330 results on '"Jeremy N Rich"'
Search Results
2. Lgr5 Marks Post-Mitotic, Lineage Restricted Cerebellar Granule Neurons during Postnatal Development.
- Author
-
Tyler E Miller, Jun Wang, Kumar Sukhdeo, Craig Horbinski, Paul J Tesar, Robert J Wechsler-Reya, and Jeremy N Rich
- Subjects
Medicine ,Science - Abstract
Wnt signaling regulates self-renewal and fate commitment of stem and progenitor cells in development and homeostasis. Leucine-rich repeat-containing G-protein coupled receptor 5 (Lgr5) is a co-receptor for Wnt signaling that marks highly proliferative stem and progenitor cells in many epithelial tissue types. Wnt signaling instructs neural developmental and homeostatic processes; however, Lgr5 expression in the developing and adult brain has not been characterized. Here we report that Lgr5 is expressed in the postnatal cerebellum during the maturation and synaptogenesis of cerebellar granule neurons (CGNs), processes controlled by Wnt signaling. Using a transgenic reporter mouse for in vivo Lgr5 expression analysis and lineage tracing, we reveal that Lgr5 specifically identified CGNs and was restricted temporally to the CGN maturation phase within the internal granule layer, but absent in the adult brain. Cells marked by Lgr5 were lineage restricted, post-mitotic and long-lived. The ligand for Lgr5, R-spondin, was secreted in a paracrine fashion that evolved during the maturation of CGNs, which coincided with the Lgr5 expression pattern. Our findings provide potential new insight into the critical regulation of Wnt signaling in the developing cerebellum and support a novel role for Lgr5 in the regulation of post-mitotic cells.
- Published
- 2014
- Full Text
- View/download PDF
3. Cadherin-11 regulates motility in normal cortical neural precursors and glioblastoma.
- Author
-
Jessica D Schulte, Maya Srikanth, Sunit Das, Jianing Zhang, Justin D Lathia, Lihui Yin, Jeremy N Rich, Eric C Olson, John A Kessler, and Anjen Chenn
- Subjects
Medicine ,Science - Abstract
Metastasizing tumor cells undergo a transformation that resembles a process in normal development when non-migratory epithelial cells modulate the expression of cytoskeletal and adhesion proteins to promote cell motility. Here we find a mesenchymal cadherin, Cadherin-11 (CDH11), is increased in cells exiting the ventricular zone (VZ) neuroepithelium during normal cerebral cortical development. When overexpressed in cortical progenitors in vivo, CDH11 causes premature exit from the neuroepithelium and increased cell migration. CDH11 expression is elevated in human brain tumors, correlating with higher tumor grade and decreased patient survival. In glioblastoma, CDH11-expressing tumor cells can be found localized near tumor vasculature. Endothelial cells stimulate TGFβ signaling and CDH11 expression in glioblastoma cells. TGFβ promotes glioblastoma cell motility, and knockdown of CDH11 expression in primary human glioblastoma cells inhibits TGFβ-stimulated migration. Together, these findings show that Cadherin-11 can promote cell migration in neural precursors and glioblastoma cells and suggest that endothelial cells increase tumor aggressiveness by co-opting mechanisms that regulate normal neural development.
- Published
- 2013
- Full Text
- View/download PDF
4. Multiplex flow cytometry barcoding and antibody arrays identify surface antigen profiles of primary and metastatic colon cancer cell lines.
- Author
-
Kumar Sukhdeo, Rosanto I Paramban, Jason G Vidal, Jeanne Elia, Jody Martin, Maricruz Rivera, Daniel R Carrasco, Awad Jarrar, Matthew F Kalady, Christian T Carson, Robert Balderas, Anita B Hjelmeland, Justin D Lathia, and Jeremy N Rich
- Subjects
Medicine ,Science - Abstract
Colon cancer is a deadly disease affecting millions of people worldwide. Current treatment challenges include management of disease burden as well as improvements in detection and targeting of tumor cells. To identify disease state-specific surface antigen signatures, we combined fluorescent cell barcoding with high-throughput flow cytometric profiling of primary and metastatic colon cancer lines (SW480, SW620, and HCT116). Our multiplexed technique offers improvements over conventional methods by permitting the simultaneous and rapid screening of cancer cells with reduced effort and cost. The method uses a protein-level analysis with commercially available antibodies on live cells with intact epitopes to detect potential tumor-specific targets that can be further investigated for their clinical utility. Multiplexed antibody arrays can easily be applied to other tumor types or pathologies for discovery-based approaches to target identification.
- Published
- 2013
- Full Text
- View/download PDF
5. Lyn facilitates glioblastoma cell survival under conditions of nutrient deprivation by promoting autophagy.
- Author
-
Wei Michael Liu, Ping Huang, Niladri Kar, Monica Burgett, Gaelle Muller-Greven, Amy S Nowacki, Clark W Distelhorst, Justin D Lathia, Jeremy N Rich, John C Kappes, and Candece L Gladson
- Subjects
Medicine ,Science - Abstract
Members of the Src family kinases (SFK) can modulate diverse cellular processes, including division, death and survival, but their role in autophagy has been minimally explored. Here, we investigated the roles of Lyn, a SFK, in promoting the survival of human glioblastoma tumor (GBM) cells in vitro and in vivo using lentiviral vector-mediated expression of constitutively-active Lyn (CA-Lyn) or dominant-negative Lyn (DN-Lyn). Expression of either CA-Lyn or DN-Lyn had no effect on the survival of U87 GBM cells grown under nutrient-rich conditions. In contrast, under nutrient-deprived conditions (absence of supplementation with L-glutamine, which is essential for growth of GBM cells, and FBS) CA-Lyn expression enhanced survival and promoted autophagy as well as inhibiting cell death and promoting proliferation. Expression of DN-Lyn promoted cell death. In the nutrient-deprived GBM cells, CA-Lyn expression enhanced AMPK activity and reduced the levels of pS6 kinase whereas DN-Lyn enhanced the levels of pS6 kinase. Similar results were obtained in vitro using another cultured GBM cell line and primary glioma stem cells. On propagation of the transduced GBM cells in the brains of nude mice, the CA-Lyn xenografts formed larger tumors than control cells and autophagosomes were detectable in the tumor cells. The DN-Lyn xenografts formed smaller tumors and contained more apoptotic cells. Our findings suggest that on nutrient deprivation in vitro Lyn acts to enhance the survival of GBM cells by promoting autophagy and proliferation as well as inhibiting cell death, and Lyn promotes the same effects in vivo in xenograft tumors. As the levels of Lyn protein or its activity are elevated in several cancers these findings may be of broad relevance to cancer biology.
- Published
- 2013
- Full Text
- View/download PDF
6. Direct in vivo evidence for tumor propagation by glioblastoma cancer stem cells.
- Author
-
Justin D Lathia, Joseph Gallagher, Jay T Myers, Meizhang Li, Amit Vasanji, Roger E McLendon, Anita B Hjelmeland, Alex Y Huang, and Jeremy N Rich
- Subjects
Medicine ,Science - Abstract
High-grade gliomas (World Health Organization grade III anaplastic astrocytoma and grade IV glioblastoma multiforme), the most prevalent primary malignant brain tumors, display a cellular hierarchy with self-renewing, tumorigenic cancer stem cells (CSCs) at the apex. While the CSC hypothesis has been an attractive model to describe many aspects of tumor behavior, it remains controversial due to unresolved issues including the use of ex vivo analyses with differential growth conditions. A CSC population has been confirmed in malignant gliomas by preferential tumor formation from cells directly isolated from patient biopsy specimens. However, direct comparison of multiple tumor cell populations with analysis of the resulting phenotypes of each population within a representative tumor environment has not been clearly described. To directly test the relative tumorigenic potential of CSCs and non-stem tumor cells in the same microenvironment, we interrogated matched tumor populations purified from a primary human tumor transplanted into a xenograft mouse model and monitored competitive in vivo tumor growth studies using serial in vivo intravital microscopy. While CSCs were a small minority of the initial transplanted cancer cell population, the CSCs, not the non-stem tumor cells, drove tumor formation and yielded tumors displaying a cellular hierarchy. In the resulting tumors, a fraction of the initial transplanted CSCs maintained expression of stem cell and proliferation markers, which were significantly higher compared to the non-stem tumor cell population and demonstrated that CSCs generated cellular heterogeneity within the tumor. These head-to-head comparisons between matched CSCs and non-stem tumor cells provide the first functional evidence using live imaging that in the same microenvironment, CSCs more than non-stem tumor cells are responsible for tumor propagation, confirming the functional definition of a CSC.
