116 results on '"Morin GB"'
Search Results
2. Cancer-associated somatic DICER1 hotspot mutations cause defective miRNA processing and reverse-strand expression bias to predominantly mature 3p strands through loss of 5p strand cleavage
- Author
-
Anglesio, MS, Wang, Y, Yang, W, Senz, J, Wan, A, Heravi-Moussavi, A, Salamanca, C, Maines-Bandiera, S, Huntsman, DG, and Morin, GB
- Published
- 2013
- Full Text
- View/download PDF
3. Phase Ib pharmacodynamic study of the MNK inhibitor Tomivosertib (eFT508) combined with paclitaxel in patients with refractory metastatic breast cancer.
- Author
-
Ferrario C, Mackey J, Gelmon KA, Levasseur N, Sorensen PH, Oo HZ, Negri GL, Tse VWL, Spencer SE, Cheng G, Morin GB, Del Rincon S, Cotechini T, Gonçalves C, Hindmarch CCT, Miller WH, Amiri M, Basiri T, Villareal-Corpuz V, Sperry S, Gregorczyk K, Spera G, Sonenberg N, and Pollak M
- Abstract
Purpose: Preclinical data motivate clinical evaluation of inhibitors of mitogen-activated protein kinase-interacting kinases 1 and 2 (MNK1/2). We conducted a phase 1b clinical trial to study target engagement and safety of tomivosertib, a MNK1/2 inhibitor, alone and in combination with paclitaxel., Methods: Eligible patients had metastatic breast cancer resistant to standard of care treatments. Biopsies were obtained at baseline and during treatment with tomivosertib, and then tomivosertib was continued with addition of paclitaxel until disease progression or toxicity. Serum drug levels were measured, and pharmacodynamic endpoints included immunohistochemistry, proteomics, translatomics, and imaging mass cytometry., Results: Tomivosertib alone and in combination with paclitaxel was well tolerated. There was no pharmacokinetic interaction between the drugs. We observed a clear reduction in phosphorylation of eIF4E at S209, a major substrate of MNK1/2, and identified tomivosertib-induced perturbations in the proteome, translatome, and cellular populations of biopsied metastatic breast cancer tissue., Conclusion: We conclude that tomivosertib effectively inhibits MNK1/2 activity in metastatic breast cancer tissue, and that it can safely be combined with paclitaxel in future phase II studies. We demonstrate feasibility of using proteomic profiles, translatomic profiles, and spatial distribution of immune cell infiltrates for clinical pharmacodynamic studies.
- Published
- 2024
- Full Text
- View/download PDF
4. Publisher Correction: Translation efficiency driven by CNOT3 subunit of the CCR4-NOT complex promotes leukemogenesis.
- Author
-
Ghashghaei M, Liu Y, Ettles J, Bombaci G, Ramkumar N, Liu Z, Escano L, Miko SS, Kim Y, Waldron JA, Do K, MacPherson K, Yuen KA, Taibi T, Yue M, Arsalan A, Jin Z, Edin G, Karsan A, Morin GB, Kuchenbauer F, Perna F, Bushell M, and Vu LP
- Published
- 2024
- Full Text
- View/download PDF
5. Changes in the tumour microenvironment mark the transition from serous borderline tumour to low-grade serous carcinoma.
- Author
-
Vallejos R, Zhantuyakova A, Negri GL, Martin SD, Spencer SE, Thornton S, Leung S, Lynch B, Qin Y, Chow C, Liang B, Zdravko S, Douglas JM, Milne K, Mateyko B, Nelson BH, Howitt BE, Kommoss FK, Horn LC, Hoang L, Singh N, Morin GB, Huntsman DG, and Cochrane D
- Subjects
- Humans, Female, Biomarkers, Tumor metabolism, Biomarkers, Tumor genetics, Neoplasm Grading, Disease Progression, Proteomics methods, Serine Endopeptidases metabolism, Serine Endopeptidases genetics, Middle Aged, Membrane Proteins metabolism, Gelatinases metabolism, Aged, Endopeptidases metabolism, Cancer-Associated Fibroblasts metabolism, Cancer-Associated Fibroblasts pathology, Lymphocytes, Tumor-Infiltrating immunology, Lymphocytes, Tumor-Infiltrating pathology, Lymphocytes, Tumor-Infiltrating metabolism, Tumor Microenvironment, Ovarian Neoplasms pathology, Ovarian Neoplasms metabolism, Ovarian Neoplasms genetics, Cystadenocarcinoma, Serous pathology, Cystadenocarcinoma, Serous metabolism
- Abstract
Low-grade serous ovarian carcinoma (LGSC) is a rare and lethal subtype of ovarian cancer. LGSC is pathologically, biologically, and clinically distinct from the more common high-grade serous ovarian carcinoma (HGSC). LGSC arises from serous borderline ovarian tumours (SBTs). The mechanism of transformation for SBTs to LGSC remains poorly understood. To better understand the biology of LGSC, we performed whole proteome profiling of formalin-fixed, paraffin-embedded tissue blocks of LGSC (n = 11), HGSC (n = 19), and SBTs (n = 26). We identified that the whole proteome is able to distinguish between histotypes of the ovarian epithelial tumours. Proteins associated with the tumour microenvironment were differentially expressed between LGSC and SBTs. Fibroblast activation protein (FAP), a protein expressed in cancer-associated fibroblasts, is the most differentially abundant protein in LGSC compared with SBT. Multiplex immunohistochemistry (IHC) for immune markers (CD20, CD79a, CD3, CD8, and CD68) was performed to determine the presence of B cells, T cells, and macrophages. The LGSC FAP
+ stroma was associated with greater abundance of Tregs and M2 macrophages, features not present in SBTs. Our proteomics cohort reveals that there are changes in the tumour microenvironment in LGSC compared with its putative precursor lesion, SBT. These changes suggest that the tumour microenvironment provides a supportive environment for LGSC tumourigenesis and progression. Thus, targeting the tumour microenvironment of LGSC may be a viable therapeutic strategy. © 2024 The Pathological Society of Great Britain and Ireland., (© 2024 The Pathological Society of Great Britain and Ireland.)- Published
- 2024
- Full Text
- View/download PDF
6. Mass Spectrometry Acquisition and Fractionation Recommendations for TMT11 and TMT16 Labeled Samples.
- Author
-
Riley RM, Negri GL, Cheng SG, Spencer Miko SE, Morin RD, and Morin GB
- Subjects
- Humans, Chemical Fractionation methods, Staining and Labeling methods, Proteomics methods, Proteomics standards, Tandem Mass Spectrometry methods, Proteome analysis
- Abstract
Proteome coverage and accurate protein quantification are both important for evaluating biological systems; however, compromises between quantification, coverage, and mass spectrometry (MS) resources are often necessary. Consequently, experimental parameters that impact coverage and quantification must be adjusted, depending on experimental goals. Among these parameters is offline prefractionation, which is utilized in MS-based proteomics to decrease sample complexity resulting in higher overall proteome coverage upon MS analysis. Prefractionation leads to increases in required MS analysis time, although this is often mitigated by isobaric labeling using tandem-mass tags (TMT), which allow samples to be multiplexed. Here we evaluate common prefractionation schemes, TMT variants, and MS acquisition methods and their impact on protein quantification and coverage. Furthermore, we provide recommendations for experimental design depending on the experimental goals.
- Published
- 2024
- Full Text
- View/download PDF
7. IMMUNOTAR - Integrative prioritization of cell surface targets for cancer immunotherapy.
- Author
-
Shraim R, Mooney B, Conkrite KL, Weiner AK, Morin GB, Sorensen PH, Maris JM, Diskin SJ, and Sacan A
- Abstract
Cancer remains a leading cause of mortality globally. Recent improvements in survival have been facilitated by the development of less toxic immunotherapies; however, identifying targets for immunotherapies remains a challenge in the field. To address this challenge, we developed IMMUNOTAR, a computational tool that systematically prioritizes and identifies candidate immunotherapeutic targets. IMMUNOTAR integrates user-provided RNA-sequencing or proteomics data with quantitative features extracted from publicly available databases based on predefined optimal immunotherapeutic target criteria and quantitatively prioritizes potential surface protein targets. We demonstrate the utility and flexibility of IMMUNOTAR using three distinct datasets, validating its effectiveness in identifying both known and new potential immunotherapeutic targets within the analyzed cancer phenotypes. Overall, IMMUNOTAR enables the compilation of data from multiple sources into a unified platform, allowing users to simultaneously evaluate surface proteins across diverse criteria. By streamlining target identification, IMMUNOTAR empowers researchers to efficiently allocate resources and accelerate immunotherapy development., Competing Interests: Competing Interests: None
- Published
- 2024
- Full Text
- View/download PDF
8. Translation efficiency driven by CNOT3 subunit of the CCR4-NOT complex promotes leukemogenesis.
- Author
-
Ghashghaei M, Liu Y, Ettles J, Bombaci G, Ramkumar N, Liu Z, Escano L, Miko SS, Kim Y, Waldron JA, Do K, MacPherson K, Yuen KA, Taibi T, Yue M, Arsalan A, Jin Z, Edin G, Karsan A, Morin GB, Kuchenbauer F, Perna F, Bushell M, and Vu LP
- Subjects
- Humans, Carcinogenesis genetics, Cell Differentiation, Receptors, CCR4, Leukemia, Myeloid, Acute genetics, Proteomics, Transcription Factors genetics, Transcription Factors metabolism
- Abstract
Protein synthesis is frequently deregulated during tumorigenesis. However, the precise contexts of selective translational control and the regulators of such mechanisms in cancer is poorly understood. Here, we uncovered CNOT3, a subunit of the CCR4-NOT complex, as an essential modulator of translation in myeloid leukemia. Elevated CNOT3 expression correlates with unfavorable outcomes in patients with acute myeloid leukemia (AML). CNOT3 depletion induces differentiation and apoptosis and delayed leukemogenesis. Transcriptomic and proteomic profiling uncovers c-MYC as a critical downstream target which is translationally regulated by CNOT3. Global analysis of mRNA features demonstrates that CNOT3 selectively influences expression of target genes in a codon usage dependent manner. Furthermore, CNOT3 associates with the protein network largely consisting of ribosomal proteins and translation elongation factors in leukemia cells. Overall, our work elicits the direct requirement for translation efficiency in tumorigenesis and propose targeting the post-transcriptional circuitry via CNOT3 as a therapeutic vulnerability in AML., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
9. Surface and Global Proteome Analyses Identify ENPP1 and Other Surface Proteins as Actionable Immunotherapeutic Targets in Ewing Sarcoma.
- Author
-
Mooney B, Negri GL, Shyp T, Delaidelli A, Zhang HF, Spencer Miko SE, Weiner AK, Radaoui AB, Shraim R, Lizardo MM, Hughes CS, Li A, El-Naggar AM, Rouleau M, Li W, Dimitrov DS, Kurmasheva RT, Houghton PJ, Diskin SJ, Maris JM, Morin GB, and Sorensen PH
- Subjects
- Child, Humans, Membrane Proteins, Proteome, Proteomics, Immunotherapy, Antigens, Neoplasm, Oxidoreductases, Sarcoma, Ewing genetics, Sarcoma, Ewing therapy, Sarcoma, Bone Neoplasms genetics, Bone Neoplasms therapy
- Abstract
Purpose: Ewing sarcoma is the second most common bone sarcoma in children, with 1 case per 1.5 million in the United States. Although the survival rate of patients diagnosed with localized disease is approximately 70%, this decreases to approximately 30% for patients with metastatic disease and only approximately 10% for treatment-refractory disease, which have not changed for decades. Therefore, new therapeutic strategies are urgently needed for metastatic and refractory Ewing sarcoma., Experimental Design: This study analyzed 19 unique Ewing sarcoma patient- or cell line-derived xenografts (from 14 primary and 5 metastatic specimens) using proteomics to identify surface proteins for potential immunotherapeutic targeting. Plasma membranes were enriched using density gradient ultracentrifugation and compared with a reference standard of 12 immortalized non-Ewing sarcoma cell lines prepared in a similar manner. In parallel, global proteome analysis was carried out on each model to complement the surfaceome data. All models were analyzed by Tandem Mass Tags-based mass spectrometry to quantify identified proteins., Results: The surfaceome and global proteome analyses identified 1,131 and 1,030 annotated surface proteins, respectively. Among surface proteins identified, both approaches identified known Ewing sarcoma-associated proteins, including IL1RAP, CD99, STEAP1, and ADGRG2, and many new cell surface targets, including ENPP1 and CDH11. Robust staining of ENPP1 was demonstrated in Ewing sarcoma tumors compared with other childhood sarcomas and normal tissues., Conclusions: Our comprehensive proteomic characterization of the Ewing sarcoma surfaceome provides a rich resource of surface-expressed proteins in Ewing sarcoma. This dataset provides the preclinical justification for exploration of targets such as ENPP1 for potential immunotherapeutic application in Ewing sarcoma. See related commentary by Bailey, p. 934., (©2023 The Authors; Published by the American Association for Cancer Research.)
