution
- Author
-
Carol A. Casey, Terrence M. Donohue, Jr., Jacy L. Kubik, Vikas Kumar, Michael J. Naldrett, Nicholas T. Woods, Cole P. Frisbie, Mark A. McNiven, and Paul G. Thomes
- Subjects
Ethanol ,steatosis ,lipid droplet ,fasting ,proteomics ,liver ,Biochemistry ,QD415-436 - Abstract
Abstract: Lipid droplets (LDs) are composed of neutral lipids enclosed in a phospholipid monolayer, which harbors membrane-associated proteins that regulate LD functions. Despite the crucial role of LDs in lipid metabolism, remodeling of LD protein composition in disease contexts, such as steatosis, remains poorly understood. We hypothesized that chronic ethanol consumption, subsequent abstinence from ethanol, or fasting differentially affects the LD membrane proteome content and that these changes influence how LDs interact with other intracellular organelles. Here, male Wistar rats were pair-fed liquid control or ethanol diets for 6 weeks, and then, randomly chosen animals from both groups were either refed a control diet for 7 days or fasted for 48 h before euthanizing. From all groups, LD membrane proteins from purified liver LDs were analyzed immunochemically and by MS proteomics. Liver LD numbers and sizes were greater in ethanol-fed rats than in pair-fed control, 7-day refed, or fasted rats. Compared with control rats, ethanol feeding markedly altered the LD membrane proteome, enriching LD structural perilipins and proteins involved in lipid biosynthesis, while lowering LD lipase levels. Ethanol feeding also lowered LD-associated mitochondrial and lysosomal proteins. In 7-day refed (i.e., ethanol-abstained) or fasted-ethanol-fed rats, we detected distinct remodeling of the LD proteome, as judged by lower levels of lipid biosynthetic proteins, and enhanced LD interaction with mitochondria and lysosomes. Our study reveals evidence of significant remodeling of the LD membrane proteome that regulates ethanol-induced steatosis, its resolution after withdrawal and abstinence, and changes in LD interactions with other intracellular organelles.
- Published
- 2021
- Full Text
- View/download PDF
4. Functional requirements for a Samd14-capping protein complex in stress erythropoiesis
- Author
-
Suhita Ray, Linda Chee, Yichao Zhou, Meg A Schaefer, Michael J Naldrett, Sophie Alvarez, Nicholas T Woods, and Kyle J Hewitt
- Subjects
Kit ,stress erythropoiesis ,erythropoietin ,Samd14 ,capping protein ,Medicine ,Science ,Biology (General) ,QH301-705.5 - Abstract
Acute anemia induces rapid expansion of erythroid precursors and accelerated differentiation to replenish erythrocytes. Paracrine signals—involving cooperation between stem cell factor (SCF)/Kit signaling and other signaling inputs—are required for the increased erythroid precursor activity in anemia. Our prior work revealed that the sterile alpha motif (SAM) domain 14 (Samd14) gene increases the regenerative capacity of the erythroid system in a mouse genetic model and promotes stress-dependent Kit signaling. However, the mechanism underlying Samd14’s role in stress erythropoiesis is unknown. We identified a protein-protein interaction between Samd14 and the α- and β-heterodimers of the F-actin capping protein (CP) complex. Knockdown of the CP β subunit increased erythroid maturation in murine ex vivo cultures and decreased colony forming potential of stress erythroid precursors. In a genetic complementation assay for Samd14 activity, our results revealed that the Samd14-CP interaction is a determinant of erythroid precursor cell levels and function. Samd14-CP promotes SCF/Kit signaling in CD71med spleen erythroid precursors. Given the roles of Kit signaling in hematopoiesis and Samd14 in Kit pathway activation, this mechanism may have pathological implications in acute/chronic anemia.
- Published
- 2022
- Full Text
- View/download PDF
5. Supplementary Figure S8 from PAXIP1 Potentiates the Combination of WEE1 Inhibitor AZD1775 and Platinum Agents in Lung Cancer
- Author
-
Alvaro N. Monteiro, Uwe Rix, Eric B. Haura, Jhanelle E. Gray, Stephen G. Brantley, John M. Koomen, W. Douglas Cress, Elizabeth Remily-Wood, Jodi L. Kroeger, Fumi Kinose, Lily Rix, Gabriela Wright, Nicholas T. Woods, and Ankita Jhuraney
- Abstract
(A) TMA containing 68 patient-derived xenograft (PDX) tumor models stained with PAXIP1 and WEE1 (B-C) Two lung adenocarcinoma PDXs with PAXIP1 and WEE1 staining were used to perform ex vivo 3-D clonogenic assays with various concentrations of AZD1775 and cisplatin and tumor/control - T/C was measured by image analysis.
- Published
- 2023
- Full Text
- View/download PDF
6. Supplementary Table S1 from PAXIP1 Potentiates the Combination of WEE1 Inhibitor AZD1775 and Platinum Agents in Lung Cancer
- Author
-
Alvaro N. Monteiro, Uwe Rix, Eric B. Haura, Jhanelle E. Gray, Stephen G. Brantley, John M. Koomen, W. Douglas Cress, Elizabeth Remily-Wood, Jodi L. Kroeger, Fumi Kinose, Lily Rix, Gabriela Wright, Nicholas T. Woods, and Ankita Jhuraney
- Abstract
Peptides used for LC-MRM based protein quantification for each kinase
- Published
- 2023
- Full Text
- View/download PDF
7. Supplementary Materials and Methods from PAXIP1 Potentiates the Combination of WEE1 Inhibitor AZD1775 and Platinum Agents in Lung Cancer
- Author
-
Alvaro N. Monteiro, Uwe Rix, Eric B. Haura, Jhanelle E. Gray, Stephen G. Brantley, John M. Koomen, W. Douglas Cress, Elizabeth Remily-Wood, Jodi L. Kroeger, Fumi Kinose, Lily Rix, Gabriela Wright, Nicholas T. Woods, and Ankita Jhuraney
- Abstract
Supplementary Materials and Methods
- Published
- 2023
- Full Text
- View/download PDF
8. Supp Materials legends from PAXIP1 Potentiates the Combination of WEE1 Inhibitor AZD1775 and Platinum Agents in Lung Cancer
- Author
-
Alvaro N. Monteiro, Uwe Rix, Eric B. Haura, Jhanelle E. Gray, Stephen G. Brantley, John M. Koomen, W. Douglas Cress, Elizabeth Remily-Wood, Jodi L. Kroeger, Fumi Kinose, Lily Rix, Gabriela Wright, Nicholas T. Woods, and Ankita Jhuraney
- Abstract
Legends for Supplementary Figures and Tables
- Published
- 2023
- Full Text
- View/download PDF
9. Supplementary Table 5 from Functional Analysis and Fine Mapping of the 9p22.2 Ovarian Cancer Susceptibility Locus
- Author
-
Alvaro N.A. Monteiro, Simon A. Gayther, Paul D. Pharoah, Ellen L. Goode, Thomas A. Sellers, Timothy R. Hughes, Houtan Noushmehr, Gerhard A. Coetzee, Catherine M. Phelan, Susan J. Ramus, Georgia Chenevix-Trench, Harvey Risch, Sean J. Yoder, Madalene A. Earp, Ally W. Yang, Howard C. Shen, Simon G. Coetzee, Paulo C. Lyra, Renato S. Carvalho, Anxhela Gjyshi, Hamed S. Najafabadi, Dennis J. Hazelett, Kate Lawrenson, Gustavo Mendoza-Fandiño, Jonathan P. Tyrer, Nicholas T. Woods, and Melissa A. Buckley
- Abstract
eQTL Associations from GTEx (Release V7 (dbGaP Accession phs000424.v7.p2). Contains single gene eQTLs for BNC2, CNTLN, c9orf92 and SH3GL2 in all tissues and single SNP eQTL for all 40 SNPs in the credible candidate set.
- Published
- 2023
- Full Text
- View/download PDF
10. Tables S5-S13 from The Proteomic Landscape of Pancreatic Ductal Adenocarcinoma Liver Metastases Identifies Molecular Subtypes and Associations with Clinical Response
- Author
-
Nicholas T. Woods, Michael A. Hollingsworth, Fang Yu, Kurt W. Fisher, Leah M. Cook, Paul M. Grandgenett, Dominick J. DiMaio, Jean L. Grem, Thomas C. Caffrey, Diane Costanzo-Garvey, Kimiko L. Krieger, Zachary S. Wagner, Fangfang Qiao, Emalie J. Clement, Dragana Lagundžin, and Henry C.-H. Law
- Abstract
Table S5: The protein expression matrix used for input in the Principle Component Analysis. Table S6: The list of gene ontology terms in each cluster observed in the SAFE network. Table S7: The regression coefficients and constants of each protein in each proteomics subtype in the Partial Least Squares-Discriminant Analysis. Table S8: The gene ontology analysis of Protein Cluster 1 from ClueGO. Table S9: The gene ontology analysis of Protein Cluster 2 from ClueGO. Table S10: The gene ontology analysis of Protein Cluster 3 from ClueGO. Table S11: The Reactome pathway enrichment analysis on the average protein expression of the 4 proteomics subtypes. Table S12: The tertile assignments of each of the 916 proteins used for the analysis in each 56 samples. Table S13: The comparative analysis of the gemcitabine-treated proteome.