- Published
- 2011
- Full Text
- View/download PDF
7. c-Myc is required for maintenance of glioma cancer stem cells.
- Author
-
Jialiang Wang, Hui Wang, Zhizhong Li, Qiulian Wu, Justin D Lathia, Roger E McLendon, Anita B Hjelmeland, and Jeremy N Rich
- Subjects
Medicine ,Science - Abstract
Malignant gliomas rank among the most lethal cancers. Gliomas display a striking cellular heterogeneity with a hierarchy of differentiation states. Recent studies support the existence of cancer stem cells in gliomas that are functionally defined by their capacity for extensive self-renewal and formation of secondary tumors that phenocopy the original tumors. As the c-Myc oncoprotein has recognized roles in normal stem cell biology, we hypothesized that c-Myc may contribute to cancer stem cell biology as these cells share characteristics with normal stem cells.Based on previous methods that we and others have employed, tumor cell populations were enriched or depleted for cancer stem cells using the stem cell marker CD133 (Prominin-1). We characterized c-Myc expression in matched tumor cell populations using real time PCR, immunoblotting, immunofluorescence and flow cytometry. Here we report that c-Myc is highly expressed in glioma cancer stem cells relative to non-stem glioma cells. To interrogate the significance of c-Myc expression in glioma cancer stem cells, we targeted its expression using lentivirally transduced short hairpin RNA (shRNA). Knockdown of c-Myc in glioma cancer stem cells reduced proliferation with concomitant cell cycle arrest in the G(0)/G(1) phase and increased apoptosis. Non-stem glioma cells displayed limited dependence on c-Myc expression for survival and proliferation. Further, glioma cancer stem cells with decreased c-Myc levels failed to form neurospheres in vitro or tumors when xenotransplanted into the brains of immunocompromised mice.These findings support a central role of c-Myc in regulating proliferation and survival of glioma cancer stem cells. Targeting core stem cell pathways may offer improved therapeutic approaches for advanced cancers.
- Published
- 2008
- Full Text
- View/download PDF
8. Transcription Elongation Machinery Is a Druggable Dependency and Potentiates Immunotherapy in Glioblastoma Stem Cells
- Author
-
Ryan C. Gimple, Guoxin Zhang, Linjie Zhao, Leo J.Y. Kim, Jia Z. Shen, Cheryl Kim, Briana C. Prager, Xujun Wang, Jean A. Bernatchez, Xiang-Dong Fu, Jeremy N. Rich, Kailin Yang, Jair L. Siqueira-Neto, Deobrat Dixit, Zhixin Qiu, Lukas Chavez, Zhe Zhu, Deguan Lv, Ye Zheng, Denise Hinz, Zhengyu Liang, Charles Spruck, Xiuxing Wang, Chunyu Jin, Qiyuan Yang, Qiulian Wu, Lihua Min, Katherine A. Jones, Zhen Dong, and Shruti Bhargava
- Subjects
Male ,Regulatory T cell ,Biology ,Article ,Epigenesis, Genetic ,Mice ,chemistry.chemical_compound ,Interferon ,Transcription (biology) ,RNA polymerase ,Gene expression ,Tumor Microenvironment ,medicine ,Animals ,Humans ,Brain Neoplasms ,Middle Aged ,Neural stem cell ,Immune checkpoint ,Gene Expression Regulation, Neoplastic ,medicine.anatomical_structure ,Oncology ,chemistry ,embryonic structures ,Neoplastic Stem Cells ,Cancer research ,Female ,Immunotherapy ,Stem cell ,Glioblastoma ,medicine.drug - Abstract
Glioblastoma (GBM) is the most lethal primary brain cancer characterized by therapeutic resistance, which is promoted by GBM stem cells (GSC). Here, we interrogated gene expression and whole-genome CRISPR/Cas9 screening in a large panel of patient-derived GSCs, differentiated GBM cells (DGC), and neural stem cells (NSC) to identify master regulators of GSC stemness, revealing an essential transcription state with increased RNA polymerase II–mediated transcription. The YY1 and transcriptional CDK9 complex was essential for GSC survival and maintenance in vitro and in vivo. YY1 interacted with CDK9 to regulate transcription elongation in GSCs. Genetic or pharmacologic targeting of the YY1–CDK9 complex elicited RNA m6A modification–dependent interferon responses, reduced regulatory T-cell infiltration, and augmented efficacy of immune checkpoint therapy in GBM. Collectively, these results suggest that YY1–CDK9 transcription elongation complex defines a targetable cell state with active transcription, suppressed interferon responses, and immunotherapy resistance in GBM. Significance: Effective strategies to rewire immunosuppressive microenvironment and enhance immunotherapy response are still lacking in GBM. YY1-driven transcriptional elongation machinery represents a druggable target to activate interferon response and enhance anti–PD-1 response through regulating the m6A modification program, linking epigenetic regulation to immunomodulatory function in GBM. This article is highlighted in the In This Issue feature, p. 275
- Published
- 2022
- Full Text
- View/download PDF
9. The RNA m6A Reader YTHDF2 Maintains Oncogene Expression and Is a Targetable Dependency in Glioblastoma Stem Cells
- Author
-
Zhen Dong, Kristoffer Vitting-Seerup, Xiuxing Wang, Li Jiang, Qiulian Wu, Qi Xie, Briana C. Prager, Shruti Bhargava, Reilly L. Kidwell, Jeremy N. Rich, Zhixin Qiu, Leo J.Y. Kim, Deobrat Dixit, Yang Wang, Samie R. Jaffrey, Petra Hamerlik, Hui Xian Poh, Jing Crystal Zhao, Ryan C. Gimple, and Zhe Zhu
- Subjects
0301 basic medicine ,endocrine system ,Cell ,Biology ,Article ,Transcriptome ,03 medical and health sciences ,0302 clinical medicine ,Cancer stem cell ,Epitranscriptomics ,medicine ,Humans ,RNA, Messenger ,Oncogene ,Brain Neoplasms ,fungi ,RNA-Binding Proteins ,RNA ,Neural stem cell ,Cell biology ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,030220 oncology & carcinogenesis ,Neoplastic Stem Cells ,Stem cell ,Glioblastoma - Abstract
Glioblastoma is a universally lethal cancer driven by glioblastoma stem cells (GSC). Here, we interrogated N6-methyladenosine (m6A) mRNA modifications in GSCs by methyl RNA immunoprecipitation followed by sequencing and transcriptome analysis, finding transcripts marked by m6A often upregulated compared with normal neural stem cells (NSC). Interrogating m6A regulators, GSCs displayed preferential expression, as well as in vitro and in vivo dependency, of the m6A reader YTHDF2, in contrast to NSCs. Although YTHDF2 has been reported to destabilize mRNAs, YTHDF2 stabilized MYC and VEGFA transcripts in GSCs in an m6A-dependent manner. We identified IGFBP3 as a downstream effector of the YTHDF2–MYC axis in GSCs. The IGF1/IGF1R inhibitor linsitinib preferentially targeted YTHDF2-expressing cells, inhibiting GSC viability without affecting NSCs and impairing in vivo glioblastoma growth. Thus, YTHDF2 links RNA epitranscriptomic modifications and GSC growth, laying the foundation for the YTHDF2–MYC–IGFBP3 axis as a specific and novel therapeutic target in glioblastoma. Significance: Epitranscriptomics promotes cellular heterogeneity in cancer. RNA m6A landscapes of cancer and NSCs identified cell type–specific dependencies and therapeutic vulnerabilities. The m6A reader YTHDF2 stabilized MYC mRNA specifically in cancer stem cells. Given the challenge of targeting MYC, YTHDF2 presents a therapeutic target to perturb MYC signaling in glioblastoma. This article is highlighted in the In This Issue feature, p. 211
- Published
- 2021
- Full Text
- View/download PDF
10. Adipocytes promote breast tumorigenesis through TAZ-dependent secretion of Resistin
- Author
-
Jeremy N. Rich, Zengqiang Yuan, Rong Wu, Qi Xie, Xiao-Song Chen, Yuhao Gao, Qing He, Liang Wang, Kun-Wei Shen, Ryan C. Gimple, and Yajin Liao
- Subjects
TAZ ,Carcinogenesis ,Breast Neoplasms ,Triple Negative Breast Neoplasms ,Biology ,medicine.disease_cause ,adipocyte ,chemistry.chemical_compound ,Breast cancer ,Transcription (biology) ,Adipocyte ,medicine ,Adipocytes ,breast tumorigenesis ,Animals ,Humans ,Secretion ,Resistin ,Obesity ,Adaptor Proteins, Signal Transducing ,Adiposity ,Cell Proliferation ,Gene knockdown ,Multidisciplinary ,Fatty Acids ,Cell Biology ,Biological Sciences ,medicine.disease ,Diet ,Gene Expression Regulation, Neoplastic ,Mice, Inbred C57BL ,PPAR gamma ,chemistry ,Transcriptional Coactivator with PDZ-Binding Motif Proteins ,Cancer research ,Neoplastic Stem Cells ,Trans-Activators ,Female ,Hormone - Abstract