- Published
- 2024
- Full Text
- View/download PDF
10. ASPSCR1-TFE3 reprograms transcription by organizing enhancer loops around hexameric VCP/p97.
- Author
-
Pozner A, Li L, Verma SP, Wang S, Barrott JJ, Nelson ML, Yu JSE, Negri GL, Colborne S, Hughes CS, Zhu JF, Lambert SL, Carroll LS, Smith-Fry K, Stewart MG, Kannan S, Jensen B, John CM, Sikdar S, Liu H, Dang NH, Bourdage J, Li J, Vahrenkamp JM, Mortenson KL, Groundland JS, Wustrack R, Senger DL, Zemp FJ, Mahoney DJ, Gertz J, Zhang X, Lazar AJ, Hirst M, Morin GB, Nielsen TO, Shen PS, and Jones KB
- Subjects
- Animals, Mice, Humans, Proteomics, Translocation, Genetic, Oncogene Proteins, Fusion genetics, Oncogene Proteins, Fusion metabolism, Chromatin genetics, Basic Helix-Loop-Helix Leucine Zipper Transcription Factors metabolism, Chromosomes, Human, X metabolism, Intracellular Signaling Peptides and Proteins genetics, Valosin Containing Protein genetics, Carcinoma, Renal Cell genetics, Carcinoma, Renal Cell pathology, Kidney Neoplasms genetics
- Abstract
The t(X,17) chromosomal translocation, generating the ASPSCR1::TFE3 fusion oncoprotein, is the singular genetic driver of alveolar soft part sarcoma (ASPS) and some Xp11-rearranged renal cell carcinomas (RCCs), frustrating efforts to identify therapeutic targets for these rare cancers. Here, proteomic analysis identifies VCP/p97, an AAA+ ATPase with known segregase function, as strongly enriched in co-immunoprecipitated nuclear complexes with ASPSCR1::TFE3. We demonstrate that VCP is a likely obligate co-factor of ASPSCR1::TFE3, one of the only such fusion oncoprotein co-factors identified in cancer biology. Specifically, VCP co-distributes with ASPSCR1::TFE3 across chromatin in association with enhancers genome-wide. VCP presence, its hexameric assembly, and its enzymatic function orchestrate the oncogenic transcriptional signature of ASPSCR1::TFE3, by facilitating assembly of higher-order chromatin conformation structures demonstrated by HiChIP. Finally, ASPSCR1::TFE3 and VCP demonstrate co-dependence for cancer cell proliferation and tumorigenesis in vitro and in ASPS and RCC mouse models, underscoring VCP's potential as a novel therapeutic target., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
11. Dependence of human cell survival and proliferation on the CASP3 prodomain.
- Author
-
Eskandari E, Negri GL, Tan S, MacAldaz ME, Ding S, Long J, Nielsen K, Spencer SE, Morin GB, and Eaves CJ
- Abstract
Mechanisms that regulate cell survival and proliferation are important for both the development and homeostasis of normal tissue, and as well as for the emergence and expansion of malignant cell populations. Caspase-3 (CASP3) has long been recognized for its proteolytic role in orchestrating cell death-initiated pathways and related processes; however, whether CASP3 has other functions in mammalian cells that do not depend on its known catalytic activity have remained unknown. To investigate this possibility, we examined the biological and molecular consequences of reducing CASP3 levels in normal and transformed human cells using lentiviral-mediated short hairpin-based knockdown experiments in combination with approaches designed to test the potential rescue capability of different components of the CASP3 protein. The results showed that a ≥50% reduction in CASP3 levels rapidly and consistently arrested cell cycle progression and survival in all cell types tested. Mass spectrometry-based proteomic analyses and more specific flow cytometric measurements strongly implicated CASP3 as playing an essential role in regulating intracellular protein aggregate clearance. Intriguingly, the rescue experiments utilizing different forms of the CASP3 protein showed its prosurvival function and effective removal of protein aggregates did not require its well-known catalytic capability, and pinpointed the N-terminal prodomain of CASP3 as the exclusive component needed in a diversity of human cell types. These findings identify a new mechanism that regulates human cell survival and proliferation and thus expands the complexity of how these processes can be controlled. The graphical abstract illustrates the critical role of CASP3 for sustained proliferation and survival of human cells through the clearance of protein aggregates., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
12. Characterizing the secretome of EGFR mutant lung adenocarcinoma.
- Author
-
Luu JK, Johnson FD, Jajarmi J, Sihota T, Shi R, Lu D, Farnsworth D, Spencer SE, Negri GL, Morin GB, and Lockwood WW
- Abstract
Background: Lung cancer is the leading cause of cancer related death worldwide, mainly due to the late stage of disease at the time of diagnosis. Non-invasive biomarkers are needed to supplement existing screening methods to enable earlier detection and increased patient survival. This is critical to EGFR -driven lung adenocarcinoma as it commonly occurs in individuals who have never smoked and do not qualify for current screening protocols., Methods: In this study, we performed mass spectrometry analysis of the secretome of cultured lung cells representing different stages of mutant EGFR driven transformation, from normal to fully malignant. Identified secreted proteins specific to the malignant state were validated using orthogonal methods and their clinical activity assessed in lung adenocarcinoma patient cohorts., Results: We quantified 1020 secreted proteins, which were compared for differential expression between stages of transformation. We validated differentially expressed proteins at the transcriptional level in clinical tumor specimens, association with patient survival, and absolute concentration to yield three biomarker candidates: MDK, GDF15, and SPINT2. These candidates were validated using ELISA and increased levels were associated with poor patient survival specifically in EGFR mutant lung adenocarcinoma patients., Conclusions: Our study provides insight into changes in secreted proteins during EGFR driven lung adenocarcinoma transformation that may play a role in the processes that promote tumor progression. The specific candidates identified can harnessed for biomarker use to identify high risk individuals for early detection screening programs and disease management for this molecular subgroup of lung adenocarcinoma patients., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2024 Luu, Johnson, Jajarmi, Sihota, Shi, Lu, Farnsworth, Spencer, Negri, Morin and Lockwood.)
- Published
- 2024
- Full Text
- View/download PDF
13. Biallelic Dicer1 Mutations in the Gynecologic Tract of Mice Drive Lineage-Specific Development of DICER1 Syndrome-Associated Cancer.
- Author
-
Wang Y, Chen SY, Ta M, Senz J, Tao LV, Thornton S, Tamvada N, Yang W, Moscovitz Y, Li E, Guo J, Shen C, Douglas JM, Ei-Naggar AM, Kommoss FKF, Underhill TM, Singh N, Gilks CB, Morin GB, and Huntsman DG
- Subjects
- Child, Humans, Female, Animals, Mice, Ribonuclease III genetics, Ribonuclease III metabolism, Mutation, Mutation, Missense, DEAD-box RNA Helicases genetics, MicroRNAs genetics, Neoplastic Syndromes, Hereditary
- Abstract
DICER1 is an RNase III enzyme essential for miRNA biogenesis through cleaving precursor-miRNA hairpins. Germline loss-of-function DICER1 mutations underline the development of DICER1 syndrome, a rare genetic disorder that predisposes children to cancer development in organs such as lung, gynecologic tract, kidney, and brain. Unlike classical tumor suppressors, the somatic "second hit" in DICER1 syndrome-associated cancers does not fully inactivate DICER1 but impairs its RNase IIIb activity only, suggesting a noncanonical two-hit hypothesis. Here, we developed a genetically engineered conditional compound heterozygous Dicer1 mutant mouse strain that fully recapitulates the biallelic DICER1 mutations in DICER1 syndrome-associated human cancers. Crossing this tool strain with tissue-specific Cre strains that activate Dicer1 mutations in gynecologic tract cells at two distinct developmental stages revealed that embryonic biallelic Dicer1 mutations caused infertility in females by disrupting oviduct and endometrium development and ultimately drove cancer development. These multicystic tubal and intrauterine tumors histologically resembled a subset of DICER1 syndrome-associated human cancers. Molecular analysis uncovered accumulation of additional oncogenic events (e.g., aberrant p53 expression, Kras mutation, and Myc activation) in murine Dicer1 mutant tumors and validated miRNA biogenesis defects in 5P miRNA strand production, of which, loss of let-7 family miRNAs was identified as a putative key player in transcriptomic rewiring and tumor development. Thus, this DICER1 syndrome-associated cancer model recapitulates the biology of human cancer and provides a unique tool for future investigation and therapeutic development., Significance: Generation of a Dicer1 mutant mouse model establishes the oncogenicity of missense mutations in the DICER1 RNase IIIb domain and provides a faithful model of DICER1 syndrome-associated cancer for further investigation., (©2023 American Association for Cancer Research.)
- Published
- 2023
- Full Text
- View/download PDF
14. ASPSCR1-TFE3 reprograms transcription by organizing enhancer loops around hexameric VCP/p97.
- Author
-
Pozner A, Verma SP, Li L, Wang S, Barrott JJ, Nelson ML, Yu JSE, Negri GL, Colborne S, Hughes CS, Zhu JF, Lambert SL, Carroll LS, Smith-Fry K, Stewart MG, Kannan S, Jensen B, Mortenson KL, John C, Sikdar S, Liu H, Dang NH, Bourdage J, Li J, Vahrenkamp JM, Groundland JS, Wustrack R, Senger DL, Zemp FJ, Mahoney DJ, Gertz J, Zhang X, Lazar AJ, Hirst M, Morin GB, Nielsen TO, Shen PS, and Jones KB
- Abstract
The t(X,17) chromosomal translocation, generating the ASPSCR1-TFE3 fusion oncoprotein, is the singular genetic driver of alveolar soft part sarcoma (ASPS) and some Xp11-rearranged renal cell carcinomas (RCC), frustrating efforts to identify therapeutic targets for these rare cancers. Proteomic analysis showed that VCP/p97, an AAA+ ATPase with known segregase function, was strongly enriched in co-immunoprecipitated nuclear complexes with ASPSCR1-TFE3. We demonstrate that VCP is a likely obligate co-factor of ASPSCR1-TFE3, one of the only such fusion oncoprotein co-factors identified in cancer biology. Specifically, VCP co-distributed with ASPSCR1-TFE3 across chromatin in association with enhancers genome-wide. VCP presence, its hexameric assembly, and its enzymatic function orchestrated the oncogenic transcriptional signature of ASPSCR1-TFE3, by facilitating assembly of higher-order chromatin conformation structures as demonstrated by HiChIP. Finally, ASPSCR1-TFE3 and VCP demonstrated co-dependence for cancer cell proliferation and tumorigenesis in vitro and in ASPS and RCC mouse models, underscoring VCP's potential as a novel therapeutic target.
- Published
- 2023
- Full Text
- View/download PDF
15. Loss of ATG4B and ATG4A results in two-stage cell cycle defects in pancreatic ductal adenocarcinoma cells.
- Author
-
Sathiyaseelan P, Chittaranjan S, Kalloger SE, Chan J, Go NE, Jardon MA, Ho CJ, Hui T, Xu J, Chow C, Gao D, Johnson FD, Lockwood WW, Morin GB, Renouf DJ, Schaeffer DF, and Gorski SM
- Subjects
- Humans, Autophagy-Related Proteins genetics, Autophagy-Related Proteins metabolism, Cysteine Endopeptidases genetics, Cysteine Endopeptidases metabolism, Autophagy genetics, Cell Line, Tumor, Cell Cycle genetics, Cell Proliferation genetics, Pancreatic Neoplasms genetics, Adenocarcinoma, Carcinoma, Pancreatic Ductal genetics, Carcinoma, Pancreatic Ductal metabolism
- Abstract
Pancreatic ductal adenocarcinoma (PDAC) exhibits elevated levels of autophagy, which promote tumor progression and treatment resistance. ATG4B is an autophagy-related cysteine protease under consideration as a potential therapeutic target, but it is largely unexplored in PDAC. Here, we investigated the clinical and functional relevance of ATG4B expression in PDAC. Using two PDAC patient cohorts, we found that low ATG4B mRNA or protein expression is associated with worse patient survival outcomes, poorly differentiated PDAC tumors and a lack of survival benefit from adjuvant chemotherapy. In PDAC cell lines, ATG4B knockout reduced proliferation, abolished processing of LC3B (also known as MAP1LC3B), and reduced GABARAP and GABARAPL1 levels, but increased ATG4A levels. ATG4B and ATG4A double knockout lines displayed a further reduction in proliferation, characterized by delays in G1-S phase transition and mitosis. Pro-LC3B accumulated aberrantly at the centrosome with a concomitant increase in centrosomal proteins PCM1 and CEP131, which was rescued by exogenous ATG4B. The two-stage cell cycle defects following ATG4B and ATG4A loss have important therapeutic implications for PDAC., Competing Interests: Competing interests The authors declare no competing or financial interests., (© 2023. Published by The Company of Biologists Ltd.)