- Published
- 2023
- Full Text
- View/download PDF
11. Data from The Proteomic Landscape of Pancreatic Ductal Adenocarcinoma Liver Metastases Identifies Molecular Subtypes and Associations with Clinical Response
- Author
-
Nicholas T. Woods, Michael A. Hollingsworth, Fang Yu, Kurt W. Fisher, Leah M. Cook, Paul M. Grandgenett, Dominick J. DiMaio, Jean L. Grem, Thomas C. Caffrey, Diane Costanzo-Garvey, Kimiko L. Krieger, Zachary S. Wagner, Fangfang Qiao, Emalie J. Clement, Dragana Lagundžin, and Henry C.-H. Law
- Abstract
Purpose:Pancreatic ductal adenocarcinoma (PDAC) is a highly metastatic disease that can be separated into distinct subtypes based on molecular signatures. Identifying PDAC subtype-specific therapeutic vulnerabilities is necessary to develop precision medicine approaches to treat PDAC.Experimental Design:A total of 56 PDAC liver metastases were obtained from the UNMC Rapid Autopsy Program and analyzed with quantitative proteomics. PDAC subtypes were identified by principal component analysis based on protein expression profiling. Proteomic subtypes were further characterized by the associated clinical information, including but not limited to survival analysis, drug treatment response, and smoking and drinking status.Results:Over 3,960 proteins were identified and used to delineate four distinct PDAC microenvironment subtypes: (i) metabolic; (ii) progenitor-like; (iii) proliferative; and (iv) inflammatory. PDAC risk factors of alcohol and tobacco consumption correlate with subtype classifications. Enhanced survival is observed in FOLFIRINOX treated metabolic and progenitor-like subtypes compared with the proliferative and inflammatory subtypes. In addition, TYMP, PDCD6IP, ERAP1, and STMN showed significant association with patient survival in a subtype-specific manner. Gemcitabine-induced alterations in the proteome identify proteins, such as serine hydroxymethyltransferase 1, associated with drug resistance.Conclusions:These data demonstrate that proteomic analysis of clinical PDAC liver metastases can identify molecular signatures unique to disease subtypes and point to opportunities for therapeutic development to improve the treatment of PDAC.
- Published
- 2023
- Full Text
- View/download PDF
12. Supplementary Figures 1-4 and extended methods from Functional Analysis and Fine Mapping of the 9p22.2 Ovarian Cancer Susceptibility Locus
- Author
-
Alvaro N.A. Monteiro, Simon A. Gayther, Paul D. Pharoah, Ellen L. Goode, Thomas A. Sellers, Timothy R. Hughes, Houtan Noushmehr, Gerhard A. Coetzee, Catherine M. Phelan, Susan J. Ramus, Georgia Chenevix-Trench, Harvey Risch, Sean J. Yoder, Madalene A. Earp, Ally W. Yang, Howard C. Shen, Simon G. Coetzee, Paulo C. Lyra, Renato S. Carvalho, Anxhela Gjyshi, Hamed S. Najafabadi, Dennis J. Hazelett, Kate Lawrenson, Gustavo Mendoza-Fandiño, Jonathan P. Tyrer, Nicholas T. Woods, and Melissa A. Buckley
- Abstract
This file contains: Supplementary Figure 1 (Related to Figure 2): Haplotype frequency for tiles Enhancer Scanning times with multiple SNPs. Supplementary Figure 2 (Related to Figure 3): Conservation and S/MAR predicted sequences at the locus. Supplementary Figure 3: Study overview and Overlap between the set of 134 SNPs obtained through the LD-guided analysis and the set of 40 SNPs obtained through the fine mapping-guided approach. Supplementary Figure 4 (Related to Figure 5): BNC2 binds to its own promoter. Extended data and detailed methods (including QC metrics) for BNC2 ChIP-Seq.
- Published
- 2023
- Full Text
- View/download PDF
13. Figures S6-S10 from The Proteomic Landscape of Pancreatic Ductal Adenocarcinoma Liver Metastases Identifies Molecular Subtypes and Associations with Clinical Response
- Author
-
Nicholas T. Woods, Michael A. Hollingsworth, Fang Yu, Kurt W. Fisher, Leah M. Cook, Paul M. Grandgenett, Dominick J. DiMaio, Jean L. Grem, Thomas C. Caffrey, Diane Costanzo-Garvey, Kimiko L. Krieger, Zachary S. Wagner, Fangfang Qiao, Emalie J. Clement, Dragana Lagundžin, and Henry C.-H. Law
- Abstract
Figure S6: Protein expression and gene ontology analysis related to PDAC subtypes. Figure S7: The characterization of Protein Cluster 1. Figure S8: The characterization of Protein Cluster 2. Figure S9: The characterization of Protein Cluster 3. Figure S10: The Kaplan-Meier curves of patients separated based on individual PDAC liver metastasis subtypes.
- Published
- 2023
- Full Text
- View/download PDF
14. Supplementary Table 2 from Functional Analysis and Fine Mapping of the 9p22.2 Ovarian Cancer Susceptibility Locus
- Author
-
Alvaro N.A. Monteiro, Simon A. Gayther, Paul D. Pharoah, Ellen L. Goode, Thomas A. Sellers, Timothy R. Hughes, Houtan Noushmehr, Gerhard A. Coetzee, Catherine M. Phelan, Susan J. Ramus, Georgia Chenevix-Trench, Harvey Risch, Sean J. Yoder, Madalene A. Earp, Ally W. Yang, Howard C. Shen, Simon G. Coetzee, Paulo C. Lyra, Renato S. Carvalho, Anxhela Gjyshi, Hamed S. Najafabadi, Dennis J. Hazelett, Kate Lawrenson, Gustavo Mendoza-Fandiño, Jonathan P. Tyrer, Nicholas T. Woods, and Melissa A. Buckley
- Abstract
Genes selected for NanoString analysis. Contains the following tabs: (1) Nanostring gene set: final list for NanoString analysis. Gene Symbol, genes selected for nanostring analysis; Pathway, annotation from literature or from pathways in Tab 10; Cell line, cell line (FTE, iFTSEC283; OSE, iOSE11) in which the BNC2 ChIP-Seq peak was identified in proximity to the TSS. P-value, significance of expression changes using a nominal p-value {less than or equal to} 0.05 (indicated in green highlight); (2) 995 gene list: list of 995 genes/transcripts with TSS within 30 kb of the BNC2 ChIP-Seq peak centers found in both iOSE11 and iFTSEC283 cells; (3)-(9) Coordinates and annotation of BNC2 ChIP-seq peaks and genes with TSS within 30 kb of the BNC2 ChIP-seq peak centers from two replicates each of FTE (iFTSEC283) and OSE (iOSE11) cells, their overlaps; (10) Panther GO analysis results.
- Published
- 2023
- Full Text
- View/download PDF
15. Data from Functional Analysis and Fine Mapping of the 9p22.2 Ovarian Cancer Susceptibility Locus
- Author
-
Alvaro N.A. Monteiro, Simon A. Gayther, Paul D. Pharoah, Ellen L. Goode, Thomas A. Sellers, Timothy R. Hughes, Houtan Noushmehr, Gerhard A. Coetzee, Catherine M. Phelan, Susan J. Ramus, Georgia Chenevix-Trench, Harvey Risch, Sean J. Yoder, Madalene A. Earp, Ally W. Yang, Howard C. Shen, Simon G. Coetzee, Paulo C. Lyra, Renato S. Carvalho, Anxhela Gjyshi, Hamed S. Najafabadi, Dennis J. Hazelett, Kate Lawrenson, Gustavo Mendoza-Fandiño, Jonathan P. Tyrer, Nicholas T. Woods, and Melissa A. Buckley
- Abstract
Genome-wide association studies have identified 40 ovarian cancer risk loci. However, the mechanisms underlying these associations remain elusive. In this study, we conducted a two-pronged approach to identify candidate causal SNPs and assess underlying biological mechanisms at chromosome 9p22.2, the first and most statistically significant associated locus for ovarian cancer susceptibility. Three transcriptional regulatory elements with allele-specific effects and a scaffold/matrix attachment region were characterized and, through physical DNA interactions, BNC2 was established as the most likely target gene. We determined the consensus binding sequence for BNC2 in vitro, verified its enrichment in BNC2 ChIP-seq regions, and validated a set of its downstream target genes. Fine-mapping by dense regional genotyping in over 15,000 ovarian cancer cases and 30,000 controls identified SNPs in the scaffold/matrix attachment region as among the most likely causal variants. This study reveals a comprehensive regulatory landscape at 9p22.2 and proposes a likely mechanism of susceptibility to ovarian cancer.Significance:Mapping the 9p22.2 ovarian cancer risk locus identifies BNC2 as an ovarian cancer risk gene.See related commentary by Choi and Brown, p. 439
- Published
- 2023
- Full Text
- View/download PDF
16. Supplementary Table 1 from Functional Analysis and Fine Mapping of the 9p22.2 Ovarian Cancer Susceptibility Locus
- Author
-
Alvaro N.A. Monteiro, Simon A. Gayther, Paul D. Pharoah, Ellen L. Goode, Thomas A. Sellers, Timothy R. Hughes, Houtan Noushmehr, Gerhard A. Coetzee, Catherine M. Phelan, Susan J. Ramus, Georgia Chenevix-Trench, Harvey Risch, Sean J. Yoder, Madalene A. Earp, Ally W. Yang, Howard C. Shen, Simon G. Coetzee, Paulo C. Lyra, Renato S. Carvalho, Anxhela Gjyshi, Hamed S. Najafabadi, Dennis J. Hazelett, Kate Lawrenson, Gustavo Mendoza-Fandiño, Jonathan P. Tyrer, Nicholas T. Woods, and Melissa A. Buckley
- Abstract
Primers used in this study.
- Published
- 2023
- Full Text
- View/download PDF
17. Supplementary Table 4 from Functional Analysis and Fine Mapping of the 9p22.2 Ovarian Cancer Susceptibility Locus
- Author
-
Alvaro N.A. Monteiro, Simon A. Gayther, Paul D. Pharoah, Ellen L. Goode, Thomas A. Sellers, Timothy R. Hughes, Houtan Noushmehr, Gerhard A. Coetzee, Catherine M. Phelan, Susan J. Ramus, Georgia Chenevix-Trench, Harvey Risch, Sean J. Yoder, Madalene A. Earp, Ally W. Yang, Howard C. Shen, Simon G. Coetzee, Paulo C. Lyra, Renato S. Carvalho, Anxhela Gjyshi, Hamed S. Najafabadi, Dennis J. Hazelett, Kate Lawrenson, Gustavo Mendoza-Fandiño, Jonathan P. Tyrer, Nicholas T. Woods, and Melissa A. Buckley
- Abstract
Fine mapping association results. Contains univariate and conditional (conditioning on rs3814113) analysis.