Significance Adipocytes are the most abundant and perhaps most active components of the tumor microenvironment in obese individuals that potentiate breast tumorigenesis through secretory mechanisms. The modulation of adipocytes can be novel therapy targets for breast cancer. Here, we revealed a specific upregulation of adipocytic TAZ through the FFA/PPARγ axis in diet-induced adiposity. Adipocytic TAZ knockdown or deficiency in mice inhibits adipocyte-induced breast cancer proliferation and stemness through impaired expression and secretion of Resistin. Immunostaining in triple-negative breast cancer samples showed that higher adipocytic TAZ/Resistin expression associates with higher clinical stages and poorer survival, demonstrating promising therapeutic targets., Adipocytes have been implicated in breast tumor growth and stemness maintenance through secreted factors. However, the mechanisms by which these cytokines are regulated during diet-induced obesity and contribute to breast tumorigenesis remain largely unknown. Here we show that transcription cofactor TAZ in adipocytes is directly up-regulated by the free fatty acid/PPARγ axis upon dietary fat stimulation. TAZ knockdown alters the expression profile of a series of secreted proteins and attenuates the tumor-supporting function of adipocytes. Moreover, we identify Resistin, an adipose-derived hormone, as a functional downstream target of TAZ, which facilitates tumorigenesis, and its expression correlated with adipocyitc TAZ in triple-negative breast cancer samples. Further, Adiponectin-cre–mediated TAZ knockout in adipocytes mitigates breast tumor growth. Taken together, our findings highlight how diet-induced TAZ expression in adipocytes promotes tumorigenesis, suggesting promising cancer therapeutic targets.
- Published
- 2020
11. A vaccine for glioma
- Author
-
Jeremy N. Rich, Duane Mitchell, Sameer Agnihotri, and Kailin Yang
- Subjects
Cancer Research ,IDH1 ,Oncology ,business.industry ,Metabolic enzymes ,Glioma ,Cancer research ,Medicine ,Cancer ,Phases of clinical research ,In patient ,business ,medicine.disease - Abstract
Despite substantial advances in understanding of the molecular features of gliomas, the therapeutic options for these aggressive tumors remain scarce. Rich, Mitchell and colleagues provide their views about a phase 1 clinical trial testing the safety and efficacy of vaccines against cancer expressing mutant metabolic enzyme IDH1 in patients with high-grade glioma.
- Published
- 2021
- Full Text
- View/download PDF
12. The Meningioma Enhancer Landscape Delineates Novel Subgroups and Drives Druggable Dependencies
- Author
-
Harish N. Vasudevan, Arie Perry, Zhe Zhu, Deobrat Dixit, Jeremy N. Rich, Qi Xie, Jean A. Bernatchez, Andrew R. Morton, Derrick Lee, Lisa C. Wallace, Qiulian Wu, Jair L. Siqueira-Neto, David R. Raleigh, Ryan C. Gimple, Briana C. Prager, Shruti Bhargava, Xiuxing Wang, Clark C. Chen, Stephen C. Mack, Gene H. Barnett, Melike Pekmezci, Lukas Chavez, Bradley H. King, and M.A. Vogelbaum
- Subjects
Epigenomics ,0301 basic medicine ,Druggability ,Neural crest ,Biology ,Prognosis ,medicine.disease ,nervous system diseases ,Meningioma ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Super-enhancer ,Oncology ,030220 oncology & carcinogenesis ,Genomic Profile ,otorhinolaryngologic diseases ,medicine ,Cancer research ,Humans ,Epigenetics ,Enhancer ,neoplasms ,Grading (tumors) - Abstract
Meningiomas are the most common primary intracranial tumor with current classification offering limited therapeutic guidance. Here, we interrogated meningioma enhancer landscapes from 33 tumors to stratify patients based upon prognosis and identify novel meningioma-specific dependencies. Enhancers robustly stratified meningiomas into three biologically distinct groups (adipogenesis/cholesterol, mesodermal, and neural crest) distinguished by distinct hormonal lineage transcriptional regulators. Meningioma landscapes clustered with intrinsic brain tumors and hormonally responsive systemic cancers with meningioma subgroups, reflecting progesterone or androgen hormonal signaling. Enhancer classification identified a subset of tumors with poor prognosis, irrespective of histologic grading. Superenhancer signatures predicted drug dependencies with superior in vitro efficacy to treatment based upon the NF2 genomic profile. Inhibition of DUSP1, a novel and druggable meningioma target, impaired tumor growth in vivo. Collectively, epigenetic landscapes empower meningioma classification and identification of novel therapies. Significance: Enhancer landscapes inform prognostic classification of aggressive meningiomas, identifying tumors at high risk of recurrence, and reveal previously unknown therapeutic targets. Druggable dependencies discovered through epigenetic profiling potentially guide treatment of intractable meningiomas. This article is highlighted in the In This Issue feature, p. 1611
- Published
- 2020
- Full Text
- View/download PDF
13. Type I Interferon Regulates a Coordinated Gene Network to Enhance Cytotoxic T Cell–Mediated Tumor Killing
- Author
-
Kei-ichiro Arimoto, Leo J.Y. Kim, Balázs Győrffy, Klaus-Peter Knobeloch, Dong-Er Zhang, Hua Cheng, Christoph Burkart, Xiang-Dong Fu, Jeremy N. Rich, Hu Cang, Jun-Bao Fan, Yu Zhou, Hui-Zhong Xu, Ming Yan, Sayuri Miyauchi, and Dan Liu
- Subjects
T-Lymphocytes ,Gene regulatory network ,Breast Neoplasms ,Ubiquitin-Activating Enzymes ,medicine.disease_cause ,Article ,Metastasis ,Mice ,Interferon ,medicine ,Animals ,Humans ,Gene Regulatory Networks ,STAT1 ,Ubiquitins ,Transcription factor ,Cell Proliferation ,Regulation of gene expression ,biology ,virus diseases ,STAT2 Transcription Factor ,medicine.disease ,ISG15 ,Gene Expression Regulation, Neoplastic ,STAT1 Transcription Factor ,Oncology ,Interferon Type I ,Cancer research ,biology.protein ,Female ,Carcinogenesis ,Transcription Factors ,medicine.drug - Abstract
Type I interferons (IFN), which activate many IFN-stimulated genes (ISG), are known to regulate tumorigenesis. However, little is known regarding how various ISGs coordinate with one another in developing antitumor effects. Here, we report that the ISG UBA7 is a tumor suppressor in breast cancer. UBA7 encodes an enzyme that catalyzes the covalent conjugation of the ubiquitin-like protein product of another ISG (ISG15) to cellular proteins in a process known as “ISGylation.” ISGylation of other ISGs, including STAT1 and STAT2, synergistically facilitates production of chemokine-receptor ligands to attract cytotoxic T cells. These gene-activation events are further linked to clustering and nuclear relocalization of STAT1/2 within IFN-induced promyelocytic leukemia (PML) bodies. Importantly, this coordinated ISG–ISGylation network plays a central role in suppressing murine breast cancer growth and metastasis, which parallels improved survival in patients with breast cancer. These findings reveal a cooperative IFN-inducible gene network in orchestrating a tumor-suppressive microenvironment. Significance: We report a highly cooperative ISG network, in which UBA7-mediated ISGylation facilitates clustering of transcription factors and activates an antitumor gene-expression program. These findings provide mechanistic insights into immune evasion in breast cancer associated with UBA7 loss, emphasizing the importance of a functional ISG–ISGylation network in tumor suppression. This article is highlighted in the In This Issue feature, p. 327
- Published
- 2020
- Full Text
- View/download PDF
14. The landscape of extrachromosomal circular DNA in medulloblastoma
- Author
-
Robert J. Wechsler-Reya, Siavash R. Dehkordi, James T. Robinson, Meghana Pagadala, Miriam Adam, Jesse R. Dixon, Vineet Bafna, Joshua T. Lange, Edwin F. Juarez, Shanqing Wang, Hannah Carter, Ivy Tsz-Lo Wong, Michael J. Levy, Sunita Sridhar, Sameena Wani, Ashutosh Tiwari, Sahaana Chandran, Owen Chapman, Richard H. Scheuermann, John Robertson Crawford, Jens Luebeck, Ernest Fraenkel, Paul S. Mischel, Scott L. Pomeroy, Lukas Chavez, Jon D. Larson, Denise M. Malicki, Nicole G. Coufal, Yingxi Lin, Jeremy N. Rich, and Jill P. Mesirov