- Published
- 2023
- Full Text
- View/download PDF
16. A MYCN-independent mechanism mediating secretome reprogramming and metastasis in MYCN -amplified neuroblastoma.
- Author
-
Zhang HF, Delaidelli A, Javed S, Turgu B, Morrison T, Hughes CS, Yang X, Pachva M, Lizardo MM, Singh G, Hoffmann J, Huang YZ, Patel K, Shraim R, Kung SHY, Morin GB, Aparicio S, Martinez D, Maris JM, Bosse KR, Williams KC, and Sorensen PH
- Subjects
- Humans, N-Myc Proto-Oncogene Protein genetics, Cell Survival, Secretome, Neuroblastoma genetics
- Abstract
MYCN amplification ( MNA ) is a defining feature of high-risk neuroblastoma (NB) and predicts poor prognosis. However, whether genes within or in close proximity to the MYCN amplicon also contribute to MNA
+ NB remains poorly understood. Here, we identify that GREB1 , a transcription factor encoding gene neighboring the MYCN locus, is frequently coexpressed with MYCN and promotes cell survival in MNA+ NB. GREB1 controls gene expression independently of MYCN, among which we uncover myosin 1B ( MYO1B ) as being highly expressed in MNA+ NB and, using a chick chorioallantoic membrane (CAM) model, as a crucial regulator of invasion and metastasis. Global secretome and proteome profiling further delineates MYO1B in regulating secretome reprogramming in MNA+ NB cells, and the cytokine MIF as an important pro-invasive and pro-metastatic mediator of MYO1B activity. Together, we have identified a putative GREB1-MYO1B-MIF axis as an unconventional mechanism promoting aggressive behavior in MNA+ NB and independently of MYCN.- Published
- 2023
- Full Text
- View/download PDF
17. Alanine supplementation exploits glutamine dependency induced by SMARCA4/2-loss.
- Author
-
Zhu X, Fu Z, Chen SY, Ong D, Aceto G, Ho R, Steinberger J, Monast A, Pilon V, Li E, Ta M, Ching K, Adams BN, Negri GL, Choiniere L, Fu L, Pavlakis K, Pirrotte P, Avizonis DZ, Trent J, Weissman BE, Klein Geltink RI, Morin GB, Park M, Huntsman DG, Foulkes WD, Wang Y, and Huang S
- Subjects
- Humans, Glucose Transporter Type 1, Adenosine Triphosphatases metabolism, Dietary Supplements, DNA Helicases metabolism, Nuclear Proteins genetics, Nuclear Proteins metabolism, Transcription Factors genetics, Transcription Factors metabolism, Glutamine, Neoplasms drug therapy, Neoplasms genetics
- Abstract
SMARCA4 (BRG1) and SMARCA2 (BRM) are the two paralogous ATPases of the SWI/SNF chromatin remodeling complexes frequently inactivated in cancers. Cells deficient in either ATPase have been shown to depend on the remaining counterpart for survival. Contrary to this paralog synthetic lethality, concomitant loss of SMARCA4/2 occurs in a subset of cancers associated with very poor outcomes. Here, we uncover that SMARCA4/2-loss represses expression of the glucose transporter GLUT1, causing reduced glucose uptake and glycolysis accompanied with increased dependency on oxidative phosphorylation (OXPHOS); adapting to this, these SMARCA4/2-deficient cells rely on elevated SLC38A2, an amino acid transporter, to increase glutamine import for fueling OXPHOS. Consequently, SMARCA4/2-deficient cells and tumors are highly sensitive to inhibitors targeting OXPHOS or glutamine metabolism. Furthermore, supplementation of alanine, also imported by SLC38A2, restricts glutamine uptake through competition and selectively induces death in SMARCA4/2-deficient cancer cells. At a clinically relevant dose, alanine supplementation synergizes with OXPHOS inhibition or conventional chemotherapy eliciting marked antitumor activity in patient-derived xenografts. Our findings reveal multiple druggable vulnerabilities of SMARCA4/2-loss exploiting a GLUT1/SLC38A2-mediated metabolic shift. Particularly, unlike dietary deprivation approaches, alanine supplementation can be readily applied to current regimens for better treatment of these aggressive cancers., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
18. Multi-Omic Analysis of CIC's Functional Networks Reveals Novel Interaction Partners and a Potential Role in Mitotic Fidelity.
- Author
-
Takemon Y, LeBlanc VG, Song J, Chan SY, Lee SD, Trinh DL, Ahmad ST, Brothers WR, Corbett RD, Gagliardi A, Moradian A, Cairncross JG, Yip S, Aparicio SAJR, Chan JA, Hughes CS, Morin GB, Gorski SM, Chittaranjan S, and Marra MA
- Abstract
CIC encodes a transcriptional repressor and MAPK signalling effector that is inactivated by loss-of-function mutations in several cancer types, consistent with a role as a tumour suppressor. Here, we used bioinformatic, genomic, and proteomic approaches to investigate CIC's interaction networks. We observed both previously identified and novel candidate interactions between CIC and SWI/SNF complex members, as well as novel interactions between CIC and cell cycle regulators and RNA processing factors. We found that CIC loss is associated with an increased frequency of mitotic defects in human cell lines and an in vivo mouse model and with dysregulated expression of mitotic regulators. We also observed aberrant splicing in CIC-deficient cell lines, predominantly at 3' and 5' untranslated regions of genes, including genes involved in MAPK signalling, DNA repair, and cell cycle regulation. Our study thus characterises the complexity of CIC's functional network and describes the effect of its loss on cell cycle regulation, mitotic integrity, and transcriptional splicing, thereby expanding our understanding of CIC's potential roles in cancer. In addition, our work exemplifies how multi-omic, network-based analyses can be used to uncover novel insights into the interconnected functions of pleiotropic genes/proteins across cellular contexts.
- Published
- 2023
- Full Text
- View/download PDF
19. Tandem mass tag-based thermal proteome profiling for the discovery of drug-protein interactions in cancer cells.
- Author
-
Johnson FD, Hughes CS, Liu A, Lockwood WW, and Morin GB
- Subjects
- Tandem Mass Spectrometry methods, Cell Line, Proteome analysis, Neoplasms drug therapy
- Abstract
Identification of effector targets is imperative to the characterization of the mechanisms of action of novel small molecules. Here, we describe steps to identify effector drug-protein interactions in lysates derived from cancer cell lines using a thermal proteome profiling (TPP) protocol. Building on existing TTP approaches, we detail the use of an in-solution trypsin digestion technique to streamline sample preparation, a nonparametric analysis to rank proteins for prioritization, and a follow-up strategy for identifying effector interactors. For complete details on the use and execution of this protocol, please refer to Johnson et al. (2022).
1 ., Competing Interests: Declaration of interests W.W.L. is a consultant of HyperBio Therapeutics., (Copyright © 2023 The Authors. Published by Elsevier Inc. All rights reserved.)- Published
- 2023
- Full Text
- View/download PDF
20. PeptideRanger: An R Package to Optimize Synthetic Peptide Selection for Mass Spectrometry Applications.
- Author
-
Riley RM, Spencer Miko SE, Morin RD, Morin GB, and Negri GL
- Subjects
- Mass Spectrometry methods, Proteins, Proteomics methods, Peptides analysis
- Abstract
Targeted and semitargeted mass spectrometry-based approaches are reliable methods to consistently detect and quantify low abundance proteins including proteins of clinical significance. Despite their potential, the development of targeted and semitargeted assays is time-consuming and often requires the purchase of costly libraries of synthetic peptides. To improve the efficiency of this rate-limiting step, we developed PeptideRanger, a tool to identify peptides from protein of interest with physiochemical properties that make them more likely to be suitable for mass spectrometry analysis. PeptideRanger is a flexible, extensively annotated, and intuitive R package that uses a random forest model trained on a diverse data set of thousands of MS experiments spanning a variety of sample types profiled with different chromatography setups and instruments. To support a variety of applications and to leverage rapidly growing public MS databases, PeptideRanger can readily be retrained with experiment-specific data sets and customized to prioritize and filter peptides based on selected properties.
- Published
- 2023
- Full Text
- View/download PDF
21. Human antibodies targeting ENPP1 as candidate therapeutics for cancers.
- Author
-
Chu X, Baek DS, Li W, Shyp T, Mooney B, Hines MG, Morin GB, Sorensen PH, and Dimitrov DS
- Subjects
- Humans, Female, Phosphoric Diester Hydrolases genetics, Phosphoric Diester Hydrolases metabolism, Immunoglobulin G, Pyrophosphatases genetics, Immunoconjugates, Breast Neoplasms
- Abstract
Ectonucleotide pyrophosphatase/phosphodiesterase 1 (ENPP1) is a type II transmembrane glycoprotein expressed in many tissues. High expression levels of ENPP1 have been observed in many cancer types such as lung cancer, ovarian cancer, and breast cancer. Such overexpression has been associated with poor prognosis in these diseases. Hence, ENPP1 is a potential target for immunotherapy across multiple cancers. Here, we isolated and characterized two high-affinity and specific anti-ENPP1 Fab antibody candidates, 17 and 3G12, from large phage-displayed human Fab libraries. After conversion to IgG1, the binding of both antibodies increased significantly due to avidity effects. Based on these antibodies, we generated antibody-drug conjugates (ADCs), IgG-based bispecific T-cell engagers (IbTEs), and CAR T-cells which all exhibited potent killing of ENPP1-expressing cells. Thus, these various antibody-derived modalities are promising therapeutic candidates for cancers expressing human ENPP1., Competing Interests: XC, D-SB, PS, and DD are co-inventors of a patent, filed on June 24 by the University of Pittsburgh US 63/367021, related to the antibodies described in this paper. Author DD is employed by Abound Bio. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2023 Chu, Baek, Li, Shyp, Mooney, Hines, Morin, Sorensen and Dimitrov.)
- Published
- 2023
- Full Text
- View/download PDF
22. The proteome of clear cell ovarian carcinoma.
- Author
-
Ji JX, Cochrane DR, Negri GL, Colborne S, Spencer Miko SE, Hoang LN, Farnell D, Tessier-Cloutier B, Huvila J, Thompson E, Leung S, Chiu D, Chow C, Ta M, Köbel M, Feil L, Anglesio M, Goode EL, Bolton K, Morin GB, and Huntsman DG
- Subjects
- Female, Humans, Carcinoma, Ovarian Epithelial pathology, Proteome, Proteomics, Adenocarcinoma, Clear Cell pathology, Ovarian Neoplasms metabolism
- Abstract
Clear cell ovarian carcinoma (CCOC) is the second most common subtype of epithelial ovarian carcinoma. Late-stage CCOC is not responsive to gold-standard chemotherapy and results in suboptimal outcomes for patients. In-depth molecular insight is urgently needed to stratify the disease and drive therapeutic development. We conducted global proteomics for 192 cases of CCOC and compared these with other epithelial ovarian carcinoma subtypes. Our results showed distinct proteomic differences in CCOC compared with other epithelial ovarian cancer subtypes including alterations in lipid and purine metabolism pathways. Furthermore, we report potential clinically significant proteomic subgroups within CCOC, suggesting the biologic plausibility of stratified treatment for this cancer. Taken together, our results provide a comprehensive understanding of the CCOC proteomic landscape to facilitate future understanding and research of this disease. © 2022 The Pathological Society of Great Britain and Ireland., (© 2022 The Pathological Society of Great Britain and Ireland.)
- Published
- 2022
- Full Text
- View/download PDF
23. Chloroquine treatment induces secretion of autophagy-related proteins and inclusion of Atg8-family proteins in distinct extracellular vesicle populations.