- Published
- 2023
- Full Text
- View/download PDF
18. Supplementary Table 3 from Functional Analysis and Fine Mapping of the 9p22.2 Ovarian Cancer Susceptibility Locus
- Author
-
Alvaro N.A. Monteiro, Simon A. Gayther, Paul D. Pharoah, Ellen L. Goode, Thomas A. Sellers, Timothy R. Hughes, Houtan Noushmehr, Gerhard A. Coetzee, Catherine M. Phelan, Susan J. Ramus, Georgia Chenevix-Trench, Harvey Risch, Sean J. Yoder, Madalene A. Earp, Ally W. Yang, Howard C. Shen, Simon G. Coetzee, Paulo C. Lyra, Renato S. Carvalho, Anxhela Gjyshi, Hamed S. Najafabadi, Dennis J. Hazelett, Kate Lawrenson, Gustavo Mendoza-Fandiño, Jonathan P. Tyrer, Nicholas T. Woods, and Melissa A. Buckley
- Abstract
List of 134 SNPs in linkage disequilibrium (r2 {greater than or equal to} 0.3) with rs3814113 (based on 1000GP v3), selected for functional analysis.
- Published
- 2023
- Full Text
- View/download PDF
19. Supplementary Data from The Proteomic Landscape of Pancreatic Ductal Adenocarcinoma Liver Metastases Identifies Molecular Subtypes and Associations with Clinical Response
- Author
-
Nicholas T. Woods, Michael A. Hollingsworth, Fang Yu, Kurt W. Fisher, Leah M. Cook, Paul M. Grandgenett, Dominick J. DiMaio, Jean L. Grem, Thomas C. Caffrey, Diane Costanzo-Garvey, Kimiko L. Krieger, Zachary S. Wagner, Fangfang Qiao, Emalie J. Clement, Dragana Lagundžin, and Henry C.-H. Law
- Abstract
Text file containing supplementary information.
- Published
- 2023
- Full Text
- View/download PDF
20. Table S4 from The Proteomic Landscape of Pancreatic Ductal Adenocarcinoma Liver Metastases Identifies Molecular Subtypes and Associations with Clinical Response
- Author
-
Nicholas T. Woods, Michael A. Hollingsworth, Fang Yu, Kurt W. Fisher, Leah M. Cook, Paul M. Grandgenett, Dominick J. DiMaio, Jean L. Grem, Thomas C. Caffrey, Diane Costanzo-Garvey, Kimiko L. Krieger, Zachary S. Wagner, Fangfang Qiao, Emalie J. Clement, Dragana Lagundžin, and Henry C.-H. Law
- Abstract
Table S4: The total list of peptides identified.
- Published
- 2023
- Full Text
- View/download PDF
21. Supplementary Methods from Anoikis, Initiated by Mcl-1 Degradation and Bim Induction, Is Deregulated during Oncogenesis
- Author
-
Hong-Gang Wang, Kapil N. Bhalla, Francis Y. Lee, Hirohito Yamaguchi, and Nicholas T. Woods
- Abstract
Supplementary Methods from Anoikis, Initiated by Mcl-1 Degradation and Bim Induction, Is Deregulated during Oncogenesis
- Published
- 2023
- Full Text
- View/download PDF
22. Supplementary Figure 3 from Anoikis, Initiated by Mcl-1 Degradation and Bim Induction, Is Deregulated during Oncogenesis
- Author
-
Hong-Gang Wang, Kapil N. Bhalla, Francis Y. Lee, Hirohito Yamaguchi, and Nicholas T. Woods
- Abstract
Supplementary Figure 3 from Anoikis, Initiated by Mcl-1 Degradation and Bim Induction, Is Deregulated during Oncogenesis
- Published
- 2023
- Full Text
- View/download PDF
23. Data from Anoikis, Initiated by Mcl-1 Degradation and Bim Induction, Is Deregulated during Oncogenesis
- Author
-
Hong-Gang Wang, Kapil N. Bhalla, Francis Y. Lee, Hirohito Yamaguchi, and Nicholas T. Woods
- Abstract
Anoikis, a Bax-dependent apoptosis triggered by detachment from the extracellular matrix, is often dysfunctional in metastatic cancer cells. Using wild-type and c-Src–transformed NIH3T3 cells as a model, we identified Mcl-1 degradation and Bim up-regulation as a critical determinant of anoikis initiation. Detachment rapidly degraded Mcl-1 via a GSK-3β–dependent proteasomal pathway and transcriptionally up-regulated Bim expression. Mcl-1 degradation in the presence of Bim was sufficient to induce anoikis. By analyzing nonmetastatic Saos-2 and metastatic derivative LM7 cells, we confirmed that dysregulation of Mcl-1 degradation and Bim induction during detachment contributes to decreased anoikis sensitivity of metastatic cells. Furthermore, knockdown of Mcl-1 or pharmacologic inhibition of the phosphoinositide-3-kinase/Akt and mitogen-activated protein kinase pathways that suppress Mcl-1 degradation and Bim expression could markedly sensitize metastatic breast cancer cells to anoikis and prevent metastases in vivo. Therefore, Mcl-1 degradation primes the cell for Bax activation and anoikis, which can be blocked by oncogenic signaling in metastatic cells. [Cancer Res 2007;67(22):10744–52]
- Published
- 2023
- Full Text
- View/download PDF
24. Supplementary Figure 2 from Anoikis, Initiated by Mcl-1 Degradation and Bim Induction, Is Deregulated during Oncogenesis
- Author
-
Hong-Gang Wang, Kapil N. Bhalla, Francis Y. Lee, Hirohito Yamaguchi, and Nicholas T. Woods
- Abstract
Supplementary Figure 2 from Anoikis, Initiated by Mcl-1 Degradation and Bim Induction, Is Deregulated during Oncogenesis
- Published
- 2023
- Full Text
- View/download PDF
25. Supplementary Figure Legends 1-4 from Anoikis, Initiated by Mcl-1 Degradation and Bim Induction, Is Deregulated during Oncogenesis
- Author
-
Hong-Gang Wang, Kapil N. Bhalla, Francis Y. Lee, Hirohito Yamaguchi, and Nicholas T. Woods
- Abstract
Supplementary Figure Legends 1-4 from Anoikis, Initiated by Mcl-1 Degradation and Bim Induction, Is Deregulated during Oncogenesis
- Published
- 2023
- Full Text
- View/download PDF
26. Supplementary Figure 4 from Anoikis, Initiated by Mcl-1 Degradation and Bim Induction, Is Deregulated during Oncogenesis
- Author
-
Hong-Gang Wang, Kapil N. Bhalla, Francis Y. Lee, Hirohito Yamaguchi, and Nicholas T. Woods
- Abstract
Supplementary Figure 4 from Anoikis, Initiated by Mcl-1 Degradation and Bim Induction, Is Deregulated during Oncogenesis
- Published
- 2023
- Full Text
- View/download PDF
27. Supplementary Figure 1 from Anoikis, Initiated by Mcl-1 Degradation and Bim Induction, Is Deregulated during Oncogenesis
- Author
-
Hong-Gang Wang, Kapil N. Bhalla, Francis Y. Lee, Hirohito Yamaguchi, and Nicholas T. Woods
- Abstract
Supplementary Figure 1 from Anoikis, Initiated by Mcl-1 Degradation and Bim Induction, Is Deregulated during Oncogenesis
- Published
- 2023
- Full Text
- View/download PDF
28. Stapling proteins in the RELA complex inhibits TNFα-induced nuclear translocation of RELA
- Author
-
Tom Huxford, Dragana Lagundžin, Nicholas T. Woods, Sandeep Rana, Smit Kour, Amarnath Natarajan, Smitha Kizhake, David Klinkebiel, and Jayapal Reddy Mallareddy
- Subjects
Chemistry ,chemistry.chemical_compound ,Chemistry (miscellaneous) ,Protein subunit ,Dimer ,Tumor necrosis factor alpha ,Biochemistry, Genetics and Molecular Biology (miscellaneous) ,Molecular Biology ,Biochemistry ,Nuclear translocation ,Cell biology - Abstract
Tumor necrosis factor (TNF) α-induced nuclear translocation of the NF-κB subunit RELA has been implicated in several pathological conditions. Here we report the discovery of a spirocyclic dimer (SpiD7) that covalently modifies RELA to inhibit TNFα-induced nuclear translocation. This is a previously unexplored strategy to inhibit TNFα-induced NF-κB activation., Discovery of a spirocyclic dimer (SpiD7) that covalently modifies RELA to generate stable high molecular weight complexes. SpiD7 inhibits TNFα-induced nuclear translocation of RELA resulting in the blockade of NF-kB gene transcription, through a previously unexplored modality.
- Published
- 2022
- Full Text
- View/download PDF
29. Two distinct mechanisms underlie estrogen-receptor-negative breast cancer susceptibility at the 2p23.2 locus
- Author
-
Nicholas T. Woods, Paulo C Lyra, Alvaro N.A. Monteiro, Marcelo A. Carvalho, Carly M. Harro, Thales C. Nepomuceno, Fergus J. Couch, Leticia Batista Azevedo Rangel, Gustavo Mendoza-Fandiño, and Xueli Li
- Subjects
Untranslated region ,Genetics ,Breast Neoplasms ,Estrogens ,Triple Negative Breast Neoplasms ,Genome-wide association study ,Locus (genetics) ,Single-nucleotide polymorphism ,Biology ,Polymorphism, Single Nucleotide ,Article ,Regulatory sequence ,Humans ,Female ,Genetic Predisposition to Disease ,Ectopic expression ,Transcription factor ,Gene ,Genetics (clinical) ,Genome-Wide Association Study - Abstract
Genome wide-association studies (GWAS) have established over 400 breast cancer risk loci defined by common single nucleotide polymorphisms (SNPs), including several associated with estrogen-receptor (ER)-negative disease. Most of these loci have not been studied systematically and the mechanistic underpinnings of risk are largely unknown. Here we explored the landscape of genomic features at an ER-negative breast cancer susceptibility locus at chromosome 2p23.2 and assessed the functionality of 81 SNPs with strong evidence of association from previous fine mapping. Five candidate regulatory regions containing risk-associated SNPs were identified. Regulatory Region 1 in the first intron of WDR43 contains SNP rs4407214, which showed allele-specific interaction with the transcription factor USF1 in in vitro assays. CRISPR-mediated disruption of Regulatory Region 1 led to expression changes in the neighboring PLB1 gene, suggesting that the region acts as a distal enhancer. Regulatory Regions 2, 4, and 5 did not provide sufficient evidence for functionality in in silico and experimental analyses. Two SNPs (rs11680458 and rs1131880) in Regulatory Region 3, mapping to the seed region for miRNA-recognition sites in the 3' untranslated region of WDR43, showed allele-specific effects of ectopic expression of miR-376 on WDR43 expression levels. Taken together, our data suggest that risk of ER-negative breast cancer associated with the 2p23.2 locus is likely driven by a combinatorial effect on the regulation of WDR43 and PLB1.