- Subjects
Whole genome sequencing ,Medulloblastoma ,Oncogene ,Cancer research ,medicine ,Wnt signaling pathway ,Drug resistance ,Biology ,Extrachromosomal circular DNA ,Enhancer ,medicine.disease ,Chromatin - Abstract
SUMMARYExtrachromosomal circular DNA (ecDNA) is an important driver of aggressive tumor growth, promoting high oncogene copy number, intratumoral heterogeneity, accelerated evolution of drug resistance, enhancer rewiring, and poor outcome. ecDNA has been reported in medulloblastoma (MB), the most common malignant pediatric brain tumor, but the ecDNA landscape and its association with specific MB subgroups, its impact on enhancer rewiring, and its potential clinical implications, are not known. We assembled a retrospective cohort of 468 MB patient samples with available whole genome sequencing (WGS) data covering the four major MB subgroups WNT, SHH, Group 3 and Group 4. Using computational methods for the detection and reconstruction of ecDNA1, we find ecDNA in 82 patients (18%) and observe that ecDNA+ MB patients are more than twice as likely to relapse and three times as likely to die of disease. In addition, we find that individual medulloblastoma tumors often harbor multiple ecDNAs, each containing different amplified oncogenes along with co-amplified non-coding regulatory enhancers. ecDNA was substantially more prevalent among 31 analyzed patient-derived xenograft (PDX) models and cell lines than in our patient cohort. By mapping the accessible chromatin and 3D conformation landscapes of MB tumors that harbor ecDNA, we observe frequent candidate “enhancer rewiring” events that spatially link oncogenes with co-amplified enhancers. Our study reveals the frequency and diversity of ecDNA in a subset of highly aggressive tumors and suggests enhancer rewiring as a frequent oncogenic mechanism of ecDNAs in MB. Further, these results demonstrate that ecDNA is a frequent and potent driver of poor outcome in MB patients.
- Published
- 2021
- Full Text
- View/download PDF
15. WDR5 represents a therapeutically exploitable target for cancer stem cells in glioblastoma
- Author
-
Kelly Mitchell, Sajina Shakya, Sonali Arora, Samuel A. Sprowls, Daniel J. Silver, Christopher M. Goins, Lisa Wallace, Gustavo Roversi, Rachel Schafer, Kristen Kay, Tyler E. Miller, Adam Lauko, John Bassett, Anjali Kashyap, J. D’Amato Kass, Erin E. Mulkearns-Hubert, Sadie Johnson, Joseph Alvarado, Jeremy N. Rich, Patrick J. Paddison, Anoop P. Patel, Shaun R. Stauffer, Christopher G. Hubert, and Justin D. Lathia
- Subjects
education.field_of_study ,Population ,Methylation ,Biology ,medicine.disease ,Histone H3 ,Cancer stem cell ,Transcription (biology) ,Cancer research ,medicine ,WDR5 ,Epigenetics ,education ,Glioblastoma - Abstract
Glioblastomas (GBMs) are heterogeneous, treatment-resistant tumors that are driven by populations of cancer stem cells (CSCs). Despite their importance for tumor growth, few molecular mechanisms critical for CSC population maintenance have been exploited for therapeutic development. We developed a spatially resolved loss-of-function screen in GBM patient-derived organoids to identify essential epigenetic regulators in the SOX2-enriched, therapy resistant niche and identified WDR5 as indispensable for this population. WDR5 is a component of the WRAD complex, which promotes SET1-family-mediated Lys4 methylation of histone H3, associated with positive regulation of transcription. In GBM CSC models, WDR5 inhibitors blocked WRAD complex assembly and reduced H3K4 trimethylation and expression of genes involved in CSC-relevant oncogenic pathways. H3K4me3 peaks lost with WDR5 inhibitor treatment occurred disproportionally on POU transcription factor motifs, including the POU5F1(OCT4)::SOX2 motif. We incorporated a SOX2/OCT4 motif driven GFP reporter system into our CSC cell models and found that WDR5 inhibitor treatment diminished reporter activity. Further, WDR5 inhibitor treatment altered the stem cell state, disrupting CSCin vitrogrowth and self-renewal as well asin vivotumor growth. These findings highlight the role of WDR5 and the WRAD complex in maintaining the CSC state and provide a rationale for therapeutic development of WDR5 inhibitors for GBM and other advanced cancers.SignificanceIn this study, we perform an epigenetic-focused functional genomics screen in glioblastoma organoids and identify WDR5 as an essential epigenetic regulator in the SOX2-enriched, therapy resistant cancer stem cell niche.Graphical Abstract
- Published
- 2021
- Full Text
- View/download PDF
16. A delicate initiation: Lipolysis of lipid droplets fuels glioblastoma
- Author
-
Kailin Yang and Jeremy N. Rich
- Subjects
Lipolysis ,Fatty Acids ,Tumor cells ,Oxidation reduction ,Cell Biology ,Lipid Droplets ,Biology ,medicine.disease ,Glucose deprivation ,Lipid droplet ,Cancer research ,medicine ,Humans ,Glioblastoma ,Molecular Biology ,Oxidation-Reduction - Abstract
Liu et al. (2021) demonstrate that CHKα2 is capable of promoting lipolysis of lipid droplets through mechanisms that require sequential steps of post-translational modifications after glucose deprivation. Intriguingly, the oxidation of fatty acids derived from lipid droplets is essential for the survival of tumor cells that informs clinical outcome among glioblastoma patients.
- Published
- 2021
17. Inhibiting DNA-PK induces glioma stem cell differentiation and sensitizes glioblastoma to radiation in mice
- Author
-
Shideng Bao, Kui Zhai, Xiaoguang Fang, Jeremy N. Rich, Zhi Huang, Leo J.Y. Kim, Jennifer S. Yu, Alexandru Almasan, Xiaoxia Li, Weiwei Tao, Qiulian Wu, Qian Huang, and George R. Stark
- Subjects
endocrine system ,DNA damage ,Cellular differentiation ,DNA-Activated Protein Kinase ,Mice ,SOX2 ,Cell Line, Tumor ,Glioma ,medicine ,Animals ,Protein kinase A ,Transcription factor ,Brain Neoplasms ,Chemistry ,SOXB1 Transcription Factors ,fungi ,Cell Differentiation ,General Medicine ,medicine.disease ,DNA-Binding Proteins ,Cell culture ,embryonic structures ,Neoplastic Stem Cells ,Cancer research ,biological phenomena, cell phenomena, and immunity ,Stem cell ,Glioblastoma - Abstract
Glioblastoma (GBM), a lethal primary brain tumor, contains glioma stem cells (GSCs) that promote malignant progression and therapeutic resistance. SOX2 is a core transcription factor that maintains the properties of stem cells, including GSCs, but mechanisms associated with posttranslational SOX2 regulation in GSCs remain elusive. Here, we report that DNA-dependent protein kinase (DNA-PK) governs SOX2 stability through phosphorylation, resulting in GSC maintenance. Mass spectrometric analyses of SOX2-binding proteins showed that DNA-PK interacted with SOX2 in GSCs. The DNA-PK catalytic subunit (DNA-PKcs) was preferentially expressed in GSCs compared to matched non-stem cell tumor cells (NSTCs) isolated from patient-derived GBM xenografts. DNA-PKcs phosphorylated human SOX2 at S251, which stabilized SOX2 by preventing WWP2-mediated ubiquitination, thus promoting GSC maintenance. We then demonstrated that when the nuclear DNA of GSCs either in vitro or in GBM xenografts in mice was damaged by irradiation or treatment with etoposide, the DNA-PK complex dissociated from SOX2, which then interacted with WWP2, leading to SOX2 degradation and GSC differentiation. These results suggest that DNA-PKcs-mediated phosphorylation of S251 was critical for SOX2 stabilization and GSC maintenance. Pharmacological inhibition of DNA-PKcs with the DNA-PKcs inhibitor NU7441 reduced GSC tumorsphere formation in vitro and impaired growth of intracranial human GBM xenografts in mice as well as sensitized the GBM xenografts to radiotherapy. Our findings suggest that DNA-PK maintains GSCs in a stem cell state and that DNA damage triggers GSC differentiation through precise regulation of SOX2 stability, highlighting that DNA-PKcs has potential as a therapeutic target in glioblastoma.