- Author
-
Xu J, Yang KC, Go NE, Colborne S, Ho CJ, Hosseini-Beheshti E, Lystad AH, Simonsen A, Guns ET, Morin GB, and Gorski SM
- Subjects
- Autophagy-Related Protein 8 Family metabolism, Autophagy-Related Proteins metabolism, Chloroquine pharmacology, Autophagy physiology, Apoptosis Regulatory Proteins metabolism, gamma-Aminobutyric Acid, Syntenins metabolism, Extracellular Vesicles metabolism
- Abstract
Chloroquine (CQ), a lysosomotropic agent, is commonly used to inhibit lysosomal degradation and macroautophagy/autophagy. Here we investigated the cell-extrinsic effects of CQ on secretion. We showed that lysosomal and autophagy inhibition by CQ altered the secretome, and induced the release of Atg8 orthologs and autophagy receptors. Atg8-family proteins, in particular, were secreted inside small extracellular vesicles (sEVs) in a lipidation-dependent manner. CQ treatment enhanced the release of Atg8-family proteins inside sEVs. Using full-length ATG16L1 and an ATG16L1 mutant that enables Atg8-family protein lipidation on double but not on single membranes, we demonstrated that LC3B is released in two distinct sEV populations: one enriched with SDCBP/Syntenin-1, CD63, and endosomal lipidated LC3B, and another that contains LC3B but is not enriched with SDCBP/Syntenin-1 or CD63, and which our data supports as originating from a double-membrane source. Our findings underscore the context-dependency of sEV heterogeneity and composition, and illustrate the integration of autophagy and sEV composition in response to lysosomal inhibition. Abbreviations: ACTB: actin beta; ANOVA: analysis of variance; ATG4B: autophagy related 4B cysteine peptidase; Atg8: autophagy related 8; ATG16L1: autophagy related 16 like 1; ATP5F1A/ATP5a: ATP synthase F1 subunit alpha; CALCOCO2: calcium binding and coiled-coil domain 2; CASP3: caspase 3; CASP7: caspase 7; CQ: chloroquine; CD9: CD9 molecule; CD63: CD63 molecule; DAPI: 4',6-diamidino-2-phenylindole; DQ-BSA: dye quenched-bovine serum albumin; ER: endoplasmic reticulum; ERN1/IRE1a: endoplasmic reticulum to nucleus signaling 1; EV: extracellular vesicles; FBS: fetal bovine serum; FDR: false discovery rate; GABARAP: GABA type A receptor-associated protein; GABARAPL2: GABA type A receptor associated protein like 2; GAPDH: glyceraldehyde-3-phosphate dehydrogenase; GFP: green fluorescent protein; GO: gene ontology; HCQ: hydroxychloroquine; HSP90AA1: heat shock protein 90 alpha family class A member 1; IP: immunoprecipitation; KO: knockout; LAMP2: lysosomal associated membrane protein 2; LIR: LC3-interacting region; LMNA: lamin A/C; MAP1LC3B/LC3B: microtubule associated protein 1 light chain 3 beta; MS: mass spectrometry; NBR1: NBR1 autophagy cargo receptor; NCOA4: nuclear receptor coactivator 4; NTA: nanoparticle tracking analysis; PE: phosphatidylethanolamine; PECA: probe-level expression change averaging; SDCBP/syntenin-1: syndecan binding protein; SD: standard deviation; SE: secreted; sEV: small extracellular vesicles; SQSTM1/p62: sequestosome 1; TAX1BP1: Tax1 binding protein 1; TEM: transmission electron microscopy; TMT: tandem-mass tag; TSG101: tumor susceptibility 101; ULK1: unc-51 like autophagy activating kinase 1; WC: whole cell.
- Published
- 2022
- Full Text
- View/download PDF
24. Integrative analysis of KRAS wildtype metastatic pancreatic ductal adenocarcinoma reveals mutation and expression-based similarities to cholangiocarcinoma.
- Author
-
Topham JT, Tsang ES, Karasinska JM, Metcalfe A, Ali H, Kalloger SE, Csizmok V, Williamson LM, Titmuss E, Nielsen K, Negri GL, Spencer Miko SE, Jang GH, Denroche RE, Wong HL, O'Kane GM, Moore RA, Mungall AJ, Loree JM, Notta F, Wilson JM, Bathe OF, Tang PA, Goodwin R, Morin GB, Knox JJ, Gallinger S, Laskin J, Marra MA, Jones SJM, Schaeffer DF, and Renouf DJ
- Subjects
- Bile Ducts, Intrahepatic, Humans, Mutation, Proto-Oncogene Proteins p21(ras) genetics, Transcription Factors genetics, Pancreatic Neoplasms, Adenocarcinoma pathology, Bile Duct Neoplasms genetics, Carcinoma, Pancreatic Ductal pathology, Cholangiocarcinoma genetics, Pancreatic Neoplasms pathology
- Abstract
Oncogenic KRAS mutations are absent in approximately 10% of patients with metastatic pancreatic ductal adenocarcinoma (mPDAC) and may represent a subgroup of mPDAC with therapeutic options beyond standard-of-care cytotoxic chemotherapy. While distinct gene fusions have been implicated in KRAS wildtype mPDAC, information regarding other types of mutations remain limited, and gene expression patterns associated with KRAS wildtype mPDAC have not been reported. Here, we leverage sequencing data from the PanGen trial to perform comprehensive characterization of the molecular landscape of KRAS wildtype mPDAC and reveal increased frequency of chr1q amplification encompassing transcription factors PROX1 and NR5A2. By leveraging data from colorectal adenocarcinoma and cholangiocarcinoma samples, we highlight similarities between cholangiocarcinoma and KRAS wildtype mPDAC involving both mutation and expression-based signatures and validate these findings using an independent dataset. These data further establish KRAS wildtype mPDAC as a unique molecular entity, with therapeutic opportunities extending beyond gene fusion events., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
25. Elucidating the importance and regulation of key enhancers for human MEIS1 expression.
- Author
-
Xiang P, Yang X, Escano L, Dhillon I, Schneider E, Clemans-Gibbon J, Wei W, Wong J, Wang SX, Tam D, Deng Y, Yung E, Morin GB, Hoodless PA, Hirst M, Karsan A, Kuchenbauer F, Humphries RK, and Rouhi A
- Subjects
- Humans, Neoplasm Proteins metabolism, Transcription Factors metabolism, Homeodomain Proteins chemistry, Leukemia, Myeloid, Acute genetics, Myeloid Ecotropic Viral Integration Site 1 Protein genetics
- Abstract
Myeloid ecotropic virus insertion site 1 (MEIS1) is essential for normal hematopoiesis and is a critical factor in the pathogenesis of a large subset of acute myeloid leukemia (AML). Despite the clinical relevance of MEIS1, its regulation is largely unknown. To understand the transcriptional regulatory mechanisms contributing to human MEIS1 expression, we created a knock-in green florescent protein (GFP) reporter system at the endogenous MEIS1 locus in a human AML cell line. Using this model, we have delineated and dissected a critical enhancer region of the MEIS1 locus for transcription factor (TF) binding through in silico prediction in combination with oligo pull-down, mass-spectrometry and knockout analysis leading to the identification of FLI1, an E-twenty-six (ETS) transcription factor, as an important regulator of MEIS1 transcription. We further show direct binding of FLI1 to the MEIS1 locus in human AML cell lines as well as enrichment of histone acetylation in MEIS1-high healthy and leukemic cells. We also observe a positive correlation between high FLI1 transcript levels and worse overall survival in AML patients. Our study expands the role of ETS factors in AML and our model constitutes a feasible tool for a more detailed understanding of transcriptional regulatory elements and their interactome., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
26. Proteomic analysis of archival breast cancer clinical specimens identifies biological subtypes with distinct survival outcomes.
- Author
-
Asleh K, Negri GL, Spencer Miko SE, Colborne S, Hughes CS, Wang XQ, Gao D, Gilks CB, Chia SKL, Nielsen TO, and Morin GB
- Subjects
- Breast pathology, Female, Gene Expression Profiling, Gene Expression Regulation, Neoplastic genetics, Humans, Proteomics, Treatment Outcome, Triple Negative Breast Neoplasms mortality, Biomarkers, Tumor metabolism, Proteome metabolism, Triple Negative Breast Neoplasms classification, Triple Negative Breast Neoplasms pathology
- Abstract
Despite advances in genomic classification of breast cancer, current clinical tests and treatment decisions are commonly based on protein level information. Formalin-fixed paraffin-embedded (FFPE) tissue specimens with extended clinical outcomes are widely available. Here, we perform comprehensive proteomic profiling of 300 FFPE breast cancer surgical specimens, 75 of each PAM50 subtype, from patients diagnosed in 2008-2013 (n = 178) and 1986-1992 (n = 122) with linked clinical outcomes. These two cohorts are analyzed separately, and we quantify 4214 proteins across all 300 samples. Within the aggressive PAM50-classified basal-like cases, proteomic profiling reveals two groups with one having characteristic immune hot expression features and highly favorable survival. Her2-Enriched cases separate into heterogeneous groups differing by extracellular matrix, lipid metabolism, and immune-response features. Within 88 triple-negative breast cancers, four proteomic clusters display features of basal-immune hot, basal-immune cold, mesenchymal, and luminal with disparate survival outcomes. Our proteomic analysis characterizes the heterogeneity of breast cancer in a clinically-applicable manner, identifies potential biomarkers and therapeutic targets, and provides a resource for clinical breast cancer classification., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
27. Characterization of a small molecule inhibitor of disulfide reductases that induces oxidative stress and lethality in lung cancer cells.
- Author
-
Johnson FD, Ferrarone J, Liu A, Brandstädter C, Munuganti R, Farnsworth DA, Lu D, Luu J, Sihota T, Jansen S, Nagelberg A, Shi R, Forcina GC, Zhang X, Cheng GSW, Spencer Miko SE, de Rappard-Yuswack G, Sorensen PH, Dixon SJ, Guha U, Becker K, Djaballah H, Somwar R, Varmus H, Morin GB, and Lockwood WW
- Subjects
- Glutathione metabolism, Humans, Lung metabolism, Lung pathology, Lung Neoplasms metabolism, Reactive Oxygen Species metabolism, Thioredoxins metabolism, Lung Neoplasms pathology, Oxidative Stress physiology, Oxidoreductases metabolism, Thioredoxin-Disulfide Reductase metabolism
- Abstract
Phenotype-based screening can identify small molecules that elicit a desired cellular response, but additional approaches are required to characterize their targets and mechanisms of action. Here, we show that a compound termed LCS3, which selectively impairs the growth of human lung adenocarcinoma (LUAD) cells, induces oxidative stress. To identify the target that mediates this effect, we use thermal proteome profiling (TPP) and uncover the disulfide reductases GSR and TXNRD1 as targets. We confirm through enzymatic assays that LCS3 inhibits disulfide reductase activity through a reversible, uncompetitive mechanism. Further, we demonstrate that LCS3-sensitive LUAD cells are sensitive to the synergistic inhibition of glutathione and thioredoxin pathways. Lastly, a genome-wide CRISPR knockout screen identifies NQO1 loss as a mechanism of LCS3 resistance. This work highlights the ability of TPP to uncover targets of small molecules identified by high-throughput screens and demonstrates the potential therapeutic utility of inhibiting disulfide reductases in LUAD., Competing Interests: Declaration of interests H.D., R.S., and H.V. are inventors on US patent #9562019 covering LCS3. W.W.L. is a consultant for HyperBio Therapeutics. R.S. has received research support from Helsinn Healthcare, Loxo Oncology, Merus, and Elevation Oncology, Inc., unrelated to the study. U.G. has a clinical trial agreement with AstraZeneca and research funding from AstraZeneca, Esanex, and Aurigene. U.G. is currently an employee of Bristol Myers Squibb., (Copyright © 2022. Published by Elsevier Inc.)
- Published
- 2022
- Full Text
- View/download PDF
28. De novo and cell line models of human mammary cell transformation reveal an essential role for Yb-1 in multiple stages of human breast cancer.