- Published
- 2021
- Full Text
- View/download PDF
30. Abstract 3504: Evaluating the molecular determinants of PDAC racial health disparities
- Author
-
Anthony E. Johansen-Sallee, Alexa M. Barber, Henry Law, Jose Trevino, and Nicholas T. Woods
- Subjects
Cancer Research ,Oncology - Abstract
Pancreatic cancer is the 3rd leading cause of cancer-related deaths with a 5-year survival rate of just 11.5%. Pancreatic ductal adenocarcinoma (PDAC), accounting for over 90% of pancreatic malignancies, has the highest incidence rate, mortality rate, and shortest survival times in non-Hispanic black (defined here as African and/or African American ancestry) patients compared to other races. Due to the racial health disparities of PDAC, this study seeks to quantify the proteomic signatures associated with tumor racial origin to identify the underlying molecular pathways that could contribute to these disparities. Using mass spectrometry on primary PDAC tumor samples collected from 30 Caucasian and 12 African American patients, 183 proteins were differentially expressed between these groups. The most over-expressed protein in African American tumors was Ring Finger Protein 2 (RNF2) with a log2 fold-change greater than six. RNF2 had previously been found to regulate GATA Binding Protein 6 (GATA6) in embryonic stem cells. Additionally, GATA6 is also an important determinant in PDAC subtyping with higher GATA6 expression leading to greater chemosensitivity. To expand upon these findings, chromatin immunoprecipitation and sequencing (ChIP-seq) was performed on MIA-PaCa2 to identify binding sites for RNF2 in PDAC cells. Peaks were called with MACS2 (q Citation Format: Anthony E. Johansen-Sallee, Alexa M. Barber, Henry Law, Jose Trevino, Nicholas T. Woods. Evaluating the molecular determinants of PDAC racial health disparities [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2023; Part 1 (Regular and Invited Abstracts); 2023 Apr 14-19; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2023;83(7_Suppl):Abstract nr 3504.
- Published
- 2023
- Full Text
- View/download PDF
31. Abstract PS18-49: A transcriptome approach to reveal CTDP1 knockdown induced G1 cell cycle arrest in triple negative breast cancer cells
- Author
-
Nicholas T. Woods and Henry Chun Hin Law
- Subjects
Transcriptome ,Cancer Research ,Gene knockdown ,Oncology ,DNA repair ,Transcriptional regulation ,E2F1 ,Biology ,Mitosis ,G1 phase ,Transcription factor ,Cell biology - Abstract
CTDP1 (C-terminal domain phosphatase 1) is the only phosphatase domain with a BRCT-domain and known to be associated with transcription and mitosis in eukaryotic systems. Our previous research showed that CTDP1 participates in the DNA damage response by mediating interstrand DNA repair through the Fanconi anemia pathway. This study also revealed that CTDP1 is an essential gene in breast cancer cell lines but not in normal breast derived epithelial MCF10A cells. This project aims to identify the changes in transcripts, kinases and interactors leading to the cell death induced by CTDP1 knockdown in the triple-negative breast cancer cell lines. Since RNA Pol II is one of the major targets of CTDP1 phosphatase activity, we initially hypothesized that the cellular changes were mediated by transcriptional regulation. The global transcriptomic profiling of shCTDP1 MDA-MB-231 cells detected more than 29000 gene products, where 1000 and 616 genes were found consistently up- and down-regulated, respectively. The downregulation of cell cycle-associated genes CCNA2, TOP2A and POLA1 in the MDA-MB-231 cells were confirmed with q-RT-PCR. While the upregulated genes did not show enrichment in the Reactome pathway database, the downregulated genes were significantly associated with DNA damage response, mitosis and transcription, which is consistent with the known functions of CTDP1. The transcription factor binding motifs analysis with iRegulon reveals that the E2F1 transcription activity decreased as CTDP1 was knocked down in MDA-MB-231. The subsequent cell cycle analysis supports this prediction and showed G1 arrest as CTDP1 is knocked down in MDA-MB-231 and MDA-MB-453, but not in MCF10A. A kinase array analysis was also conducted to further identify kinases activated or deactivated as CTDP1 is knocked down. A nested network analysis of the predicted transcription factors, kinases and CTDP1 interactors is used to identify potential pathways contributing to cell death of the triple-negative breast cancer cells upon loss of CTDP1 expression. Citation Format: Henry Chun Hin Law, Nicholas Woods. A transcriptome approach to reveal CTDP1 knockdown induced G1 cell cycle arrest in triple negative breast cancer cells [abstract]. In: Proceedings of the 2020 San Antonio Breast Cancer Virtual Symposium; 2020 Dec 8-11; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2021;81(4 Suppl):Abstract nr PS18-49.
- Published
- 2021
- Full Text
- View/download PDF
32. Small Molecule Activators of B56-PP2A Restore 4E-BP Expression and Function to Suppress Cap-dependent Translation in Cancer Cells
- Author
-
Michelle A. Lum, Kayla A. Jonas-Breckenridge, Adrian R. Black, Nicholas T. Woods, Caitlin O’Connor, Rita A. Avelar, Analisa DiFeo, Goutham Narla, and Jennifer D. Black
- Abstract
Dysregulation of cap-dependent translation is a hallmark of cancer, with key roles in supporting the transformed phenotype. The eIF4E binding proteins (4E-BP1, 2, 3) are major negative regulators of cap-dependent translation that are inactivated in tumors through inhibitory phosphorylation by oncogenic kinases (e.g., mTOR) or by downregulation. Previous studies from our group and others have linked tumor suppressive PP2A family serine/threonine phosphatases to activation of 4E-BP1. Here, we leveraged novel small molecule activators of PP2A (SMAPs) (e.g., DT-061, DT-1154) that are being developed as antitumor agents to (a) explore the role of a subset of B56-PP2As in regulation of 4E-BP activity, and (b) to evaluate the potential of B56-PP2A reactivation for restoring translation control in tumor cells. We show that SMAPs promote PP2A-dependent hypophosphorylation of 4E-BP1/4EBP2 in the presence of active upstream inhibitory kinases (mTOR, ERK, AKT), supporting a role for B56-PP2As as 4E-BP phosphatases. Unexpectedly, DT-061 also led to robust PP2A-dependent upregulation of 4E-BP1, but not 4E-BP2 or 4E-BP3. Cap-binding assays and eIF4E immunoprecipitation showed that SMAP/B56-PP2A blocks the formation of the eIF4F translation initiation complex. Bicistronic reporter assays that directly measure cap-dependent translation activity confirmed the translational consequences of these effects. siRNA knockdown pointed to B56α-PP2A as a mediator of SMAP effects on 4E-BPs, although B56β- and/or B56ε-PP2A may also play a role. 4E-BP1 upregulation involved ATF4-dependent transcription of the 4E-BP1 gene (EIF4EBP1) and the effect was partially dependent on TFE3/TFEB transcription factors. Thus, B56-PP2A orchestrates a translation repressive program involving transcriptional induction and hypophosphorylation of 4E-BP1, highlighting the potential of PP2A-based therapeutic strategies for restoration of translation control in cancer cells.
- Published
- 2022
- Full Text
- View/download PDF
33. Author response: Functional requirements for a Samd14-capping protein complex in stress erythropoiesis
- Author
-
Suhita Ray, Linda Chee, Yichao Zhou, Meg A Schaefer, Michael J Naldrett, Sophie Alvarez, Nicholas T Woods, and Kyle J Hewitt
- Published
- 2022
- Full Text
- View/download PDF
34. Combined Alcohol Exposure and KRAS Mutation in Human Pancreatic Ductal Epithelial Cells Induces Proliferation and Alters Subtype Signatures Determined by Multi-Omics Analysis
- Author
-
Emalie J. Clement, Henry C.-H. Law, Fangfang Qiao, Dragana Noe, Jose G. Trevino, and Nicholas T. Woods
- Subjects
Cancer Research ,Oncology ,endocrine system diseases ,alcohol ,pancreatic cancer ,Proteomics ,KRAS ,SERPINE1 ,neoplasms ,digestive system diseases - Abstract
Pancreatic Ductal adenocarcinoma (PDAC) is an aggressive cancer commonly exhibiting KRAS-activating mutations. Alcohol contributes to the risk of developing PDAC in humans, and murine models have shown alcohol consumption in the context of KRAS mutation in the pancreas promotes the development of PDAC. The molecular signatures in pancreas cells altered by alcohol exposure in the context of mutant KRAS could identify pathways related to the etiology of PDAC. In this study, we evaluated the combined effects of alcohol exposure and KRAS mutation status on the transcriptome and proteome of pancreatic HPNE cell models. These analyses identified alterations in transcription and translational processes in mutant KRAS cells exposed to alcohol. In addition, multi-omics analysis suggests an increase in the correlation between mRNA transcript and protein abundance in cells exposed to alcohol with an underlying KRAS mutation. Through differential co-expression, SERPINE1 was found to be influential for PDAC development in the context of mutant KRAS and ethanol. In terms of PDAC subtypes, alcohol conditioning of HPNE cells expressing mutant KRAS decreases the Inflammatory subtype signature and increases the Proliferative and Metabolic signatures, as we previously observed in patient samples. The alterations in molecular subtypes were associated with an increased sensitivity to chemotherapeutic agents gemcitabine, irinotecan, and oxaliplatin. These results provide a framework for distinguishing the molecular dysregulation associated with combined alcohol and mutant KRAS in a pancreatic cell line model.