- Published
- 2021
- Full Text
- View/download PDF
18. HAUSP Stabilizes SOX2 through Deubiquitination to Maintain Self-renewal and Tumorigenic Potential of Glioma Stem Cells
- Author
-
Weiwei Tao, Shideng Bao, Jeremy N. Rich, Justin D. Lathia, Jennifer S. Yu, Qiulian Wu, Xiaoguang Fang, Kui Zhai, Zhi Huang, and Qian Huang
- Subjects
endocrine system ,Chemistry ,fungi ,medicine.disease ,Small hairpin RNA ,SOX2 ,Glioma ,Radioresistance ,embryonic structures ,medicine ,Cancer research ,Post-translational regulation ,Stem cell ,Transcription factor ,Deubiquitination - Abstract
Glioblastoma (GBM) is the most lethal brain tumor containing glioma stem cells (GSCs) that promote malignant growth and therapeutic resistance. The self-renewal and tumorigenic potential of GSCs are maintained by core stem cell transcription factors including SOX2. Defining the posttranslational regulation of SOX2 may offer new insights into GSC biology and potential therapeutic opportunity. Here, we discover that HAUSP stabilizes SOX2 through deubiquitination to maintain GSC self-renewal and tumorigenic potential. HAUSP is preferentially expressed in GSCs in perivascular niches in GBMs. Disrupting HAUSP by shRNA or its inhibitor P22077 promoted SOX2 degradation, induced GSC differentiation, impaired GSC tumorigenic potential, and suppressed GBM tumor growth. Importantly, pharmacological inhibition of HAUSP synergized with radiation to inhibit GBM growth and extended animal survival, indicating that targeting HAUSP may overcome GSC-mediated radioresistance. Our findings reveal an unappreciated crucial role of HAUSP in the GSC maintenance and provide a promising target for developing effective anti-GSC therapeutics to improve GBM treatment.HighlightsHAUSP deubiquitinates and stabilizes SOX2 in glioma stem cells (GSCs).HAUSP is preferentially expressed by GSCs in perivascular niches in GBMs.HAUSP is required for maintaining GSC self-renewal and tumorigenic potential.Targeting HAUSP inhibited malignant growth in GSC-derived GBM xenografts.Inhibition of HAUSP synergized with radiation to suppress GBM tumor growth.
- Published
- 2021
- Full Text
- View/download PDF
19. Glioma Stem Cell–Specific Superenhancer Promotes Polyunsaturated Fatty-Acid Synthesis to Support EGFR Signaling
- Author
-
Stephen C. Mack, Leo J.Y. Kim, Hu Cang, Zhen Dong, Deguan Lv, Jeremy N. Rich, Megan E Wolf, Reilly L. Kidwell, Qi Xie, Tao Huan, Qing Ye, J. Mark Brown, Xiuxing Wang, Tengqian Sun, Junfeng Bi, Guoxin Zhang, Derrick Lee, Ryan C. Gimple, Andrew E. Sloan, Zhe Zhu, Shruti Bhargava, Anthony D. Gromovsky, Linjie Zhao, Briana C. Prager, Li Jiang, Qiulian Wu, and Paul S. Mischel
- Subjects
0301 basic medicine ,endocrine system ,Fatty Acid Elongases ,Cell ,Biology ,Methylation ,Article ,Epigenesis, Genetic ,Histones ,03 medical and health sciences ,0302 clinical medicine ,Downregulation and upregulation ,Cell Line, Tumor ,Glioma ,medicine ,Animals ,Humans ,Cell Proliferation ,Epigenomics ,Regulation of gene expression ,Brain Neoplasms ,Cell growth ,SOXB1 Transcription Factors ,fungi ,medicine.disease ,Up-Regulation ,ErbB Receptors ,Gene Expression Regulation, Neoplastic ,Enhancer Elements, Genetic ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,030220 oncology & carcinogenesis ,Fatty Acids, Unsaturated ,Neoplastic Stem Cells ,Cancer research ,Stem cell ,Signal transduction ,Glioblastoma ,Signal Transduction - Abstract
Glioblastoma ranks among the most aggressive and lethal of all human cancers. Functionally defined glioma stem cells (GSC) contribute to this poor prognosis by driving therapeutic resistance and maintaining cellular heterogeneity. To understand the molecular processes essential for GSC maintenance and tumorigenicity, we interrogated the superenhancer landscapes of primary glioblastoma specimens and in vitro GSCs. GSCs epigenetically upregulated ELOVL2, a key polyunsaturated fatty-acid synthesis enzyme. Targeting ELOVL2 inhibited glioblastoma cell growth and tumor initiation. ELOVL2 depletion altered cellular membrane phospholipid composition, disrupted membrane structural properties, and diminished EGFR signaling through control of fatty-acid elongation. In support of the translational potential of these findings, dual targeting of polyunsaturated fatty-acid synthesis and EGFR signaling had a combinatorial cytotoxic effect on GSCs. Significance: Glioblastoma remains a devastating disease despite extensive characterization. We profiled epigenomic landscapes of glioblastoma to pinpoint cell state–specific dependencies and therapeutic vulnerabilities. GSCs utilize polyunsaturated fatty-acid synthesis to support membrane architecture, inhibition of which impairs EGFR signaling and GSC proliferation. Combinatorial targeting of these networks represents a promising therapeutic strategy. See related commentary by Affronti and Wellen, p. 1161. This article is highlighted in the In This Issue feature, p. 1143
- Published
- 2019
- Full Text
- View/download PDF
20. The dystroglycan receptor maintains glioma stem cells in the vascular niche
- Author
-
Courtney L.R. Jurd, Kathleen S. Ensbey, Thomas Robertson, Zara C. Bruce, Paul R. Jamieson, Carolin Offenhäuser, Rochelle C.J. D’Souza, Bryan W. Day, Fiona M. Smith, Andrew W. Boyd, Po Inglis, Rosalind L. Jeffree, Kevin P. Campbell, Seckin Akgul, Yuchen Li, Justin D. Lathia, Zarnie Lwin, Yi Chieh Lim, Jeremy N. Rich, Terrance Grant Johns, Brett W. Stringer, Krishna P.L. Bhat, and Ulrich Baumgartner
- Subjects
0301 basic medicine ,animal structures ,Central nervous system ,EphA3 ,Mice, SCID ,Biology ,Pathology and Forensic Medicine ,Extracellular matrix ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,0302 clinical medicine ,Mice, Inbred NOD ,Glioma ,Glioblastoma (GBM) ,Glioma stem cell (GSC) commitment ,medicine ,Dystroglycan ,Tumor Microenvironment ,Compartment (development) ,Animals ,Humans ,Integrin-α6 ,Receptor ,Dystroglycans ,Extracellular Signal-Regulated MAP Kinases ,Cells, Cultured ,Original Paper ,Brain Neoplasms ,Dystroglycan (DG) ,medicine.disease ,Cell biology ,030104 developmental biology ,medicine.anatomical_structure ,Cell Transformation, Neoplastic ,MAPK signalling ,biology.protein ,Perivascular niche ,Neoplastic Stem Cells ,Female ,Neurology (clinical) ,Stem cell ,Antibody ,MES-like GBM ,030217 neurology & neurosurgery ,Neoplasm Transplantation - Abstract
Glioblastomas (GBMs) are malignant central nervous system (CNS) neoplasms with a very poor prognosis. They display cellular hierarchies containing self-renewing tumourigenic glioma stem cells (GSCs) in a complex heterogeneous microenvironment. One proposed GSC niche is the extracellular matrix (ECM)-rich perivascular bed of the tumour. Here, we report that the ECM binding dystroglycan (DG) receptor is expressed and functionally glycosylated on GSCs residing in the perivascular niche. Glycosylated αDG is highly expressed and functional on the most aggressive mesenchymal-like (MES-like) GBM tumour compartment. Furthermore, we found that DG acts to maintain an MES-like state via tight control of MAPK activation. Antibody-based blockade of αDG induces robust ERK-mediated differentiation leading to reduced GSC potential. DG was shown to be required for tumour initiation in MES-like GBM, with constitutive loss significantly delaying or preventing tumourigenic potential in-vivo. These findings reveal a central role of the DG receptor, not only as a structural element, but also as a critical factor promoting MES-like GBM and the maintenance of GSCs residing in the perivascular niche. Electronic supplementary material The online version of this article (10.1007/s00401-019-02069-x) contains supplementary material, which is available to authorized users.