- Author
-
Lefort S, El-Naggar A, Tan S, Colborne S, Negri GL, Pellacani D, Hirst M, Gusterson B, Morin GB, Sorensen PH, and Eaves CJ
- Subjects
- Animals, Cell Line, Tumor, DNA Helicases metabolism, Female, Humans, Mice, Poly-ADP-Ribose Binding Proteins, RNA Helicases metabolism, RNA Recognition Motif Proteins, Transcription Factors metabolism, Y-Box-Binding Protein 1 genetics, Y-Box-Binding Protein 1 metabolism, Breast Neoplasms pathology
- Abstract
Breast cancer heterogeneity has made it challenging to identify mechanisms critical to the initial stages of their genesis in vivo. Here, we sought to interrogate the role of YB-1 in newly arising human breast cancers as well as in established cell lines. In a first series of experiments, we found that short-hairpin RNA-mediated knockdown of YB-1 in MDA-MB-231 cells blocked both their local tumour-forming and lung-colonising activity in immunodeficient mice. Conversely, upregulated expression of YB-1 enhanced the poor in vivo tumorigenicity of T47D cells. We then found that YB-1 knockdown also inhibits the initial generation in mice of invasive ductal carcinomas and ductal carcinomas in situ from freshly isolated human mammary cells transduced, respectively, with KRAS
G12D or myristoylated-AKT1. Interestingly, increased expression of HIF1α and G3BP1, two YB-1 translational targets and elements of a stress-adaptive programme, mirrored the levels of YB-1 in both transformed primary and established MDA-MB-231 breast cancer cells., (© 2021. The Author(s).)- Published
- 2022
- Full Text
- View/download PDF
29. Proteomic Screens for Suppressors of Anoikis Identify IL1RAP as a Promising Surface Target in Ewing Sarcoma.
- Author
-
Zhang HF, Hughes CS, Li W, He JZ, Surdez D, El-Naggar AM, Cheng H, Prudova A, Delaidelli A, Negri GL, Li X, Ørum-Madsen MS, Lizardo MM, Oo HZ, Colborne S, Shyp T, Scopim-Ribeiro R, Hammond CA, Dhez AC, Langman S, Lim JKM, Kung SHY, Li A, Steino A, Daugaard M, Parker SJ, Geltink RIK, Orentas RJ, Xu LY, Morin GB, Delattre O, Dimitrov DS, and Sorensen PH
- Subjects
- Adult, Cell Line, Tumor, Child, Humans, Proteomics, Receptors, Interleukin-1, Anoikis, Interleukin-1 Receptor Accessory Protein, Sarcoma, Ewing genetics, Sarcoma, Ewing metabolism, Sarcoma, Ewing pathology
- Abstract
Cancer cells must overcome anoikis (detachment-induced death) to successfully metastasize. Using proteomic screens, we found that distinct oncoproteins upregulate IL1 receptor accessory protein (IL1RAP) to suppress anoikis. IL1RAP is directly induced by oncogenic fusions of Ewing sarcoma, a highly metastatic childhood sarcoma. IL1RAP inactivation triggers anoikis and impedes metastatic dissemination of Ewing sarcoma cells. Mechanistically, IL1RAP binds the cell-surface system X
c - transporter to enhance exogenous cystine uptake, thereby replenishing cysteine and the glutathione antioxidant. Under cystine depletion, IL1RAP induces cystathionine gamma lyase (CTH) to activate the transsulfuration pathway for de novo cysteine synthesis. Therefore, IL1RAP maintains cyst(e)ine and glutathione pools, which are vital for redox homeostasis and anoikis resistance. IL1RAP is minimally expressed in pediatric and adult normal tissues, and human anti-IL1RAP antibodies induce potent antibody-dependent cellular cytotoxicity of Ewing sarcoma cells. Therefore, we define IL1RAP as a new cell-surface target in Ewing sarcoma, which is potentially exploitable for immunotherapy. SIGNIFICANCE: Here, we identify cell-surface protein IL1RAP as a key driver of metastasis in Ewing sarcoma, a highly aggressive childhood sarcoma. Minimal expression in pediatric and adult normal tissues nominates IL1RAP as a promising target for immunotherapy. See related commentary by Yoon and DeNicola, p. 2679 . This article is highlighted in the In This Issue feature, p. 2659 ., (©2021 The Authors; Published by the American Association for Cancer Research.)- Published
- 2021
- Full Text
- View/download PDF
30. Proteotranscriptomic classification and characterization of pancreatic neuroendocrine neoplasms.
- Author
-
Yang KC, Kalloger SE, Aird JJ, Lee MKC, Rushton C, Mungall KL, Mungall AJ, Gao D, Chow C, Xu J, Karasinska JM, Colborne S, Jones SJM, Schrader J, Morin RD, Loree JM, Marra MA, Renouf DJ, Morin GB, Schaeffer DF, and Gorski SM
- Subjects
- Cell Differentiation, Cell Proliferation, Co-Repressor Proteins genetics, Co-Repressor Proteins metabolism, Databases, Genetic, Female, Gene Expression Profiling, Gene Expression Regulation, Neoplastic, Genetic Heterogeneity, Humans, Male, Molecular Chaperones genetics, Molecular Chaperones metabolism, Mutation, Neuroendocrine Tumors pathology, Neuroendocrine Tumors therapy, Pancreatic Neoplasms pathology, Pancreatic Neoplasms therapy, Prognosis, Proteomics, Proto-Oncogene Proteins genetics, Proto-Oncogene Proteins metabolism, Signal Transduction, Transcriptional Coactivator with PDZ-Binding Motif Proteins genetics, Transcriptional Coactivator with PDZ-Binding Motif Proteins metabolism, YAP-Signaling Proteins genetics, YAP-Signaling Proteins metabolism, Biomarkers, Tumor genetics, Biomarkers, Tumor metabolism, Neuroendocrine Tumors genetics, Neuroendocrine Tumors metabolism, Pancreatic Neoplasms genetics, Pancreatic Neoplasms metabolism, Proteome, Transcriptome
- Abstract
Pancreatic neuroendocrine neoplasms (PNENs) are biologically and clinically heterogeneous. Here, we use a multi-omics approach to uncover the molecular factors underlying this heterogeneity. Transcriptomic analysis of 84 PNEN specimens, drawn from two cohorts, is substantiated with proteomic profiling and identifies four subgroups: Proliferative, PDX1-high, Alpha cell-like and Stromal/Mesenchymal. The Proliferative subgroup, consisting of both well- and poorly differentiated specimens, is associated with inferior overall survival probability. The PDX1-high and Alpha cell-like subgroups partially resemble previously described subtypes, and we further uncover distinctive metabolism-related features in the Alpha cell-like subgroup. The Stromal/Mesenchymal subgroup exhibits molecular characteristics of YAP1/WWTR1(TAZ) activation suggestive of Hippo signaling pathway involvement in PNENs. Whole-exome sequencing reveals subgroup-enriched mutational differences, supported by activity inference analysis, and identifies hypermorphic proto-oncogene variants in 14.3% of sequenced PNENs. Our study reveals differences in cellular signaling axes that provide potential directions for PNEN patient stratification and treatment strategies., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2021 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
31. Protein feature analysis of heat shock induced ubiquitination sites reveals preferential modification site localization.
- Author
-
Kuechler ER, Rose A, Bolten M, Madero A, Kammoonah S, Colborne S, Gsponer J, Morin GB, and Mayor T
- Subjects
- Animals, Humans, Proteasome Endopeptidase Complex metabolism, Protein Folding, Saccharomyces cerevisiae metabolism, Ubiquitination, Heat-Shock Response, Ubiquitin metabolism
- Abstract
Protein aggregation is indicative of failing protein quality control systems. These systems are responsible for the refolding or degradation of aberrant and misfolded proteins. Heat stress can cause proteins to misfold, triggering cellular responses including a marked increase in the ubiquitination of proteins. This response has been characterized in yeast, however more studies are needed within mammalian cells. Herein, we examine proteins that become ubiquitinated during heat shock in human tissue culture cells using diGly enrichment coupled with mass spectrometry. A majority of these proteins are localized in the nucleus or cytosol. Proteins which are conjugated under stress display longer sequence lengths, more interaction partners, and more hydrophobic patches than controls but do not show lower melting temperatures. Furthermore, heat-induced conjugation sites occur less frequently in disordered regions and are closer to hydrophobic patches than other ubiquitination sites; perhaps providing novel insight into the molecular mechanism mediating this response. Nuclear and cytosolic pools of modified proteins appear to have different protein features. Using a pulse-SILAC approach, we found that both long-lived and newly-synthesized proteins are conjugated under stress. Modified long-lived proteins are predominately nuclear and were distinct from newly-synthesized proteins, indicating that different pathways may mediate the heat-induced increase of polyubiquitination. SIGNIFICANCE: The maintenance of protein homeostasis requires a balance of protein synthesis, folding, and degradation. Under stress conditions, the cell must rapidly adapt by increasing its folding capacity to eliminate aberrant proteins. A major pathway for proteolysis is mediated by the ubiquitin proteasome system. While increased ubiquitination after heat stress was observed over 30 years ago, it remains unclear which proteins are conjugated during heat shock in mammalian cells and by what means this conjugation occurs. In this study, we combined SILAC-based mass spectrometry with computational analyses to reveal features associated to proteins ubiquitinated while under heat shock. Interestingly, we found that conjugation sites induced by the stress are less often located within disordered regions and more often located near hydrophobic patches. Our study showcases how proteomics can reveal distinct feature associated to a cohort of proteins that are modified post translationally and how the ubiquitin conjugation sites are preferably selected in these conditions. Our work opens a new path for delineating the molecular mechanisms leading to the heat stress response and the regulation of protein homeostasis., (Copyright © 2021 Elsevier B.V. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
32. Multiomics Characterization of Low-Grade Serous Ovarian Carcinoma Identifies Potential Biomarkers of MEK Inhibitor Sensitivity and Therapeutic Vulnerability.
- Author
-
Shrestha R, Llaurado Fernandez M, Dawson A, Hoenisch J, Volik S, Lin YY, Anderson S, Kim H, Haegert AM, Colborne S, Wong NKY, McConeghy B, Bell RH, Brahmbhatt S, Lee CH, DiMattia GE, Le Bihan S, Morin GB, Collins CC, and Carey MS
- Subjects
- Antineoplastic Combined Chemotherapy Protocols therapeutic use, Biomarkers, Tumor genetics, Biomarkers, Tumor isolation & purification, Biomarkers, Tumor metabolism, Cell Line, Tumor, Cohort Studies, Cystadenocarcinoma, Serous genetics, Cystadenocarcinoma, Serous metabolism, Cystadenocarcinoma, Serous pathology, Drug Resistance, Neoplasm genetics, Female, Genomics methods, Humans, Metabolomics methods, Neoplasm Grading, Ovarian Neoplasms genetics, Ovarian Neoplasms metabolism, Ovarian Neoplasms pathology, Proteomics methods, Systems Integration, Biomarkers, Pharmacological analysis, Cystadenocarcinoma, Serous drug therapy, Mitogen-Activated Protein Kinase Kinases antagonists & inhibitors, Ovarian Neoplasms drug therapy, Protein Kinase Inhibitors therapeutic use
- Abstract
Low-grade serous ovarian carcinoma (LGSOC) is a rare tumor subtype with high case fatality rates in patients with metastatic disease. There is a pressing need to develop effective treatments using newly available preclinical models for therapeutic discovery and drug evaluation. Here, we use multiomics integration of whole-exome sequencing, RNA sequencing, and mass spectrometry-based proteomics on 14 LGSOC cell lines to elucidate novel biomarkers and therapeutic vulnerabilities. Comparison of LGSOC cell line data with LGSOC tumor data enabled predictive biomarker identification of MEK inhibitor (MEKi) efficacy, with KRAS mutations found exclusively in MEKi-sensitive cell lines and NRAS mutations found mostly in MEKi-resistant cell lines. Distinct patterns of Catalogue of Somatic Mutations in Cancer mutational signatures were identified in MEKi-sensitive and MEKi-resistant cell lines. Deletions of CDKN2A/B and MTAP genes were more frequent in cell lines than tumor samples and possibly represent key driver events in the absence of KRAS/NRAS/BRAF mutations. These LGSOC cell lines were representative models of the molecular aberrations found in LGSOC tumors. For prediction of in vitro MEKi efficacy, proteomic data provided better discrimination than gene expression data. Condensin, minichromosome maintenance, and replication factor C protein complexes were identified as potential treatment targets in MEKi-resistant cell lines. This study suggests that CDKN2A/B or MTAP deficiency may be exploited using synthetically lethal treatment strategies, highlighting the importance of using proteomic data as a tool for molecular drug prediction. Multiomics approaches are crucial to improving our understanding of the molecular underpinnings of LGSOC and applying this information to develop new therapies. SIGNIFICANCE: These findings highlight the utility of global multiomics to characterize LGSOC cell lines as research models, to determine biomarkers of MEKi resistance, and to identify potential novel therapeutic targets., (©2021 American Association for Cancer Research.)