- Published
- 2022
- Full Text
- View/download PDF
35. Functional requirements for a Samd14-capping protein complex in stress erythropoiesis
- Author
-
Suhita Ray, Linda Chee, Yichao Zhou, Meg A Schaefer, Michael J Naldrett, Sophie Alvarez, Nicholas T Woods, and Kyle J Hewitt
- Subjects
Erythroid Precursor Cells ,Mice ,Erythrocytes ,General Immunology and Microbiology ,General Neuroscience ,hemic and lymphatic diseases ,Animals ,Proteins ,Anemia ,Cell Differentiation ,Erythropoiesis ,General Medicine ,General Biochemistry, Genetics and Molecular Biology - Abstract
Acute anemia induces rapid expansion of erythroid precursors and accelerated differentiation to replenish erythrocytes. Paracrine signals-involving cooperation between stem cell factor (SCF)/Kit signaling and other signaling inputs-are required for the increased erythroid precursor activity in anemia. Our prior work revealed that the sterile alpha motif (SAM) domain 14 (Anemia is a condition in which the body has a shortage of healthy red blood cells to carry enough oxygen to support its organs. A range of factors are known to cause anemia, including traumatic blood loss, toxins or nutritional deficiency. An estimated one-third of all women of reproductive age are anemic, which can cause tiredness, weakness and shortness of breath. Severe anemia drives the release of hormones and growth factors, leading to a rapid regeneration of precursor red blood cells to replenish the supply in the blood. To understand how red blood cell regeneration is controlled, Ray et al. studied proteins involved in regenerating blood using mice in which anemia had been induced with chemicals. Previous research had shown that the protein Samd14 is produced at higher quantities in individuals with anemia, and is involved with the recovery of lost red blood cells. However, it is not known how the Samd14 protein plays a role in regenerating blood cells, or whether Samd14 interacts with other proteins required for red blood cell production. To shed light on these questions, mouse cells exposed to anemia conditions were used to see what proteins Samd14 binds to. Purifying Samd14 revealed that it interacts with the actin capping protein. This interaction relies on a specific region of Samd14 that is similar to regions in other proteins that bind capping proteins. Ray et al. found that the interaction between Samd14 and the actin capping protein increased the signals needed for the development and survival of new red blood cells. These results identify a signaling mechanism that, if disrupted, could cause anemia to develop. They lead to a better understanding of how our bodies recover from anemia, and potential avenues to treat this condition.
- Published
- 2021
36. Malondialdehyde-Acetaldehyde Extracellular Matrix Protein Adducts Attenuate Unfolded Protein Response During Alcohol and Smoking–Induced Pancreatitis
- Author
-
Rakesh, Bhatia, Christopher M, Thompson, Emalie J, Clement, Koelina, Ganguly, Jesse L, Cox, Sanchita, Rauth, Jawed Akhtar, Siddiqui, Simran S, Mashiana, Maneesh, Jain, Todd A, Wyatt, Harmeet S, Mashiana, Shailender, Singh, Nicholas T, Woods, Kusum K, Kharbanda, Surinder K, Batra, and Sushil, Kumar
- Subjects
Proteomics ,Aldehydes ,Extracellular Matrix Proteins ,Ethanol ,Proteome ,Hepatology ,Smoking ,Gastroenterology ,Acetaldehyde ,Cyclin-Dependent Kinases ,Mice ,Malondialdehyde ,Pancreatitis, Chronic ,Acute Disease ,Unfolded Protein Response ,Animals ,Ceruletide - Abstract
Epidemiological studies have established alcohol and smoking as independent risk factors for recurrent acute pancreatitis and chronic pancreatitis. However, the molecular players responsible for the progressive loss of pancreatic parenchyma and fibroinflammatory response are poorly characterized.Tandem mass tag-based proteomic and bioinformatics analyses were performed on the pancreata of mice exposed to alcohol, cigarette smoke, or a combination of alcohol and cigarette smoke. Biochemical, immunohistochemistry, and transcriptome analyses were performed on the pancreatic tissues and primary acinar cells treated with cerulein in combination with ethanol (50 mmol/L) and cigarette smoke extract (40 μg/mL) for the mechanistic studies.A unique alteration in the pancreatic proteome was observed in mice exposed chronically to the combination of alcohol and cigarette smoke (56.5%) compared with cigarette smoke (21%) or alcohol (17%) alone. The formation of toxic metabolites (P.001) and attenuated unfolded protein response (P.04) were the significantly altered pathways on combined exposure. The extracellular matrix (ECM) proteins showed stable malondialdehyde-acetaldehyde (MAA) adducts in the pancreata of the combination group and chronic pancreatitis patients with a history of smoking and alcohol consumption. Interestingly, MAA-ECM adducts significantly suppressed expression of X-box-binding protein-1, leading to acinar cell death in the presence of alcohol and smoking. The stable MAA-ECM adducts persist even after alcohol and smoking cessation, and significantly delay pancreatic regeneration by abrogating the expression of cyclin-dependent kinases (CDK7 and CDK5) and regeneration markers.The combined alcohol and smoking generate stable MAA-ECM adducts that increase endoplasmic reticulum stress and acinar cell death due to attenuated unfolded protein response and suppress expression of cell cycle regulators. Targeting aldehyde adducts might provide a novel therapeutic strategy for the management of recurrent acute pancreatitis and chronic pancreatitis.
- Published
- 2022
- Full Text
- View/download PDF
37. The Proteomic Landscape of Pancreatic Ductal Adenocarcinoma Liver Metastases Identifies Molecular Subtypes and Associations with Clinical Response – Response
- Author
-
Henry C.-H. Law, Emalie J. Clement, Paul M. Grandgenett, Michael A. Hollingsworth, and Nicholas T. Woods
- Subjects
Pancreatic Neoplasms ,Proteomics ,Cancer Research ,Oncology ,Liver Neoplasms ,Humans ,Article ,Carcinoma, Pancreatic Ductal - Published
- 2021
38. Ctdp1 deficiency leads to early embryonic lethality in mice and defects in cell cycle progression in MEFs
- Author
-
Henry C.-H. Law, Shannon M. Buckley, Emalie J. Clement, Kimiko L. Krieger, Yi Xiao, Fangfang Qiao, and Nicholas T. Woods
- Subjects
Cell cycle checkpoint ,Splice site mutation ,biology ,embryonic lethality ,Cell growth ,QH301-705.5 ,Science ,Cyclin B ,Cre recombinase ,knockout ,ccfdn ,Cell cycle ,mefs ,General Biochemistry, Genetics and Molecular Biology ,Cell biology ,cell death ,cell cycle arrest ,Knockout mouse ,biology.protein ,Transcriptional regulation ,Biology (General) ,General Agricultural and Biological Sciences ,ctdp1 ,Research Article - Abstract
RNA polymerase II subunit A Carboxy-Terminal Domain Phosphatase 1 (CTDP1), a member of the haloacid dehalogenase superfamily phosphatases, has a defined role in transcriptional regulation, but emerging evidence suggests an expanded functional repertoire in the cell cycle and DNA damage response. In humans, a splice site mutation in CTDP1 gives rise to the rare Congenital Cataracts Facial Dysmorphism and Neuropathy syndrome, and recent evidence from our lab indicates CTDP1 is required for breast cancer growth and proliferation. To explore the physiological function of CTDP1 in a mammalian system, we generated a conditional Ctdp1 knockout mouse model by insertion of loxP sites upstream of exon 3 and downstream of exon 4. Biallelic deletion of Ctdp1 results in lethality before embryonic day 7.5, with morphological features indicating embryo cell death and resorption. However, Ctdp1+/− mice are haplosufficient for phenotypic traits including body weight, hematological parameters, exploratory and locomotive functions. To investigate the potential mechanisms of the embryonic death caused by biallelic Ctdp1 knockout, mouse embryonic fibroblasts (MEFs) were established from Ctdp1+/+ and Ctdp1flox/flox mice. Lentivirus delivered Cre-mediated biallelic deletion of Ctdp1 in MEFs results in cell death preceded by impaired proliferation characterized by an increase in G1- and G2-phase populations and a reduction in the S-phase population. These cell cycle alterations caused by deletion of Ctdp1 are associated with an increase in p27 protein expression and a decrease in phosphorylated RB, phosphorylated Histone H3, and Cyclin B expression. Together, these results reveal that Ctdp1 plays an essential role in early mouse embryo development and cell growth and survival in part by regulating the cell cycle., Summary: Knockout of Ctdp1 reveals its essential role in mammalian embryogenesis and regulation of the cell cycle.
- Published
- 2021
39. Spirocyclic dimer SpiD7 activates the unfolded protein response to selectively inhibit growth and induce apoptosis of cancer cells
- Author
-
Smit Kour, Sandeep Rana, Sydney P. Kubica, Smitha Kizhake, Mudassier Ahmad, Catalina Muñoz-Trujillo, David Klinkebiel, Sarbjit Singh, Jayapal Reddy Mallareddy, Surabhi Chandra, Nicholas T. Woods, Adam R. Karpf, and Amarnath Natarajan
- Subjects
Cell Line, Tumor ,Carcinoma ,Drug Discovery ,Eukaryotic Initiation Factor-2 ,Unfolded Protein Response ,Humans ,Apoptosis ,Cell Biology ,Endoplasmic Reticulum ,Endoplasmic Reticulum Stress ,Molecular Biology ,Biochemistry - Abstract
The unfolded protein response (UPR) is an adaptation mechanism activated to resolve transient accumulation of unfolded/misfolded proteins in the endoplasmic reticulum. Failure to resolve the transient accumulation of such proteins results in UPR-mediated programmed cell death. Loss of tumor suppressor gene or oncogene addiction in cancer cells can result in sustained higher basal UPR levels; however, it is not clear if these higher basal UPR levels in cancer cells can be exploited as a therapeutic strategy. We hypothesized that covalent modification of surface-exposed cysteine (SEC) residues could simulate unfolded/misfolded proteins to activate the UPR, and that higher basal UPR levels in cancer cells would provide the necessary therapeutic window. To test this hypothesis, here we synthesized analogs that can covalently modify multiple SEC residues and evaluated them as UPR activators. We identified a spirocyclic dimer, SpiD7, and evaluated its effects on UPR activation signals, that is, XBP1 splicing, phosphorylation of eIF2α, and a decrease in ATF 6 levels, in normal and cancer cells, which were further confirmed by RNA-Seq analyses. We found that SpiD7 selectively induced caspase-mediated apoptosis in cancer cells, whereas normal cells exhibited robust XBP1 splicing, indicating adaptation to stress. Furthermore, SpiD7 inhibited the growth of high-grade serous carcinoma cell lines ~3-15-fold more potently than immortalized fallopian tube epithelial (paired normal control) cells and reduced clonogenic growth of high-grade serous carcinoma cell lines. Our results suggest that induction of the UPR by covalent modification of SEC residues represents a cancer cell vulnerability and can be exploited to discover novel therapeutics.