- Published
- 2019
21. Glioblastoma stem cells: lessons from the tumor hierarchy in a lethal cancer
- Author
-
Deobrat Dixit, Shruti Bhargava, Jeremy N. Rich, and Ryan C. Gimple
- Subjects
endocrine system ,Population ,Brain tumor ,Review ,Biology ,Epigenesis, Genetic ,03 medical and health sciences ,0302 clinical medicine ,Cancer stem cell ,Tumor Microenvironment ,Genetics ,medicine ,Animals ,Humans ,Epigenetics ,education ,030304 developmental biology ,0303 health sciences ,education.field_of_study ,Brain Neoplasms ,fungi ,Cancer ,Cell Differentiation ,medicine.disease ,Cell culture ,030220 oncology & carcinogenesis ,Neoplastic Stem Cells ,Cancer research ,Stem cell ,Glioblastoma ,Developmental Biology - Abstract
Glioblastoma ranks among the most lethal of all human cancers. Glioblastomas display striking cellular heterogeneity, with stem-like glioblastoma stem cells (GSCs) at the apex. Although the original identification of GSCs dates back more than a decade, the purification and characterization of GSCs remains challenging. Despite these challenges, the evidence that GSCs play important roles in tumor growth and response to therapy has grown. Like normal stem cells, GSCs are functionally defined and distinguished from their differentiated tumor progeny at core transcriptional, epigenetic, and metabolic regulatory levels, suggesting that no single therapeutic modality will be universally effective against a heterogenous GSC population. Glioblastomas induce a systemic immunosuppression with mixed responses to oncoimmunologic modalities, suggesting the potential for augmentation of response with a deeper consideration of GSCs. Unfortunately, the GSC literature has been complicated by frequent use of inferior cell lines and a lack of proper functional analyses. Collectively, glioblastoma offers a reliable cancer to study cancer stem cells to better model the human disease and inform improved biologic understanding and design of novel therapeutics.
- Published
- 2019
- Full Text
- View/download PDF
22. Altered lipid metabolism marks glioblastoma stem and non-stem cells in separate tumor niches
- Author
-
H. Alex Brown, J. Mark Brown, Jeremy N. Rich, Justin D. Lathia, Luiz O. F. Penalva, Briana C. Prager, Arnon Møldrup Knudsen, Sajina Shakya, Christopher G. Hubert, Bjarne Winther Kristensen, Anthony D. Gromovsky, James S. Hale, and Lisa C. Wallace
- Subjects
Organoid ,Tumor heterogeneity ,FADS1 ,Cell ,Biology ,Pathology and Forensic Medicine ,Cellular and Molecular Neuroscience ,Downregulation and upregulation ,Cancer stem cell ,Lipid droplet ,medicine ,Tumor Cells, Cultured ,Tumor Microenvironment ,Humans ,RC346-429 ,Brain Neoplasms ,Research ,Lipid metabolism ,Lipid Metabolism ,Lipid droplets ,Phenotype ,Organoids ,medicine.anatomical_structure ,Cancer research ,Neoplastic Stem Cells ,Neurology. Diseases of the nervous system ,Neurology (clinical) ,Stem cell ,Glioblastoma - Abstract
BackgroundGlioblastoma (GBM) is marked by cellular heterogeneity, including metabolic heterogeneity, that varies among cellular microenvironments in the same tumor. Altered cellular metabolism in cancer is well-established, but how lipid metabolism is altered to suit different microenvironmental conditions and cellular states within a tumor remains unexplored.MethodsWe assessed GBM organoid models that mimic the transition zone between nutrient-rich and nutrient-poor pseudopalisading/perinecrotic tumor zones and performed spatial RNA-sequencing of cells to interrogate lipid metabolism. Using targeted lipidomic analysis, we assessed differences in acutely enriched cancer stem cells (CSCs) and non-CSCs from multiple patient-derived models to explore the link between the stem cell state and lipid metabolism.ResultsSpatial analysis revealed a striking difference in lipid content between microenvironments, with lipid enrichment in the hypoxic organoid cores and the perinecrotic and pseudopalisading regions of primary patient tumors. This was accompanied by regionally restricted upregulation of hypoxia-inducible lipid droplet-associated (HILPDA) gene expression in organoid cores and in clinical GBM specimens, but not lower-grade brain tumors, that was specifically localized to pseudopalisading regions of patient tumors. CSCs have low lipid droplet accumulation compared to non-CSCs in organoid models and xenograft tumors, and prospectively sorted lipid-low GBM cells are functionally enriched for stem cell activity. Targeted lipidomic analysis revealed that CSCs had decreased levels of major classes of neutral lipids compared to non-CSCs but had significantly increased polyunsaturated fatty acid production due to high fatty acid desaturase (FADS1/2) expression.ConclusionsOur data demonstrate that lipid metabolism is differentially altered across GBM microenvironments and cellular hierarchies, providing guidance for targeting of these altered lipid metabolic pathways.Key pointsGBM cells in nutrient-poor tumor regions have increased accumulation of lipid droplets.CSCs have reduced lipid content compared to non-CSCs.GBM CSCs and non-CSCs have disparate lipid metabolisms that may be uniquely targetable.Importance of the StudyMetabolic targeting has long been advocated as a therapy against many tumors including GBM, and it remains an outstanding question whether cancer stem cells (CSCs) have altered lipid metabolism. We demonstrated striking differences in lipid metabolism between diverse cell populations from the same patient. These spatially and phenotypically distinct lipid phenotypes occur clinically in the majority of patients and can be recapitulated in laboratory models. Lipidomic analysis of multiple patient-derived models shows a significant shift in lipid metabolism between GBM CSCs and non-CSCs, suggesting that lipid levels may not be simply a product of the microenvironment but also may be a reflection of cellular state. Our results suggest that therapeutic targeting of GBM lipid metabolism must consider multiple separate tumor cell populations to be effective, and we provide a methodologic framework for studying these metabolically diverse cellular populations.