- Published
- 2021
- Full Text
- View/download PDF
33. Whole-proteome analysis of mesonephric-derived cancers describes new potential biomarkers.
- Author
-
Gibbard E, Cochrane DR, Pors J, Negri GL, Colborne S, Cheng AS, Chow C, Farnell D, Tessier-Cloutier B, McAlpine JN, Morin GB, Schmidt D, Kommoss S, Kommoss F, Keul J, Gilks B, Huntsman DG, and Hoang L
- Subjects
- Female, Humans, Proteomics methods, Biomarkers, Tumor analysis, Glutathione Transferase analysis, Histocompatibility Antigens analysis, Histone-Lysine N-Methyltransferase analysis, Mesonephroma diagnosis, Peptide Elongation Factor 1 analysis
- Abstract
Mesonephric carcinomas (MEs) and female adnexal tumors of probable Wolffian origin (FATWO) are derived from embryologic remnants of Wolffian/mesonephric ducts. Mesonephric-like carcinomas (MLCs) show identical morphology to ME of the cervix but occur in the uterus and ovary without convincing mesonephric remnants. ME, MLC, and FATWO are challenging to diagnose due to their morphologic similarities to Müllerian/paramesonephric tumors, contributing to a lack of evidence-based and tumor-specific treatments. We performed whole-proteomic analysis on 9 ME/MLC and 56 endometrial carcinomas (ECs) to identify potential diagnostic biomarkers. Although there were no convincing differences between ME and MLC, 543 proteins showed increased expression in ME/MLC relative to EC. From these proteins, euchromatic histone lysine methyltransferase 2 (EHMT2), glutathione S-transferase Mu 3 (GSTM3), eukaryotic translation elongation factor 1 alpha 2 (EEF1A2), and glycogen synthase kinase 3 beta were identified as putative biomarkers. Immunohistochemistry was performed on these candidates and GATA3 in 14 ME/MLC, 8 FATWO, 155 EC, and normal tissues. Of the candidates, only GATA3 and EHMT2 were highly expressed in mesonephric remnants and mesonephric-derived male tissues. GATA3 had the highest sensitivity and specificity for ME/MLC versus EC (93% and 99%) but was absent in FATWO. EHMT2 was 100% sensitive for ME/MLC & FATWO but was not specific (65%). Similarly, EEF1A2 was reasonably sensitive to ME/MLC (92%) and FATWO (88%) but was the least specific (38%). GSTM3 performed intermediately (sensitivity for ME/MLC and FATWO: 83% and 38%, respectively; specificity 67%). Although GATA3 remained the best diagnostic biomarker for ME/MLC, we have identified EHMT2, EEF1A2, and GSTM3 as proteins of interest in these cancers. FATWO's cell of origin is uncertain and remains an area for future research., (Crown Copyright © 2020. Published by Elsevier Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
34. Re-expression of SMARCA4/BRG1 in small cell carcinoma of ovary, hypercalcemic type (SCCOHT) promotes an epithelial-like gene signature through an AP-1-dependent mechanism.
- Author
-
Orlando KA, Douglas AK, Abudu A, Wang Y, Tessier-Cloutier B, Su W, Peters A, Sherman LS, Moore R, Nguyen V, Negri GL, Colborne S, Morin GB, Kommoss F, Lang JD, Hendricks WP, Raupach EA, Pirrotte P, Huntsman DG, Trent JM, Parker JS, Raab JR, and Weissman BE
- Subjects
- Biomarkers, Tumor analysis, Carcinoma, Small Cell genetics, Cell Line, Tumor, Cell Transformation, Neoplastic genetics, DNA Helicases metabolism, Female, Humans, Hypercalcemia genetics, Mutation genetics, Nuclear Proteins metabolism, Ovarian Neoplasms pathology, Ovary metabolism, Ovary pathology, Transcription Factor AP-1 genetics, Transcription Factors metabolism, Carcinoma, Small Cell pathology, DNA Helicases genetics, Hypercalcemia pathology, Nuclear Proteins genetics, Ovarian Neoplasms metabolism, Transcription Factor AP-1 metabolism, Transcription Factors genetics
- Abstract
Small cell carcinoma of the ovary, hypercalcemic type (SCCOHT) is a rare and aggressive form of ovarian cancer. SCCOHT tumors have inactivating mutations in SMARCA4 (BRG1), one of the two mutually exclusive ATPases of the SWI/SNF chromatin remodeling complex. To address the role that BRG1 loss plays in SCCOHT tumorigenesis, we performed integrative multi-omic analyses in SCCOHT cell lines +/- BRG1 reexpression. BRG1 reexpression induced a gene and protein signature similar to an epithelial cell and gained chromatin accessibility sites correlated with other epithelial originating TCGA tumors. Gained chromatin accessibility and BRG1 recruited sites were strongly enriched for transcription-factor-binding motifs of AP-1 family members. Furthermore, AP-1 motifs were enriched at the promoters of highly upregulated epithelial genes. Using a dominant-negative AP-1 cell line, we found that both AP-1 DNA-binding activity and BRG1 reexpression are necessary for the gene and protein expression of epithelial genes. Our study demonstrates that BRG1 reexpression drives an epithelial-like gene and protein signature in SCCOHT cells that depends upon by AP-1 activity., Competing Interests: KO, AD, AA, YW, BT, WS, AP, LS, RM, VN, GN, SC, GM, FK, JL, WH, ER, PP, DH, JT, JP, JR, BW No competing interests declared, (© 2020, Orlando et al.)
- Published
- 2020
- Full Text
- View/download PDF
35. Dynamic pH barrage junction focusing of amino acids, peptides, and digested monoclonal antibodies in capillary electrophoresis-mass spectrometry.
- Author
-
Wang L, Cheng J, McNutt JE, Morin GB, and Chen DDY
- Subjects
- Amino Acids chemistry, Antibodies, Monoclonal chemistry, Electrophoresis, Capillary instrumentation, Equipment Design, Hydrogen-Ion Concentration, Peptides chemistry, Tandem Mass Spectrometry instrumentation, Trypsin, Amino Acids analysis, Antibodies, Monoclonal analysis, Electrophoresis, Capillary methods, Peptides analysis, Tandem Mass Spectrometry methods
- Abstract
Dynamic pH barrage junction focusing in CE enables effective signal enhancement, quantitative capture efficiencies, and straightforward optimization. The method is a technical variant of dynamic pH junction focusing. CE separation with dynamic pH barrage junction focusing is compatible with both optical and mass spectrometric detection. We developed a CE-MS/MS method using hydrophilic polyethyleneimine-coated capillaries and validated it for the qualitative analysis of amino acids, peptides, and tryptic peptides of digested monoclonal antibodies. The S/N of extracted ion electropherograms of zwitterionic analytes were enhanced by approximately two orders of magnitude with a tradeoff of a shortened separation window. Online focusing improved the MS signal intensity of a diluted antibody digest, enabling more precursor ions to be analyzed with subsequent tandem mass spectrometric identification. It also broadened the concentration range of protein digest samples for which adequate sequence coverage data can be obtained. With only 0.9 ng of digested infliximab sample loaded into the capillary, 76% and 100% sequence coverage was realized for antibody heavy and light chains, respectively, after online focusing. Full coverage was achieved with 9 ng of injected digest., (© 2020 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim.)
- Published
- 2020
- Full Text
- View/download PDF
36. The Pathognomonic FOXL2 C134W Mutation Alters DNA-Binding Specificity.
- Author
-
Carles A, Trigo-Gonzalez G, Cao Q, Cheng SG, Moksa M, Bilenky M, Huntsman DG, Morin GB, and Hirst M
- Subjects
- Cell Line, Tumor, Female, Granulosa Cell Tumor metabolism, Humans, Mutation, Missense, Point Mutation, Protein Binding, DNA metabolism, Forkhead Box Protein L2 genetics, Forkhead Box Protein L2 metabolism, Gene Expression Regulation, Neoplastic genetics, Granulosa Cell Tumor genetics
- Abstract
The somatic missense point mutation c.402C>G (p.C134W) in the FOXL2 transcription factor is pathognomonic for adult-type granulosa cell tumors (AGCT) and a diagnostic marker for this tumor type. However, the molecular consequences of this mutation and its contribution to the mechanisms of AGCT pathogenesis remain unclear. To explore these mechanisms, we engineered V5-FOXL2
WT - and V5-FOXL2C134W -inducible isogenic cell lines and performed chromatin immunoprecipitation sequencing and transcriptome profiling. FOXL2C134W associated with the majority of the FOXL2 wild-type DNA elements as well as a large collection of unique elements genome wide. This model enabled confirmation of altered DNA-binding specificity for FOXL2C134W and identification of unique targets of FOXL2C134W including SLC35F2 , whose expression increased sensitivity to YM155. Our results suggest FOXL2C134W drives AGCT by altering the binding affinity of FOXL2-containing complexes to engage an oncogenic transcriptional program. SIGNIFICANCE: A mechanistic understanding of FOXL2C134W -induced regulatory state alterations drives discovery of a rationally designed therapeutic strategy., (©2020 American Association for Cancer Research.)- Published
- 2020
- Full Text
- View/download PDF
37. Arginine Depletion Therapy with ADI-PEG20 Limits Tumor Growth in Argininosuccinate Synthase-Deficient Ovarian Cancer, Including Small-Cell Carcinoma of the Ovary, Hypercalcemic Type.
- Author
-
Ji JX, Cochrane DR, Tessier-Cloutier B, Chen SY, Ho G, Pathak KV, Alcazar IN, Farnell D, Leung S, Cheng A, Chow C, Colborne S, Negri GL, Kommoss F, Karnezis A, Morin GB, McAlpine JN, Gilks CB, Weissman BE, Trent JM, Hoang L, Pirrotte P, Wang Y, and Huntsman DG
- Subjects
- Animals, Arginine antagonists & inhibitors, Arginine genetics, Argininosuccinate Synthase deficiency, Carcinoma, Small Cell genetics, Carcinoma, Small Cell pathology, Cell Line, Tumor, Cell Proliferation drug effects, Female, Humans, Mice, Ovarian Neoplasms genetics, Ovarian Neoplasms pathology, Ovary metabolism, Ovary pathology, Parathyroid Hormone-Related Protein genetics, Parathyroid Hormone-Related Protein immunology, Proteomics, Xenograft Model Antitumor Assays, Argininosuccinate Synthase genetics, Carcinoma, Small Cell drug therapy, Hydrolases pharmacology, Ovarian Neoplasms drug therapy, Polyethylene Glycols pharmacology
- Abstract
Purpose: Many rare ovarian cancer subtypes, such as small-cell carcinoma of the ovary, hypercalcemic type (SCCOHT), have poor prognosis due to their aggressive nature and resistance to standard platinum- and taxane-based chemotherapy. The development of effective therapeutics has been hindered by the rarity of such tumors. We sought to identify targetable vulnerabilities in rare ovarian cancer subtypes., Experimental Design: We compared the global proteomic landscape of six cases each of endometrioid ovarian cancer (ENOC), clear cell ovarian cancer (CCOC), and SCCOHT to the most common subtype, high-grade serous ovarian cancer (HGSC), to identify potential therapeutic targets. IHC of tissue microarrays was used as validation of arginosuccinate synthase (ASS1) deficiency. The efficacy of arginine-depriving therapeutic ADI-PEG20 was assessed in vitro using cell lines and patient-derived xenograft mouse models representing SCCOHT., Results: Global proteomic analysis identified low ASS1 expression in ENOC, CCOC, and SCCOHT compared with HGSC. Low ASS1 levels were validated through IHC in large patient cohorts. The lowest levels of ASS1 were observed in SCCOHT, where ASS1 was absent in 12 of 31 cases, and expressed in less than 5% of the tumor cells in 9 of 31 cases. ASS1-deficient ovarian cancer cells were sensitive to ADI-PEG20 treatment regardless of subtype in vitro . Furthermore, in two cell line mouse xenograft models and one patient-derived mouse xenograft model of SCCOHT, once-a-week treatment with ADI-PEG20 (30 mg/kg and 15 mg/kg) inhibited tumor growth in vivo ., Conclusions: Preclinical in vitro and in vivo studies identified ADI-PEG20 as a potential therapy for patients with rare ovarian cancers, including SCCOHT., (©2020 American Association for Cancer Research.)
- Published
- 2020
- Full Text
- View/download PDF
38. Complementary Methods for de Novo Monoclonal Antibody Sequencing to Achieve Complete Sequence Coverage.
- Author
-
Cheng J, Wang L, Rive CM, Holt RA, Morin GB, and Chen DDY
- Subjects
- Chromatography, Liquid, Electrophoresis, Capillary, Sequence Analysis, Antibodies, Monoclonal, Tandem Mass Spectrometry
- Abstract
Mass spectrometry is a powerful tool for de novo sequencing of novel proteins. Recent efforts in this area have mainly focused on liquid chromatography coupled with tandem mass spectrometry (LC-MS/MS). Here, we present an alternative method, capillary electrophoresis tandem mass spectrometry (CE-MS/MS), for sequencing novel monoclonal antibodies. Using less than 200 ng in total of tryptic digest sample in a triplicated measurement, CE-MS/MS with pH-mediated focusing successfully sequenced mAb infliximab with 100% sequence coverage and 100% accuracy for the light chain and 96% coverage and 93% accuracy for the heavy chain. It was also demonstrated that CE-MS/MS gives comparable results, and in some cases, even better results, as compared to LC-MS/MS when used as a standalone technique. A combined workflow using both CE-MS/MS and LC-MS/MS was also used to sequence a novel antibody, anti-CD-176, resulting in the first proposed sequence for this mAb.