- Published
- 2022
- Full Text
- View/download PDF
40. UBR5 HECT domain mutations identified in mantle cell lymphoma control maturation of B cells
- Author
-
Nicholas T. Woods, Catalina Amador, Shannon M. Buckley, Samantha A. Swenson, Heather Vahle, Michael R. Green, Henry C.-H. Law, Tyler J. Gilbreath, Jared H. Graham, and R. Willow Hynes-Smith
- Subjects
0301 basic medicine ,HECT domain ,Spliceosome ,Immunobiology and Immunotherapy ,Ubiquitin-Protein Ligases ,Immunology ,Protein domain ,Lymphoma, Mantle-Cell ,Biochemistry ,Malignant transformation ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Ubiquitin ,Protein Domains ,Animals ,Humans ,B-Lymphocytes ,biology ,Chemistry ,Alternative splicing ,Cell Biology ,Hematology ,Mice, Mutant Strains ,Cell biology ,Ubiquitin ligase ,Neoplasm Proteins ,030104 developmental biology ,030220 oncology & carcinogenesis ,RNA splicing ,Mutation ,biology.protein - Abstract
Coordination of a number of molecular mechanisms including transcription, alternative splicing, and class switch recombination are required to facilitate development, activation, and survival of B cells. Disruption of these pathways can result in malignant transformation. Recently, next-generation sequencing has identified a number of novel mutations in mantle cell lymphoma (MCL) patients including mutations in the ubiquitin E3 ligase UBR5. Approximately 18% of MCL patients were found to have mutations in UBR5, with the majority of mutations within the HECT domain of the protein that can accept and transfer ubiquitin molecules to the substrate. Determining if UBR5 controls the maturation of B cells is important to fully understand malignant transformation to MCL. To elucidate the role of UBR5 in B-cell maturation and activation, we generated a conditional mutant disrupting UBR5′s C-terminal HECT domain. Loss of the UBR5 HECT domain leads to a block in maturation of B cells in the spleen and upregulation of proteins associated with messenger RNA splicing via the spliceosome. Our studies reveal a novel role of UBR5 in B-cell maturation by stabilization of spliceosome components during B-cell development and suggests UBR5 mutations play a role in MCL transformation.
- Published
- 2020
41. Lrig1 Regulates Epithelial to Mesenchyme Transition (EMT) in Pancreatic Duct Glands (PDG), an Epithelial Stem Cell Compartment Important in Regeneration and Cancer
- Author
-
Michael A. Hollingsworth, Pinaki Mondal, Sarah Perry Thayer, Tristan S. Caffrey, Kyle L. McAndrews, Paul M. Grandgenett, Dulce Maroni, Vikash Kansal, Jeffrey D. Price, and Nicholas T. Woods
- Subjects
medicine.anatomical_structure ,Chemistry ,Mesenchyme ,Regeneration (biology) ,Mesenchymal stem cell ,Cell ,medicine ,Compartment (development) ,Epithelial–mesenchymal transition ,Stem cell ,Epithelium ,Cell biology - Abstract
Lrig1, an epithelial stem cell marker and tumor suppressor, is uniquely expressed within PDGs in pancreas. PDGs are an epithelial progenitor compartment and likely compartment-of-origin for PDAC. Here, in vivo GFP-lineage tagging of Lrig1 expressing PDG cells identify GFP tagged cells in mesenchyme but not in the main duct epithelium, identifying a cell within a PDG epithelial compartment capable of EMT to regenerate its own micro-environment. Mechanisms regulating this EMT switch results from a loss of Lrig1. Characterization of the GFP-positive mesenchyme and PDG cells confirms EMT. ScRNASeq of epithelial spheroids revealed enhanced mesenchymal features in Lrig1 deficient cells, while Lrig1 overexpression abrogates mesenchymal differentiation. In vivo, in vitro and ex vivo studies reveal the Lrig1 loss within this stem cell compartment results in enhanced EGF and TGF-β signaling, expansion of stem cell, proliferative and mesenchymal markers in addition to the up-regulation of a master regulator of stemness and EMT, PSPC1. In vivo murine models of PDAC (KPC) which tag and lose Lrig1 in the PDG compartment results in the expansion of the PDG stem cell compartment which now resembles low-grade PanINs in addition to the formation of an extensive GFP-positive desmoplasia. In vitro, Lrig1 regulates proliferation and EMT in cancer cell lines. These studies identify Lrig1 as a regulator of stemness and EMT in regeneration and cancer.
- Published
- 2020
- Full Text
- View/download PDF
42. Abstract PO-127: Proteome profiling of pancreatic Ductal Adenocarcinoma (PDAC) primary tumors in Caucasian, African Americans and Latinx patients
- Author
-
Henry C.-H. Law, Andrea N. Riner, Jose G. Trevino, and Nicholas T. Woods
- Subjects
Oncology ,Epidemiology - Abstract
The clinical management of pancreatic ductal adenocarcinoma (PDAC) faces difficult challenges due to its aggressive metastatic potential, complex microenvironment, and lack of targeted therapies. Health disparities also exacerbate these challenges. For instance, Black and African American (AA) patients have higher incidence rates and worse clinical outcomes than White patients even when socioeconomic and tumor stages are controlled. To advance the understanding of the biological differences across the racial groups, PDAC primary tumors collected from 30 Caucasian, 12 African American (AA) and 3 Latinx patients were analyzed by quantitative proteomics. In collaboration with the IDeA National Resource for Quantitative Proteomics, 5820 proteins were identified and quantified using data-independent acquisition (DIA) in the tumor proteome. Comparing the Latinx and the Caucasian tumor proteome, 120 and 95 proteins were found up- and down-regulated in the Latinx proteome, respectively. Proteins involved in the fatty acid metabolism, urea cycle, bile acid and bile salt metabolism were found enriched among the upregulated proteins. 108 and 75 proteins were found up- and down-regulated in African American tumor proteome over the Caucasians, respectively. The 108 upregulated proteins were submitted for Reactome Pathway Analysis. Pathways such as the complement cascade, extracellular matrix (ECM) organization and ECM proteoglycans were found enriched. Haptoglobin-related protein (HPR) was one of the 108 upregulated proteins in the AA tumor proteome, which is also observed at the transcript level in the The Cancer Genome Atlas data. The HPR is known for its trypanolytic function and gene amplifications are observed in those of African descent. HPR works with haptoglobin to clear the free hemoglobin in blood to prevent oxidative damage. We believe that the proteins overexpressed, and the biological processes activated are contributing to the PDAC disparities observed in the African descendants. Therefore, the characterization of the PDAC proteome is a valuable method to delineate the underlying molecular signatures that may contribute to the health disparities. Citation Format: Henry C.-H. Law, Andrea N. Riner, Jose G. Trevino, Nicholas T. Woods. Proteome profiling of pancreatic Ductal Adenocarcinoma (PDAC) primary tumors in Caucasian, African Americans and Latinx patients [abstract]. In: Proceedings of the AACR Virtual Conference: 14th AACR Conference on the Science of Cancer Health Disparities in Racial/Ethnic Minorities and the Medically Underserved; 2021 Oct 6-8. Philadelphia (PA): AACR; Cancer Epidemiol Biomarkers Prev 2022;31(1 Suppl):Abstract nr PO-127.
- Published
- 2022
- Full Text
- View/download PDF
43. Abstract PO-005: Proteome profiling of pancreatic ductal adenocarcinoma (PDAC) primary tumors in Caucasian, African American and Latinx patients
- Author
-
Jose G. Trevino, Henry C.-H. Law, Andrea N. Riner, and Nicholas T. Woods
- Subjects
Cancer Research ,biology ,business.industry ,Quantitative proteomics ,Haptoglobin ,Cancer ,medicine.disease ,Oncology ,Downregulation and upregulation ,Pancreatic cancer ,Proteome ,Cancer research ,medicine ,biology.protein ,business ,Gene ,Extracellular matrix organization - Abstract
The clinical management of pancreatic ductal adenocarcinoma (PDAC) faces difficult challenges due to its aggressive metastatic potential, complex microenvironment, and lack of targeted therapies. Health disparities also exacerbate these challenges. For instance, African and African Americans have higher incidence rates and worse clinical outcomes than Whites even when socioeconomic and tumor stages are controlled. To advance the understanding of the biological differences across the racial groups, the PDAC primary tumors collected from 30 Caucasian, 12 African American (AA) and 3 Latinx patients were analyzed by quantitative proteomics. In collaboration with the IDeA National Resource for Quantitative Proteomics, 5820 proteins were identified and quantified using data-independent acquisition (DIA) in the tumor proteome. Comparing the Latinx and the Caucasian tumor proteome, 120 and 95 proteins were found up- and down-regulated in the Latinx proteome. Proteins involved in the fatty acid metabolism, urea cycle, bile acid and bile salt metabolism were found enriched among the upregulated proteins. 108 and 75 proteins were found up- and down-regulated in African American tumor proteome over the Caucasians. The 108 upregulated proteins were submitted for Reactome Pathway Analysis. Pathways such as the complement cascade, extracellular matrix organization and ECM proteoglycans were found enriched. Haptoglobin-related protein (HPR) was one of the 108 upregulated proteins in the AA tumor proteome, which is also observed at the transcript level in the TCGA data. The HPR is known for its trypanolytic function and gene amplifications are observed in those of African descent. HPR works with haptoglobin (HP) to clear the free hemoglobin in blood to prevent oxidative damage. We believe that the proteins overexpressed, and the biological processes activated are contributing to the PDAC disparities observed in the African descendants. Therefore, the characterization of the PDAC proteome is a valuable method to delineate the underlying molecular signatures that may contribute to the health disparities. Citation Format: Henry C. H. Law, Andrea N. Riner, Jose G. Trevino, Nicholas T. Woods. Proteome profiling of pancreatic ductal adenocarcinoma (PDAC) primary tumors in Caucasian, African American and Latinx patients [abstract]. In: Proceedings of the AACR Virtual Special Conference on Pancreatic Cancer; 2021 Sep 29-30. Philadelphia (PA): AACR; Cancer Res 2021;81(22 Suppl):Abstract nr PO-005.