- Published
- 2021
- Full Text
- View/download PDF
23. Brd4-bound enhancers drive cell-intrinsic sex differences in glioblastoma
- Author
-
Briana C. Prager, Kristopher Berrett, Jeremy N. Rich, Michael N. Wilkinson, Joshua B. Rubin, Sumithra Sankararaman, Lauren Broestl, Xuhua Chen, Robi D. Mitra, Zongtai Qi, Najla Kfoury, Jason Gertz, and Arnav Moudgil
- Subjects
sex differences ,Male ,BRD4 ,Cell ,Gene Expression ,Cell Cycle Proteins ,Regulatory Sequences, Nucleic Acid ,Biology ,medicine.disease_cause ,Brd4-bound enhancers ,Histones ,Proto-Oncogene Proteins c-myc ,Mice ,Sex Factors ,Cell Line, Tumor ,Genetics ,medicine ,Animals ,Humans ,BET inhibitors ,Enhancer ,sex-specific transcriptional programs ,Cell Proliferation ,Sex Characteristics ,Gene knockdown ,Multidisciplinary ,Brain Neoplasms ,glioblastoma ,Nuclear Proteins ,Cancer ,Biological Sciences ,medicine.disease ,Gene Expression Regulation, Neoplastic ,medicine.anatomical_structure ,Cell culture ,Cancer research ,Female ,Tumor Suppressor Protein p53 ,Stem cell ,Carcinogenesis ,Protein Binding ,Transcription Factors - Abstract
Significance Consistent sex differences in incidence and outcome have been reported in numerous cancers including brain tumors. GBM, the most common and aggressive primary brain tumor, occurs with higher incidence and shorter survival in males compared to females. Brd4 is essential for regulating transcriptome-wide gene expression and specifying cell identity, including that of GBM. We report that sex-biased Brd4 activity drives sex differences in GBM and renders male and female tumor cells differentially sensitive to BET inhibitors. The observed sex differences in BETi treatment strongly indicate that sex differences in disease biology translate into sex differences in therapeutic responses. This has critical implications for clinical use of BET inhibitors further affirming the importance of inclusion of sex as a biological variable., Sex can be an important determinant of cancer phenotype, and exploring sex-biased tumor biology holds promise for identifying novel therapeutic targets and new approaches to cancer treatment. In an established isogenic murine model of glioblastoma (GBM), we discovered correlated transcriptome-wide sex differences in gene expression, H3K27ac marks, large Brd4-bound enhancer usage, and Brd4 localization to Myc and p53 genomic binding sites. These sex-biased gene expression patterns were also evident in human glioblastoma stem cells (GSCs). These observations led us to hypothesize that Brd4-bound enhancers might underlie sex differences in stem cell function and tumorigenicity in GBM. We found that male and female GBM cells exhibited sex-specific responses to pharmacological or genetic inhibition of Brd4. Brd4 knockdown or pharmacologic inhibition decreased male GBM cell clonogenicity and in vivo tumorigenesis while increasing both in female GBM cells. These results were validated in male and female patient-derived GBM cell lines. Furthermore, analysis of the Cancer Therapeutic Response Portal of human GBM samples segregated by sex revealed that male GBM cells are significantly more sensitive to BET (bromodomain and extraterminal) inhibitors than are female cells. Thus, Brd4 activity is revealed to drive sex differences in stem cell and tumorigenic phenotypes, which can be abrogated by sex-specific responses to BET inhibition. This has important implications for the clinical evaluation and use of BET inhibitors.
- Published
- 2021
- Full Text
- View/download PDF
24. PI3Kγ inhibition suppresses microglia/TAM accumulation in glioblastoma microenvironment to promote exceptional temozolomide response
- Author
-
Tomoyuki Koga, Jianfang Ning, Jun Ma, Anhua Wu, Clark C. Chen, Beibei Xu, Tao Jiang, Gan You, Megan M. Kaneda, Frank B. Furnari, Judith A. Varner, Kunal S. Patel, Ryan M. Shepard, Jeremy N. Rich, Sanjay Dhawan, Ming Li, Andrew S. Venteicher, Aaron L. Sarver, Jie Li, Hua Zhu, and Christopher K. Glass
- Subjects
Male ,Myeloid ,Nude ,Cell ,Drug Resistance ,Inbred C57BL ,PI3Kγ ,microglia/macrophages ,Mice ,Tumor-Associated Macrophages ,Tumor Microenvironment ,Class Ib Phosphatidylinositol 3-Kinase ,2.1 Biological and endogenous factors ,Aetiology ,Phosphoinositide-3 Kinase Inhibitors ,Cancer ,Tumor ,Multidisciplinary ,Microglia ,Brain Neoplasms ,Chemistry ,Biological Sciences ,Interleukin-11 ,Interleukin 11 ,medicine.anatomical_structure ,Female ,Stem cell ,Signal Transduction ,medicine.drug ,Adult ,Mice, Nude ,Cell Line ,Rare Diseases ,Cell Line, Tumor ,Temozolomide ,Genetics ,medicine ,Animals ,Humans ,Secretion ,Tumor microenvironment ,glioblastoma ,Neurosciences ,Brain Disorders ,Mice, Inbred C57BL ,Brain Cancer ,exceptional responders ,Drug Resistance, Neoplasm ,PI3K gamma ,Cancer research ,Neoplasm ,IL11 ,Phosphatidylinositol 3-Kinase ,Glioblastoma - Abstract
Precision medicine in oncology leverages clinical observations of exceptional response. Toward an understanding of the molecular features that define this response, we applied an integrated, multiplatform analysis of RNA profiles derived from clinically annotated glioblastoma samples. This analysis suggested that specimens from exceptional responders are characterized by decreased accumulation of microglia/macrophages in the glioblastoma microenvironment. Glioblastoma-associated microglia/macrophages secreted interleukin 11 (IL11) to activate STAT3-MYC signaling in glioblastoma cells. This signaling induced stem cell states that confer enhanced tumorigenicity and resistance to the standard-of-care chemotherapy, temozolomide (TMZ). Targeting a myeloid cell restricted an isoform of phosphoinositide-3-kinase, phosphoinositide-3-kinase gamma isoform (PI3Kγ), by pharmacologic inhibition or genetic inactivation disrupted this signaling axis by reducing microglia/macrophage-associated IL11 secretion in the tumor microenvironment. Mirroring the clinical outcomes of exceptional responders, PI3Kγ inhibition synergistically enhanced the anti-neoplastic effects of TMZ in orthotopic murine glioblastoma models. Moreover, inhibition or genetic inactivation of PI3Kγ in murine glioblastoma models recapitulated expression profiles observed in clinical specimens isolated from exceptional responders. Our results suggest key contributions from tumor-associated microglia/macrophages in exceptional responses and highlight the translational potential for PI3Kγ inhibition as a glioblastoma therapy.
- Published
- 2021
- Full Text
- View/download PDF
25. Tumor cells upregulate normoxic HIF-1α in response to doxorubicin
- Author
-
Yiting Cao, Jeremy N. Rich, Mark W. Dewhirst, Joseph M. Eble, Gabi Hanna, Douglas H. Weitzel, Chelsea D. Landon, Hong Yuan, James M. Provenzale, Charleen Yu Chih Nien, and Ejung Moon
- Subjects
Cancer Research ,medicine.medical_specialty ,Angiogenesis ,Cellular homeostasis ,Mice, Nude ,Nitric Oxide Synthase Type II ,Breast Neoplasms ,Nitric Oxide ,Article ,Mice ,Random Allocation ,Downregulation and upregulation ,Internal medicine ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Doxorubicin ,STAT1 ,Antibiotics, Antineoplastic ,biology ,Mammary Neoplasms, Experimental ,Combination chemotherapy ,Hypoxia-Inducible Factor 1, alpha Subunit ,Up-Regulation ,Endocrinology ,STAT1 Transcription Factor ,Oncology ,Cell culture ,Cancer cell ,biology.protein ,Cancer research ,MCF-7 Cells ,Female ,medicine.drug ,Transcription Factors - Abstract
Hypoxia-inducible factor 1 (HIF-1) is a master transcription factor that controls cellular homeostasis. Although its activation benefits normal tissue, HIF-1 activation in tumors is a major risk factor for angiogenesis, therapeutic resistance, and poor prognosis. HIF-1 activity is usually suppressed under normoxic conditions because of rapid oxygen-dependent degradation of HIF-1α. Here, we show that, under normoxic conditions, HIF-1α is upregulated in tumor cells in response to doxorubicin, a chemotherapeutic agent used to treat many cancers. In addition, doxorubicin enhanced VEGF secretion by normoxic tumor cells and stimulated tumor angiogenesis. Doxorubicin-induced accumulation of HIF-1α in normoxic cells was caused by increased expression and activation of STAT1, the activation of which stimulated expression of iNOS and its synthesis of nitric oxide (NO) in tumor cells. Mechanistic investigations established that blocking NO synthesis or STAT1 activation was sufficient to attenuate the HIF-1α accumulation induced by doxorubicin in normoxic cancer cells. To our knowledge, this is the first report that a chemotherapeutic drug can induce HIF-1α accumulation in normoxic cells, an efficacy-limiting activity. Our results argue that HIF-1α-targeting strategies may enhance doxorubicin efficacy. More generally, they suggest a broader perspective on the design of combination chemotherapy approaches with immediate clinical impact. Cancer Res; 73(20); 6230–42. ©2013 AACR.