- Published
- 2020
- Full Text
- View/download PDF
39. Proteomic analysis of transitional cell carcinoma-like variant of tubo-ovarian high-grade serous carcinoma.
- Author
-
Tessier-Cloutier B, Cochrane DR, Karnezis AN, Colborne S, Magrill J, Talhouk A, Zhang J, Leung S, Hughes CS, Piskorz A, Cheng AS, Greening K, du Bois A, Pfisterer J, Soslow RA, Kommoss S, Brenton JD, Morin GB, Gilks CB, Huntsman DG, and Kommoss F
- Subjects
- Female, Humans, Proteomics methods, Biomarkers, Tumor analysis, Carcinoma, Transitional Cell diagnosis, Cystadenocarcinoma, Serous diagnosis, Fallopian Tube Neoplasms diagnosis, Ovarian Neoplasms diagnosis
- Abstract
The current World Health Organization classification does not distinguish transitional cell carcinoma of the ovary (TCC) from conventional tubo-ovarian high-grade serous carcinoma (HGSC), despite evidence suggesting improved prognosis for patients with TCC; instead, it is considered a morphologic variant of HGSC. The immunohistochemical (IHC) markers applied to date do not distinguish between TCC and HGSC. Therefore, we sought to compare the proteomic profiles of TCC and conventional HGSC to identify proteins enriched in TCC. Prognostic biomarkers in HGSC have proven to be elusive, and our aim was to identify biomarkers of TCC as a way of reliably and reproducibly identifying patients with a favorable prognosis and better response to chemotherapy compared with those with conventional HGSC. Quantitative global proteome analysis was performed on archival material of 12 cases of TCC and 16 cases of HGSC using SP3 (single-pot, solid phase-enhanced, sample preparation)-Clinical Tissue Proteomics, a recently described protocol for full-proteome analysis from formalin-fixed paraffin-embedded tissues. We identified 430 proteins that were significantly enriched in TCC over HGSC. Unsupervised co-clustering perfectly distinguished TCC from HGSC based on protein expression. Pathway analysis showed that proteins associated with cell death, necrosis, and apoptosis were highly expressed in TCCs, whereas proteins associated with DNA homologous recombination, cell mitosis, proliferation and survival, and cell cycle progression pathways had reduced expression. From the proteomic analysis, three potential biomarkers for TCC were identified, claudin-4 (CLDN4), ubiquitin carboxyl-terminal esterase L1 (UCHL1), and minichromosome maintenance protein 7 (MCM7), and tested by IHC analysis on tissue microarrays. In agreement with the proteomic analysis, IHC expression of those proteins was stronger in TCC than in HGSC (p < 0.0001). Using global proteomic analysis, we are able to distinguish TCC from conventional HGSC. Follow-up studies will be necessary to confirm that these molecular and morphologic differences are clinically significant., (Copyright © 2020. Published by Elsevier Inc.)
- Published
- 2020
- Full Text
- View/download PDF
40. Loss of m 1 acp 3 Ψ Ribosomal RNA Modification Is a Major Feature of Cancer.
- Author
-
Babaian A, Rothe K, Girodat D, Minia I, Djondovic S, Milek M, Spencer Miko SE, Wieden HJ, Landthaler M, Morin GB, and Mager DL
- Subjects
- Base Sequence genetics, Humans, Nucleic Acid Conformation, Pseudouridine genetics, Neoplasms genetics, Oncogenes genetics, RNA, Ribosomal metabolism, Ribosomal Proteins metabolism, Ribosomes metabolism
- Abstract
The ribosome is an RNA-protein complex that is essential for translation in all domains of life. The structural and catalytic core of the ribosome is its ribosomal RNA (rRNA). While mutations in ribosomal protein (RP) genes are known drivers of oncogenesis, oncogenic rRNA variants have remained elusive. We identify a cancer-specific single-nucleotide variation in 18S rRNA at nucleotide 1248.U in up to 45.9% of patients with colorectal carcinoma (CRC) and present across >22 cancer types. This is the site of a unique hyper-modified base, 1-methyl-3-α-amino-α-carboxyl-propyl pseudouridine (m
1 acp3 Ψ), a >1-billion-years-conserved RNA modification at the peptidyl decoding site of the ribosome. A subset of CRC tumors we call hypo-m1 acp3 Ψ shows sub-stoichiometric m1 acp3 Ψ modification, unlike normal control tissues. An m1 acp3 Ψ knockout model and hypo-m1 acp3 Ψ patient tumors share a translational signature characterized by highly abundant ribosomal proteins. Thus, m1 acp3 Ψ-deficient rRNA forms an uncharacterized class of "onco-ribosome" which may serve as a chemotherapeutic target for treating cancer patients., Competing Interests: Declaration of Interests The authors declare no competing interests., (Copyright © 2020 The Author(s). Published by Elsevier Inc. All rights reserved.)- Published
- 2020
- Full Text
- View/download PDF
41. Bottom-up proteomics of envelope proteins extracted from spinach chloroplast via high organic content CE-MS.
- Author
-
Cheng J, Morin GB, and Chen DDY
- Subjects
- Chloroplasts chemistry, Hydrophobic and Hydrophilic Interactions, Plant Leaves chemistry, Proteome analysis, Proteome chemistry, Chloroplast Proteins analysis, Chloroplast Proteins chemistry, Chloroplast Proteins isolation & purification, Electrophoresis, Capillary methods, Proteomics methods, Spinacia oleracea chemistry, Tandem Mass Spectrometry methods
- Abstract
A high organic content CE-MS/MS (HOCE-MS/MS) method was developed for the proteomic analysis of envelope proteins extracted from spinach leaves. Separation was performed in a 1-m long hydroxypropyl cellulose coated capillary, using 8% (v/v) formic acid in 70% (v/v) methanol and 22% water as the BGE. A flow-through microvial interface was used to couple the CE system with an Orbitrap Fusion Lumos mass spectrometer, and field-amplified sample stacking was used to improve the concentration sensitivity. Using this optimized method, 3579 peptides and 1141 proteins were identified using the Proteome Discoverer software with a 1% false discovery rate at the protein level. Relative to conventional aqueous CE, HOCE-MS did a better job of discovering hydrophobic peptides and provided more peptide and protein identifications. Relative to nano-LC-MS, it achieved comparable peptide and protein identification performance and detected peptides not identified by LC-MS: of the full set of peptides identified using the two techniques, 19% were identified only using HOCE-MS. It also outperformed nano-LC-MS with respect to the detection of low molecular weight peptides., (© 2020 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim.)
- Published
- 2020
- Full Text
- View/download PDF
42. Class I HDAC inhibitors enhance YB-1 acetylation and oxidative stress to block sarcoma metastasis.
- Author
-
El-Naggar AM, Somasekharan SP, Wang Y, Cheng H, Negri GL, Pan M, Wang XQ, Delaidelli A, Rafn B, Cran J, Zhang F, Zhang H, Colborne S, Gleave M, Mandinova A, Kedersha N, Hughes CS, Surdez D, Delattre O, Wang Y, Huntsman DG, Morin GB, and Sorensen PH
- Subjects
- Acetylation, Animals, Bone Neoplasms metabolism, Bone Neoplasms pathology, Cell Line, Tumor, Cells, Cultured, Humans, Hypoxia-Inducible Factor 1, alpha Subunit metabolism, Mice, NF-E2-Related Factor 2 metabolism, Neoplasm Metastasis, Oxidative Stress, Sarcoma, Ewing metabolism, Sarcoma, Ewing pathology, Antineoplastic Agents therapeutic use, Benzamides therapeutic use, Bone Neoplasms drug therapy, Histone Deacetylase Inhibitors therapeutic use, Pyridines therapeutic use, Sarcoma, Ewing drug therapy, Transcription Factors metabolism
- Abstract
Outcomes for metastatic Ewing sarcoma and osteosarcoma are dismal and have not changed for decades. Oxidative stress attenuates melanoma metastasis, and melanoma cells must reduce oxidative stress to metastasize. We explored this in sarcomas by screening for oxidative stress sensitizers, which identified the class I HDAC inhibitor MS-275 as enhancing vulnerability to reactive oxygen species (ROS) in sarcoma cells. Mechanistically, MS-275 inhibits YB-1 deacetylation, decreasing its binding to 5'-UTRs of NFE2L2 encoding the antioxidant factor NRF2, thereby reducing NFE2L2 translation and synthesis of NRF2 to increase cellular ROS. By global acetylomics, MS-275 promotes rapid acetylation of the YB-1 RNA-binding protein at lysine-81, blocking binding and translational activation of NFE2L2, as well as known YB-1 mRNA targets, HIF1A, and the stress granule nucleator, G3BP1. MS-275 dramatically reduces sarcoma metastasis in vivo, but an MS-275-resistant YB-1K81-to-alanine mutant restores metastatic capacity and NRF2, HIF1α, and G3BP1 synthesis in MS-275-treated mice. These studies describe a novel function for MS-275 through enhanced YB-1 acetylation, thus inhibiting YB-1 translational control of key cytoprotective factors and its pro-metastatic activity., (© 2019 The Authors. Published under the terms of the CC BY 4.0 license.)
- Published
- 2019
- Full Text
- View/download PDF
43. The SNAP25 Interactome in Ventromedial Caudate in Schizophrenia Includes the Mitochondrial Protein ARF1.
- Author
-
Ramos-Miguel A, Barakauskas V, Alamri J, Miyauchi M, Barr AM, Beasley CL, Rosoklija G, Mann JJ, Dwork AJ, Moradian A, Morin GB, and Honer WG
- Subjects
- Adult, Aged, Caudate Nucleus physiopathology, Female, Humans, Male, Middle Aged, ADP-Ribosylation Factor 1 metabolism, Caudate Nucleus metabolism, Schizophrenia metabolism, Schizophrenia physiopathology, Synaptosomal-Associated Protein 25 metabolism
- Abstract
Abnormalities of SNAP25 (synaptosome-associated protein 25) amount and protein-protein interactions occur in schizophrenia, and may contribute to abnormalities of neurotransmitter release in patients. However, presynaptic terminal function depends on multiple subcellular mechanisms, including energy provided by mitochondria. To explore the SNAP25 interactome in schizophrenia, we immunoprecipitated SNAP25 along with interacting proteins from the ventromedial caudate of 15 cases of schizophrenia and 13 controls. Proteins were identified with mass spectrometry-based proteomics. As well as 15 SNARE- (soluble N-ethylmaleimide-sensitive factor attachment protein receptor) associated proteins, we identified 17 mitochondria-associated and four other proteins. The mitochondrial small GTPase ARF1 (ADP-ribosylation factor 1) was identified in eight schizophrenia SNAP25 immunoprecipitates and none from controls (P = 0.004). Although the ARF1-SNAP25 interaction may be increased, immunoblotting demonstrated 21% lower ARF1-21 (21 kiloDaltons) in schizophrenia samples (P = 0.04). In contrast, the mitochondrial protein UQCRC1 (ubiquinol-cytochrome c reductase core protein 1) did not differ. Lower ARF1-21 levels were associated with the previously reported increased SNAP25-syntaxin interaction in schizophrenia (r = -0.39, P = 0.04). Additional immunoprecipitation studies confirmed the ARF1-21-SNAP25 interaction, independent of UQCRC1. Both ARF1 and SNAP25 were localized to synaptosomes. Confocal microscopy demonstrated co-localization of ARF1 and SNAP25, and further suggested fivefold enrichment of ARF1 in synaptosomes containing an excitatory marker (vesicular glutamate transporter) compared with synaptosomes containing an inhibitory marker (vesicular GABA transporter). The present findings suggest an association between abnormalities of SNARE proteins involved with vesicular neurotransmission and the mitochondrial protein ARF1 that may contribute to the pathophysiology of schizophrenia., (Copyright © 2019 IBRO. Published by Elsevier Ltd. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
44. The FUS-DDIT3 Interactome in Myxoid Liposarcoma.
- Author
-
Yu JSE, Colborne S, Hughes CS, Morin GB, and Nielsen TO
- Subjects
- Carrier Proteins chemistry, Cell Line, Tumor, Humans, Liposarcoma, Myxoid genetics, Oncogene Proteins, Fusion chemistry, Protein Binding, Protein Interaction Domains and Motifs, Protein Interaction Mapping, Proteome, Proteomics methods, Reproducibility of Results, Carrier Proteins metabolism, Liposarcoma, Myxoid metabolism, Oncogene Proteins, Fusion metabolism
- Abstract
Myxoid liposarcoma is a malignant lipogenic tumor that develops in deep soft tissues. While local control rates are good, current chemotherapy options remain ineffective against metastatic disease. Myxoid liposarcoma is characterized by the FUS-DDIT3 fusion oncoprotein that is proposed to function as an aberrant transcription factor, but its exact mechanism of action has remained unclear. To identify the key functional interacting partners of FUS-DDIT3, this study utilized immunoprecipitation-mass spectrometry (IP-MS) to identify the FUS-DDIT3 interactome in whole cell lysates of myxoid liposarcoma cells, and results showed an enrichment of RNA processing proteins. Further quantitative MS analyses of FUS-DDIT3 complexes isolated from nuclear lysates showed that members of several chromatin regulatory complexes were present in the FUS-DDIT3 interactome, including NuRD, SWI/SNF, PRC1, PRC2, and MLL1 COMPASS-like complexes. Co-immunoprecipitation validated the associations of FUS-DDIT3 with BRG1/SMARCA4, BAF155/SMARCC1, BAF57/SMARCE1, and KDM1A. Data from this study provides candidates for functional validation as potential therapeutic targets, particularly for emerging epigenetic drugs., (Copyright © 2019 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
45. Response to Comment on "PP2A inhibition sensitizes cancer stem cells to ABL tyrosine kinase inhibitors in BCR-ABL + human leukemia".