- Published
- 2021
- Full Text
- View/download PDF
44. Functional Requirements of a Samd14-Capping Protein Interaction in Stress Erythropoiesis
- Author
-
Kyle J. Hewitt, Linda Chee, Suhita Ray, and Nicholas T. Woods
- Subjects
Stress (mechanics) ,Chemistry ,Immunology ,Erythropoiesis ,Functional requirement ,Cell Biology ,Hematology ,Biochemistry ,Cell biology - Abstract
Stress erythropoiesis describes the process of accelerating red blood cell (RBC) production in anemia. Among a number of important mediators of stress erythropoiesis, paracrine signals - involving cooperation between SCF/c-Kit signaling and other signaling inputs - are required for the activation/function of stress erythroid progenitors. Whereas many critical factors required to drive erythropoiesis in normal physiological conditions have been described, whether distinct mechanisms control developmental, steady-state, and stress erythropoiesis in anemia is poorly understood. Our prior work revealed that the Sterile Alpha Motif (SAM) Domain 14 (Samd14) gene is transcriptionally upregulated in a model of acute hemolytic anemia induced by the RBC-lysing chemical phenylhydrazine. Samd14 is regulated by GATA binding transcription factors via an intronic enhancer (Samd14-Enh). In a mouse knockout of Samd14-Enh (Samd14-Enh -/-), we established that the Samd14-Enh is dispensable for steady-state erythropoiesis but is required for recovery from severe hemolytic anemia. Samd14 promotes c-Kit signaling in vivo and ex vivo, and the SAM domain of Samd14 facilitates c-Kit-mediated cellular signaling and stress progenitor activity. In addition, the Samd14 SAM domain is functionally distinct from closely related SAM domains, which demonstrates a unique role for this SAM domain in stress signaling and cell survival. In our working model, Samd14-Enh is part of an ensemble of anemia-responsive enhancers which promote stress erythroid progenitor activity. However, the mechanism underlying Samd14's role in stress erythropoiesis is unknown. To identify potential Samd14-interacting proteins that mediate its function, we performed immunoprecipitation-mass spectrometry on the Samd14 protein. We found that Samd14 interacted with α- and β heterodimers of the F-actin capping protein (CP) complex independent of the SAM domain. CP binds to actin during filament assembly/disassembly and plays a role in cell morphology, migration, and signaling. Deleting a 17 amino acid sequence near the N-terminus of Samd14 disrupted the Samd14-CP interaction. However, mutating the canonical RxR of the CP interaction (CPI) motif, which is required for CP-binding in other proteins, does not abrogate the Samd14-CP interaction. Moreover, replacing this sequence with the canonical CPI domain of CKIP-1 completely disrupts the interaction, indicating that other sequence features are required to maintain the Samd14-CP complex. Ex vivo knockdown of the β-subunit of CP (CPβ), which disrupts the integrity of the CP complex, decreased the percentage of early erythroid precursors (p To test the function of the Samd14-CP complex, we designed an ex vivo genetic complementation assay to express Samd14 lacking the CPB-domain (Samd14∆CPB) in stress erythroid progenitors isolated from anemic Samd14-Enh -/- mice. Phospho-AKT (Ser473) and phospho-ERK (Thr202/Tyr204) levels in Samd14∆CPB were, respectively, 2.2 fold (p=0.007) and ~7 fold (n=3) lower than wild type Samd14 expressing cells, 5 min post SCF stimulation. Relative to Samd14, Samd14∆CPB expression reduced burst forming unit-erythroid (BFU-E) (2.0 fold) and colony forming unit-erythroid (CFU-E) (1.5 fold). These results revealed that the Samd14-CP interaction is a determinant of BFU-E and CFU-E progenitor cell levels and function. Remarkably, as the requirement of the CPB domain in BFU-E and CFU-E progenitors is distinct from the Samd14-SAM domain (which promotes BFU-E but not CFU-E), the function of Samd14 in these two cell types may differ. Ongoing studies will examine whether the function of Samd14 extends beyond SCF/c-Kit signaling and establish cell type-dependent functions of Samd14 and Samd14-interacting proteins. Given the critical importance of c-Kit signaling in hematopoiesis, the role of Samd14 in mediating pathway activation, and our discovery implicating the capping protein complex in erythropoiesis, it is worth considering the pathological implications of this mechanism in acute/chronic anemia and leukemia. Disclosures No relevant conflicts of interest to declare.
- Published
- 2021
- Full Text
- View/download PDF
45. Aminopyrazole based CDK9 PROTAC sensitizes pancreatic cancer cells to venetoclax
- Author
-
Hannah M. King, Edward L. Ezell, Smitha Kizhake, Muhammad Zahid, Sydney P. Kubica, Michael J. Naldrett, Jayapal Reddy Mallareddy, Amarnath Natarajan, Henry C.-H. Law, Sophie Alvarez, Nicholas T. Woods, and Sandeep Rana
- Subjects
Clinical Biochemistry ,Pharmaceutical Science ,Antineoplastic Agents ,01 natural sciences ,Biochemistry ,Article ,Structure-Activity Relationship ,chemistry.chemical_compound ,Cyclin-dependent kinase ,Drug Discovery ,Humans ,Kinome ,Protein Kinase Inhibitors ,Molecular Biology ,Cell Proliferation ,Sulfonamides ,Dose-Response Relationship, Drug ,Molecular Structure ,biology ,010405 organic chemistry ,Venetoclax ,Kinase ,Organic Chemistry ,Proteolysis targeting chimera ,Bridged Bicyclo Compounds, Heterocyclic ,Cyclin-Dependent Kinase 9 ,0104 chemical sciences ,Pancreatic Neoplasms ,010404 medicinal & biomolecular chemistry ,chemistry ,Proteolysis ,Cancer research ,biology.protein ,Pyrazoles ,Molecular Medicine ,Oncogene MYC ,Cyclin-dependent kinase 9 ,Drug Screening Assays, Antitumor ,Growth inhibition - Abstract
Cyclin-dependent kinase 9 (CDK9) is a member of the cyclin-dependent kinase (CDK) family which is involved in transcriptional regulation of several genes, including the oncogene Myc, and is a validated target for pancreatic cancer. Here we report the development of an aminopyrazole based proteolysis targeting chimera (PROTAC 2) that selectively degrades CDK9 (DC50 = 158 ± 6 nM). Mass spectrometry-based kinome profiling shows PROTAC 2 selectively degrades CDK9 in MiaPaCa2 cells and sensitizes them to Venetoclax mediated growth inhibition.
- Published
- 2021
- Full Text
- View/download PDF
46. Functional Impacts of the BRCA1-mTORC2 Interaction in Breast Cancer
- Author
-
Kimiko L. Krieger, Wen-Feng Hu, Nicholas T. Woods, and Tyler J. Ripperger
- Subjects
endocrine system diseases ,protein-protein interactions ,Fluorescent Antibody Technique ,mTORC1 ,medicine.disease_cause ,DNA damage response ,mTORC2 ,0302 clinical medicine ,PRR5 ,Phosphorylation ,skin and connective tissue diseases ,Spectroscopy ,0303 health sciences ,Mutation ,BRCT domain ,BRCA1 Protein ,Intracellular Signaling Peptides and Proteins ,General Medicine ,3. Good health ,Computer Science Applications ,030220 oncology & carcinogenesis ,Signal Transduction ,DNA damage ,Cell Survival ,Breast Neoplasms ,Mechanistic Target of Rapamycin Complex 2 ,Biology ,Catalysis ,Article ,Inorganic Chemistry ,03 medical and health sciences ,Breast cancer ,Cell Line, Tumor ,Two-Hybrid System Techniques ,medicine ,Humans ,Physical and Theoretical Chemistry ,Molecular Biology ,PI3K/AKT/mTOR pathway ,030304 developmental biology ,Organic Chemistry ,mTORC2 complex ,medicine.disease ,BRCA1 ,Cancer research ,SIN1 ,Homologous recombination ,Carcinogenesis ,RICTOR ,DNA Damage - Abstract
Deleterious mutations in Breast Cancer 1 (BRCA1) are associated with an increased risk of breast and ovarian cancer. Mutations in the tandem BRCA1 C-terminal (tBRCT) protein domain disrupt critical protein interactions required for the faithful repair of DNA through homologous recombination, which contributes to oncogenesis. Our studies have identified RICTOR, PRR5, and SIN1 subunits of the mammalian target of rapamycin complex 2 (mTORC2) as interacting partners with the tBRCT domain of BRCA1 leading to the disruption of the mTORC2 complex. However, the interplay between mTORC2 signaling and BRCA1 function in the DNA damage response (DDR) remains to be determined. In this study, we used protein interaction assays to determine the binary interactions between the tBRCT domain and mTORC2 subunits, evaluated the impact of mTOR inhibition on the transcriptional function of the tBRCT, evaluated the impact of mTOR signaling on BRCA1 recruitment to DNA damage-induced foci and determined the breast cancer cell line response to mTOR inhibition dependent upon BRCA1 expression and mutation. This study determined that PRR5, RICTOR, and SIN1 could each independently interact with the BRCA1 tBRCT. Inhibition of mTORC1, but not mTORC1/2, increases BRCA1 transcriptional activation activity. Treatment with pan-mTOR inhibitor PP242 diminishes DNA damage-induced &gamma, H2AX and BRCA1 foci formation. Breast cancer cells lacking expression of functional BRCA1 are more sensitive to mTOR inhibitors. These data suggest that mTOR signaling is required for BRCA1 response to DNA damage and breast cancer cells lacking BRCA1 are more sensitive to pan-mTOR inhibition. This work suggests chemotherapeutic strategies using mTOR inhibitors could be tailored for patients that lack functional BRCA1.