- Author
-
Lai D, Chen M, Su J, Liu X, Rothe K, Hu K, Forrest DL, Eaves CJ, Morin GB, and Jiang X
- Subjects
- Drug Resistance, Neoplasm drug effects, Fusion Proteins, bcr-abl, Humans, Neoplastic Stem Cells drug effects, Protein Kinase Inhibitors, Leukemia, Myelogenous, Chronic, BCR-ABL Positive
- Abstract
LB100 sensitizes resistant chronic phase CML stem and progenitor cells to TKIs and spares healthy bone marrow cells., (Copyright © 2019 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of Science. No claim to original U.S. Government Works.)
- Published
- 2019
- Full Text
- View/download PDF
46. BAP1 haploinsufficiency predicts a distinct immunogenic class of malignant peritoneal mesothelioma.
- Author
-
Shrestha R, Nabavi N, Lin YY, Mo F, Anderson S, Volik S, Adomat HH, Lin D, Xue H, Dong X, Shukin R, Bell RH, McConeghy B, Haegert A, Brahmbhatt S, Li E, Oo HZ, Hurtado-Coll A, Fazli L, Zhou J, McConnell Y, McCart A, Lowy A, Morin GB, Chen T, Daugaard M, Sahinalp SC, Hach F, Le Bihan S, Gleave ME, Wang Y, Churg A, and Collins CC
- Subjects
- Biomarkers, Tumor metabolism, Humans, Immunotherapy, Mesothelioma classification, Mesothelioma therapy, Mutation, Peritoneal Neoplasms classification, Peritoneal Neoplasms therapy, Tumor Microenvironment, Tumor Suppressor Proteins metabolism, Ubiquitin Thiolesterase metabolism, Biomarkers, Tumor genetics, Haploinsufficiency, Mesothelioma genetics, Peritoneal Neoplasms genetics, Tumor Suppressor Proteins genetics, Ubiquitin Thiolesterase genetics
- Abstract
Background: Malignant peritoneal mesothelioma (PeM) is a rare and fatal cancer that originates from the peritoneal lining of the abdomen. Standard treatment of PeM is limited to cytoreductive surgery and/or chemotherapy, and no effective targeted therapies for PeM exist. Some immune checkpoint inhibitor studies of mesothelioma have found positivity to be associated with a worse prognosis., Methods: To search for novel therapeutic targets for PeM, we performed a comprehensive integrative multi-omics analysis of the genome, transcriptome, and proteome of 19 treatment-naïve PeM, and in particular, we examined BAP1 mutation and copy number status and its relationship to immune checkpoint inhibitor activation., Results: We found that PeM could be divided into tumors with an inflammatory tumor microenvironment and those without and that this distinction correlated with haploinsufficiency of BAP1. To further investigate the role of BAP1, we used our recently developed cancer driver gene prioritization algorithm, HIT'nDRIVE, and observed that PeM with BAP1 haploinsufficiency form a distinct molecular subtype characterized by distinct gene expression patterns of chromatin remodeling, DNA repair pathways, and immune checkpoint receptor activation. We demonstrate that this subtype is correlated with an inflammatory tumor microenvironment and thus is a candidate for immune checkpoint blockade therapies., Conclusions: Our findings reveal BAP1 to be a potential, easily trackable prognostic and predictive biomarker for PeM immunotherapy that refines PeM disease classification. BAP1 stratification may improve drug response rates in ongoing phases I and II clinical trials exploring the use of immune checkpoint blockade therapies in PeM in which BAP1 status is not considered. This integrated molecular characterization provides a comprehensive foundation for improved management of a subset of PeM patients.
- Published
- 2019
- Full Text
- View/download PDF
47. RawTools: Rapid and Dynamic Interrogation of Orbitrap Data Files for Mass Spectrometer System Management.
- Author
-
Kovalchik KA, Colborne S, Spencer SE, Sorensen PH, Chen DDY, Morin GB, and Hughes CS
- Subjects
- Data Analysis, Database Management Systems, Mass Spectrometry methods, Information Storage and Retrieval methods, Proteomics methods, Software
- Abstract
Optimizing the quality of proteomics data collected from a mass spectrometer (MS) requires careful selection of acquisition parameters and proper assessment of instrument performance. Software tools capable of extracting a broad set of information from raw files, including meta, scan, quantification, and identification data, are needed to provide guidance for MS system management. In this work, direct extraction and utilization of these data is demonstrated using RawTools, a standalone tool for extracting meta and scan data directly from raw MS files generated on Thermo Orbitrap instruments. RawTools generates summarized and detailed plain text outputs after parsing individual raw files, including scan rates and durations, duty cycle characteristics, precursor and reporter ion quantification, and chromatography performance. RawTools also contains a diagnostic module that includes an optional "preview" database search for facilitating informed decision-making related to optimization of MS performance based on a variety of metrics. RawTools has been developed in C# and utilizes the Thermo RawFileReader library and thus can process raw MS files with high speed and high efficiency on all major operating systems (Windows, MacOS, Linux). To demonstrate the utility of RawTools, the extraction of meta and scan data from both individual and large collections of raw MS files was carried out to identify problematic characteristics of instrument performance. Taken together, the combined rich feature-set of RawTools with the capability for interrogation of MS and experiment performance makes this software a valuable tool for proteomics researchers.
- Published
- 2019
- Full Text
- View/download PDF
48. A Standardized and Reproducible Proteomics Protocol for Bottom-Up Quantitative Analysis of Protein Samples Using SP3 and Mass Spectrometry.
- Author
-
Hughes CS, Sorensen PH, and Morin GB
- Subjects
- Chromatography, High Pressure Liquid, Chromatography, Liquid, Data Interpretation, Statistical, Humans, Peptides, Proteome, Workflow, Magnetite Nanoparticles, Mass Spectrometry methods, Proteomics methods, Proteomics standards
- Abstract
The broad utility of mass spectrometry (MS) for investigating the proteomes of a diverse array of sample types has significantly expanded the use of this technology in biological studies. This widespread use has resulted in a substantial collection of protocols and acquisition approaches designed to obtain the highest-quality data for each experiment. As a result, distilling this information to develop a standard operating protocol for essential workflows, such as bottom-up quantitative shotgun whole proteome analysis, can be complex for users new to MS technology. Further complicating this matter, in-depth description of the methodological choices is seldom given in the literature. In this work, we describe a workflow for quantitative whole proteome analysis that is suitable for biomarker discovery, giving detailed consideration to important stages, including (1) cell lysis and protein cleanup using SP3 paramagnetic beads, (2) quantitative labeling, (3) offline peptide fractionation, (4) MS analysis, and (5) data analysis and interpretation. Special attention is paid to providing comprehensive details for all stages of this proteomics workflow to enhance transferability to external labs. The standardized protocol described here will provide a simplified resource to the proteomics community toward efficient adaptation of MS technology in proteomics studies.
- Published
- 2019
- Full Text
- View/download PDF
49. Single-pot, solid-phase-enhanced sample preparation for proteomics experiments.
- Author
-
Hughes CS, Moggridge S, Müller T, Sorensen PH, Morin GB, and Krijgsveld J
- Subjects
- Buffers, Chemical Fractionation instrumentation, Detergents chemistry, Guanidine chemistry, HEK293 Cells, Humans, Hydrophobic and Hydrophilic Interactions, Isothiocyanates chemistry, Proteomics instrumentation, Solvents chemistry, Specimen Handling standards, Urea chemistry, Chemical Fractionation methods, Proteome isolation & purification, Proteomics methods, Solid Phase Microextraction methods
- Abstract
A critical step in proteomics analysis is the optimal extraction and processing of protein material to ensure the highest sensitivity in downstream detection. Achieving this requires a sample-handling technology that exhibits unbiased protein manipulation, flexibility in reagent use, and virtually lossless processing. Addressing these needs, the single-pot, solid-phase-enhanced sample-preparation (SP3) technology is a paramagnetic bead-based approach for rapid, robust, and efficient processing of protein samples for proteomic analysis. SP3 uses a hydrophilic interaction mechanism for exchange or removal of components that are commonly used to facilitate cell or tissue lysis, protein solubilization, and enzymatic digestion (e.g., detergents, chaotropes, salts, buffers, acids, and solvents) before downstream proteomic analysis. The SP3 protocol consists of nonselective protein binding and rinsing steps that are enabled through the use of ethanol-driven solvation capture on the surface of hydrophilic beads, and elution of purified material in aqueous conditions. In contrast to alternative approaches, SP3 combines compatibility with a substantial collection of solution additives with virtually lossless and unbiased recovery of proteins independent of input quantity, all in a simplified single-tube protocol. The SP3 protocol is simple and efficient, and can be easily completed by a standard user in ~30 min, including reagent preparation. As a result of these properties, SP3 has successfully been used to facilitate examination of a broad range of sample types spanning simple and complex protein mixtures in large and very small amounts, across numerous organisms. This work describes the steps and extensive considerations involved in performing SP3 in bottom-up proteomics, using a simplified protein cleanup scenario for illustration.
- Published
- 2019
- Full Text
- View/download PDF
50. Histone Deacetylase Inhibitors Synergize with Catalytic Inhibitors of EZH2 to Exhibit Antitumor Activity in Small Cell Carcinoma of the Ovary, Hypercalcemic Type.
- Author
-
Wang Y, Chen SY, Colborne S, Lambert G, Shin CY, Santos ND, Orlando KA, Lang JD, Hendricks WPD, Bally MB, Karnezis AN, Hass R, Underhill TM, Morin GB, Trent JM, Weissman BE, and Huntsman DG
- Subjects
- Animals, Apoptosis drug effects, Biocatalysis drug effects, Carcinoma, Small Cell complications, Carcinoma, Small Cell genetics, Carcinoma, Small Cell pathology, Cell Cycle Checkpoints drug effects, Cell Differentiation drug effects, Cell Line, Tumor, Cell Lineage drug effects, DNA Helicases metabolism, Drug Synergism, Enhancer of Zeste Homolog 2 Protein metabolism, Epigenesis, Genetic drug effects, Female, Gene Expression Regulation, Neoplastic drug effects, Histone Deacetylase Inhibitors pharmacology, Humans, Hydroxamic Acids pharmacology, Hydroxamic Acids therapeutic use, Hypercalcemia complications, Mice, Nuclear Proteins metabolism, Ovarian Neoplasms complications, Ovarian Neoplasms genetics, Ovarian Neoplasms pathology, Proteome metabolism, Transcription Factors metabolism, Xenograft Model Antitumor Assays, Antineoplastic Agents therapeutic use, Carcinoma, Small Cell drug therapy, Enhancer of Zeste Homolog 2 Protein antagonists & inhibitors, Histone Deacetylase Inhibitors therapeutic use, Hypercalcemia drug therapy, Ovarian Neoplasms drug therapy
- Abstract
Small cell carcinoma of the ovary, hypercalcemic type (SCCOHT) is a rare but extremely lethal malignancy that mainly impacts young women. SCCOHT is characterized by a diploid genome with loss of SMARCA4 and lack of SMARCA2 expression, two mutually exclusive ATPases of the SWI/SNF chromatin-remodeling complex. We and others have identified the histone methyltransferase EZH2 as a promising therapeutic target for SCCOHT, suggesting that SCCOHT cells depend on the alternation of epigenetic pathways for survival. In this study, we found that SCCOHT cells were more sensitive to pan-HDAC inhibitors compared with other ovarian cancer lines or immortalized cell lines tested. Pan-HDAC inhibitors, such as quisinostat, reversed the expression of a group of proteins that were deregulated in SCCOHT cells due to SMARCA4 loss, leading to growth arrest, apoptosis, and differentiation in vitro and suppressed tumor growth of xenografted tumors of SCCOHT cells. Moreover, combined treatment of HDAC inhibitors and EZH2 inhibitors at sublethal doses synergistically induced histone H3K27 acetylation and target gene expression, leading to rapid induction of apoptosis and growth suppression of SCCOHT cells and xenografted tumors. Therefore, our preclinical study highlighted the therapeutic potential of combined treatment of HDAC inhibitors with EZH2 catalytic inhibitors to treat SCCOHT., (©2018 American Association for Cancer Research.)
- Published
- 2018
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.