- Published
- 2019
47. Proximity Labeling To Map Host-Pathogen Interactions at the Membrane of a Bacterium-Containing Vacuole in Chlamydia trachomatis-Infected Human Cells
- Author
-
Scot P. Ouellette, Elizabeth A. Rucks, Macy G. Olson, Lisa M. Jorgenson, Alyssa Lawrence, Ray E. Widner, Dragana Lagundzin, and Nicholas T. Woods
- Subjects
APEX2 ,Inc proteins ,Immunoprecipitation ,Immunology ,Chlamydia trachomatis ,Vacuole ,Biology ,medicine.disease_cause ,Microbiology ,Interactome ,LRRF1 ,Mass Spectrometry ,03 medical and health sciences ,inclusion membrane ,Bacterial Proteins ,medicine ,host-pathogen interactions ,Humans ,Biotinylation ,030304 developmental biology ,0303 health sciences ,Cellular Microbiology: Pathogen-Host Cell Molecular Interactions ,030306 microbiology ,Intracellular parasite ,Membrane Proteins ,Gene Expression Regulation, Bacterial ,Recombinant Proteins ,3. Good health ,Cell biology ,Cytosol ,Infectious Diseases ,Membrane protein ,Parasitology ,Streptavidin ,proximity labeling ,HeLa Cells - Abstract
Many intracellular bacteria, including the obligate intracellular pathogen Chlamydia trachomatis, grow within a membrane-bound bacterium-containing vacuole (BCV). Secreted cytosolic effectors modulate host activity, but an understanding of the host-pathogen interactions that occur at the BCV membrane is limited by the difficulty in purifying membrane fractions from infected host cells., Many intracellular bacteria, including the obligate intracellular pathogen Chlamydia trachomatis, grow within a membrane-bound bacterium-containing vacuole (BCV). Secreted cytosolic effectors modulate host activity, but an understanding of the host-pathogen interactions that occur at the BCV membrane is limited by the difficulty in purifying membrane fractions from infected host cells. We used the ascorbate peroxidase (APEX2) proximity labeling system, which labels proximal proteins with biotin in vivo, to study the protein-protein interactions that occur at the chlamydial vacuolar, or inclusion, membrane. An in vivo understanding of the secreted chlamydial inclusion membrane protein (Inc) interactions (e.g., Inc-Inc and Inc-eukaryotic protein) and how these contribute to overall host-chlamydia interactions at this unique membrane is lacking. We hypothesize some Incs organize the inclusion membrane, whereas other Incs bind eukaryotic proteins to promote chlamydia-host interactions. To study this, Incs fused to APEX2 were expressed in C. trachomatis L2. Affinity purification-mass spectrometry (AP-MS) identified biotinylated proteins, which were analyzed for statistical significance using significance analysis of the interactome (SAINT). Broadly supporting both Inc-Inc and Inc-host interactions, our Inc-APEX2 constructs labeled Incs as well as known and previously unreported eukaryotic proteins localizing to the inclusion. We demonstrate, using bacterial two-hybrid and coimmunoprecipitation assays, that endogenous LRRFIP1 (LRRF1) is recruited to the inclusion by the Inc CT226. We further demonstrate interactions between CT226 and the Incs used in our study to reveal a model for inclusion membrane organization. Combined, our data highlight the utility of APEX2 to capture the complex in vivo protein-protein interactions at the chlamydial inclusion.
- Published
- 2019
48. E3 Ligase UBR5 HECT domain mutations in lymphoma control maturation of B cells via alternative splicing
- Author
-
Jared H. Graham, Shannon M. Buckley, R. Willow Hynes-Smith, Samantha A. Swenson, Henry Chun Hin Law, Nicholas T. Woods, Tyler J. Gilbreath, Heather Vahle, and Michael R. Green
- Subjects
HECT domain ,0303 health sciences ,Spliceosome ,biology ,Alternative splicing ,Ubiquitin ligase ,Cell biology ,03 medical and health sciences ,0302 clinical medicine ,medicine.anatomical_structure ,Ubiquitin ,Immunoglobulin class switching ,030220 oncology & carcinogenesis ,RNA splicing ,biology.protein ,medicine ,B cell ,030304 developmental biology - Abstract
Coordination of a number of molecular mechanisms including transcription, alternative splicing, and class switch recombination are required to facilitate development, activation, and survival of B cells. Disruption of these pathways can result in malignant transformation. Recently, next generation sequencing has identified a number of novel mutations in mantle cell lymphoma (MCL) patients including the ubiquitin E3 ligase UBR5. Approximately 18% of MCL patients were found to have mutations in UBR5 with the majority of mutations within the HECT domain of the protein which can accept and transfer ubiquitin molecules to the substrate. Determining if UBR5 controls the maturation of B cells is important to fully understand malignant transformation to MCL. To elucidate the role of UBR5 in B cell maturation and activation we generated a conditional mutant disrupting UBR5’s C-terminal HECT domain. Loss of the UBR5 HECT domain leads to a block in maturation of B cells in the spleen and up-regulation of proteins associated with mRNA splicing via the spliceosome. Our studies reveal a novel role of UBR5 in B cell maturation by regulating alternative splicing of key transcripts during B cell development and suggests UBR5 mutations may promote mantle cell lymphoma initiation.KEY POINTSUtilizing a novel mouse model mimicking MCL patient mutations, the loss of UBR5’s HECT domain causes alterations in B cell development.UBR5 mutations lead to stabilization of UBR5 and aberrant splicing.
- Published
- 2019
- Full Text
- View/download PDF
49. Proximity Labeling to Map Host-Pathogen Interactions at the Membrane of a Bacteria Containing Vacuole inChlamydia trachomatisInfected Human Cells
- Author
-
Macy G. Olson, Ray E. Widner, Elizabeth A. Rucks, Scot P. Ouellette, Lisa M. Jorgenson, Nicholas T. Woods, Alyssa Lawrence, and Dragana Lagundzin
- Subjects
medicine.anatomical_structure ,Membrane protein ,Chemistry ,Intracellular parasite ,Biotinylation ,Lysosome ,medicine ,Vacuole ,Chlamydia trachomatis ,medicine.disease_cause ,Interactome ,Cell biology ,Protein–protein interaction - Abstract
As an obligate intracellular pathogenic bacterium,C. trachomatisdevelops within a membrane-bound vacuole, termed the inclusion. The inclusion membrane is modified by chlamydial inclusion membrane proteins (Incs), which act as the mediators of host-pathogen interactions. Anin vivounderstanding of Inc-Inc and Inc-eukaryotic protein interactions and how these contribute to overall host-chlamydial interactions at this unique membrane is lacking. Previous bacterial two-hybrid studies established that certain Incs have the propensity to bind other Incs while others have limited Inc-Inc interactions. We hypothesize some Incs organize the inclusion membrane whereas other Incs bind eukaryotic proteins to promote chlamydial-host interactions. To test this hypothesis, we used the ascorbate peroxidase proximity labeling system (APEX2), which labels proximal proteins with biotinin vivo, and chose to analyze Inc proteins with varying Inc-binding propensities. We inducibly expressed these Incs fused to APEX2 inChlamydia trachomatisL2, verified their localization and labeling activities by transmission electron microscopy, and used affinity purification-mass spectrometry to identify biotinylated proteins. To analyze our mass spectrometry results for statistical significance, we used Significance Analysis of INTeractome (SAINT), which demonstrated that our Inc-APEX2 constructs labeled Inc proteins as well as known and previously unreported eukaryotic proteins that localize to the inclusion. Our results broadly support two types of Inc interactions: Inc-Inc versus Inc-host. One eukaryotic protein, LRRFIP1 (LRRF1) was found in all of our Inc-APEX2 datasets, which is consistent with previously published AP-MS datasets. For the first time, we demonstrate by confocal and super-resolution microscopy that endogenous LRRF1 localizes to the chlamydial inclusion. We also used bacterial two-hybrid studies and pulldown assays to determine if LRRF1 was identified as a true interacting protein or was proximal to our Inc-APEX2 constructs. Combined, our data highlight the utility of APEX2 to capture the complexin vivoprotein-protein interactions at the chlamydial inclusion.Author summaryMany intracellular bacteria, including the obligate intracellular pathogenChlamydia trachomatis, grow within a membrane-bound “bacteria containing vacuole” (BCV) that, in most cases, prevents association with the lysosome. Secreted cytosolic effectors modulate host activity, but an understanding of the host-pathogen interactions that occur at the BCV membrane is limited by the difficulty in purifying membrane fractions from infected host cells. Here, we used the ascorbate peroxidase proximity labeling system (APEX2), which labels proximal proteins with biotinin vivo, to study the interactions that occur at the chlamydial vacuolar, or inclusion, membrane. The inclusion membrane is modified by chlamydial type III secreted inclusion membrane proteins (Incs), which act as the mediators of host-pathogen interactions. Our results broadly support two types of Inc interactions: Inc-Inc versus Inc-host. Our data highlight the utility of APEX2 to capture the complex protein-protein interactions at a membrane sitein vivoin the context of infection.
- Published
- 2019
- Full Text
- View/download PDF
50. CTDP1 regulates breast cancer survival and DNA repair through BRCT-specific interactions with FANCI
- Author
-
Keith R. Johnson, Xueli Li, Kimiko L. Krieger, Alvaro N.A. Monteiro, Toshiyasu Taniguchi, Nicholas T. Woods, Dragana Lagundžin, Ronald S. Cheung, Wen Feng Hu, and Tadayoshi Bessho
- Subjects
0301 basic medicine ,Cancer Research ,DNA damage ,DNA repair ,Immunology ,lcsh:RC254-282 ,Article ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,0302 clinical medicine ,Breast cancer ,Fanconi anemia ,hemic and lymphatic diseases ,FANCD2 ,medicine ,lcsh:QH573-671 ,Chemistry ,lcsh:Cytology ,Cell Biology ,DNA Repair Pathway ,medicine.disease ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,FANCA ,Cell biology ,Chromatin ,Protein-protein interaction networks ,030104 developmental biology ,030220 oncology & carcinogenesis ,Homologous recombination - Abstract
BRCA1 C-terminal domains are found in a specialized group of 23 proteins that function in the DNA damage response to protect genomic integrity. C-terminal domain phosphatase 1 (CTDP1) is the only phosphatase with a BRCA1 C-terminal domain in the human proteome, yet direct participation in the DNA damage response has not been reported. Examination of the CTDP1 BRCA1 C-terminal domain-specific protein interaction network revealed 103 high confidence interactions enriched in DNA damage response proteins, including FANCA and FANCI that are central to the Fanconi anemia DNA repair pathway necessary for the resolution of DNA interstrand crosslink damage. CTDP1 expression promotes DNA damage-induced FANCA and FANCD2 foci formation and enhances homologous recombination repair efficiency. CTDP1 was found to regulate multiple aspects of FANCI activity, including chromatin localization, interaction with γ-H2AX, and SQ motif phosphorylations. Knockdown of CTDP1 increases MCF-10A sensitivity to DNA interstrand crosslinks and double-strand breaks, but not ultraviolet radiation. In addition, CTDP1 knockdown impairs in vitro and in vivo growth of breast cancer cell lines. These results elucidate the molecular functions of CTDP1 in Fanconi anemia interstrand crosslink repair and identify this protein as a potential target for breast cancer therapy.
- Published
- 2019
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.