- Published
- 2021
26. Targeting glioblastoma signaling and metabolism with a re-purposed brain-penetrant drug
- Author
-
Alex Reed, Ryan C. Gimple, Derek A. Wainwright, Timothy F. Cloughesy, Atif Ali Khan, Jeremy N. Rich, Harley I. Kornblum, Tomoyuki Koga, Huijun Yang, Jun Tang, Frank B. Furnari, Junfeng Bi, Benjamin F. Cravatt, Yuchao Gu, Paul S. Mischel, Oswald Quehenberger, Ivy Tsz-Lo Wong, Hui Jing, Shunichiro Miki, Briana C. Prager, Aaron M. Armando, Andrey Rzhetsky, Ellis J. Curtis, Sihan Wu, and Wei Zhang
- Subjects
chemistry.chemical_compound ,Antineoplastic Combined Chemotherapy Protocols ,Tumor Cells, Cultured ,Electronic Health Records ,Epidermal growth factor receptor ,Membrane lipids ,Biology (General) ,education.field_of_study ,biology ,Brain Neoplasms ,Sphingomyelins ,Tumor Burden ,ErbB Receptors ,Sphingomyelin Phosphodiesterase ,Blood-Brain Barrier ,sphingolipid metabolism ,Antidepressant ,Sphingomyelin phosphodiesterase 1 ,Female ,EGFR signaling ,medicine.drug ,Signal Transduction ,Ceramide ,Combination therapy ,QH301-705.5 ,Serotonin reuptake inhibitor ,Mice, Nude ,Antineoplastic Agents ,General Biochemistry, Genetics and Molecular Biology ,Permeability ,Cell Line, Tumor ,Fluoxetine ,medicine ,Temozolomide ,Animals ,Humans ,SMPD1 ,education ,Retrospective Studies ,business.industry ,Drug Repositioning ,Xenograft Model Antitumor Assays ,nervous system diseases ,chemistry ,Cancer research ,biology.protein ,business ,Energy Metabolism ,Glioblastoma - Abstract
Summary The highly lethal brain cancer glioblastoma (GBM) poses a daunting challenge because the blood-brain barrier renders potentially druggable amplified or mutated oncoproteins relatively inaccessible. Here, we identify sphingomyelin phosphodiesterase 1 (SMPD1), an enzyme that regulates the conversion of sphingomyelin to ceramide, as an actionable drug target in GBM. We show that the highly brain-penetrant antidepressant fluoxetine potently inhibits SMPD1 activity, killing GBMs, through inhibition of epidermal growth factor receptor (EGFR) signaling and via activation of lysosomal stress. Combining fluoxetine with temozolomide, a standard of care for GBM, causes massive increases in GBM cell death and complete tumor regression in mice. Incorporation of real-world evidence from electronic medical records from insurance databases reveals significantly increased survival in GBM patients treated with fluoxetine, which was not seen in patients treated with other selective serotonin reuptake inhibitor (SSRI) antidepressants. These results nominate the repurposing of fluoxetine as a potentially safe and promising therapy for patients with GBM and suggest prospective randomized clinical trials.
- Published
- 2021
27. STEM-14. THE WRAD COMPLEX REPRESENTS A THERAPEUTIC TARGET FOR CANCER STEM CELLS IN GLIOBLASTOMA
- Author
-
Sadie Johnson, Rachel Schafer, Shaun Stauffer, Jonathan Macdonald, Gustavo Roversi, Justin D. Lathia, Joseph Alvarado, Erin E. Mulkearns-Hubert, Anjali Kashyap, Jeremy N. Rich, Adam Lauko, Christopher Goins, Kristen Kay, Daniel J. Silver, Kelly Mitchell, Steven Martinez, and Christopher G. Hubert
- Subjects
Cancer Research ,Standard of care ,Growth retardation ,business.industry ,Cell cycle quiescence ,Tumor initiation ,26th Annual Meeting & Education Day of the Society for Neuro-Oncology ,medicine.disease ,Oncology ,Cancer stem cell ,Cancer research ,Medicine ,Neurology (clinical) ,Epigenetics ,Stem cell ,business ,Glioblastoma - Abstract
Glioblastoma (GBM) progression and resistance to conventional therapies is driven in part by cells within the tumor with stem cell properties including quiescence, self-renewal and drug efflux potential. It is thought that eliminating these cancer stem cells (CSCs) is a key component to successful clinical management of GBM. However, currently, few known molecular mechanisms driving CSCs can be exploited for therapeutic development. Core transcription factors such as SOX2, OLIG2, OCT4 and NANOG maintain the CSC state in GBM. Our laboratory recently uncovered a self-renewal signaling axis involving RBBP5 that is necessary and sufficient for CSC maintenance through driving expression of these core stem cell maintenance transcription factors. RBBP5 is a component of the WRAD complex, which promotes Lys4 methylation of histone H3 to positively regulate transcription. We hypothesized that targeting RBBP5 could be a means to disrupt epigenetic programs that maintain CSCs in stemness transcriptional states. We found that genetic and pharmacologic inhibition of the WRAD complex reduced CSC growth, self-renewal and tumor initiation potential. WRAD inhibitors partially dissembled the WRAD complex and reduced H3K4 trimethylation both globally and at the promoters of key stem cell maintenance transcription factors. Using a CSC reporter system, we demonstrated that WRAD complex inhibition decreased growth of SOX2/OCT4 expressing CSCs in a concentration-dependent manner as quantified by live imaging. Overall, our studies assess the function of the WRAD complex and the effect of WRAD complex inhibitors in preclinical models and specifically on the stem cell state for the first time in GBM. Studying the functions of the WRAD complex in CSCs may improve understanding of GBM pathogenesis and elucidate how CSCs survive despite aggressive chemotherapy and radiation. Our ongoing studies aim to develop brain penetrant inhibitors targeting the WRAD complex as an anti-CSC strategy that could potentially synergize with standard of care treatments.
- Published
- 2021
- Full Text
- View/download PDF
28. Plasma cells shape the mesenchymal identity of ovarian cancers through transfer of exosome-derived microRNAs
- Author
-
Shengtao Zhou, Wei Wang, Linjie Zhao, Ryan C. Gimple, Yuan Wang, Xin Wang, Lian Xu, Jeremy N. Rich, and Zhengnan Yang
- Subjects
endocrine system diseases ,Plasma Cells ,Plasma cell ,Exosomes ,Exosome ,03 medical and health sciences ,0302 clinical medicine ,microRNA ,medicine ,Humans ,Research Articles ,030304 developmental biology ,Cancer ,Cell Proliferation ,Ovarian Neoplasms ,0303 health sciences ,Multidisciplinary ,Bortezomib ,Chemistry ,Mesenchymal stem cell ,SciAdv r-articles ,Mesenchymal Stem Cells ,medicine.disease ,In vitro ,Microvesicles ,MicroRNAs ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Cancer research ,Female ,Ovarian cancer ,medicine.drug ,Research Article - Abstract
Plasma cells shape the mesenchymal identity of ovarian cancers through exosome-mediated miR-330-3p/JAM2 axis., Ovarian cancer represents a highly lethal disease that poses a substantial burden for females, with four main molecular subtypes carrying distinct clinical outcomes. Here, we demonstrated that plasma cells, a subset of antibody-producing B cells, were enriched in the mesenchymal subtype of high-grade serous ovarian cancers (HGSCs). Plasma cell abundance correlated with the density of mesenchymal cells in clinical specimens of HGSCs. Coculture of nonmesenchymal ovarian cancer cells and plasma cells induced a mesenchymal phenotype of tumor cells in vitro and in vivo. Phenotypic switch was mediated by the transfer of plasma cell–derived exosomes containing miR-330-3p into nonmesenchymal ovarian cancer cells. Exosome-derived miR-330-3p increased expression of junctional adhesion molecule B in a noncanonical fashion. Depletion of plasma cells by bortezomib reversed the mesenchymal characteristics of ovarian cancer and inhibited in vivo tumor growth. Collectively, our work suggests targeting plasma cells may be a novel approach for ovarian cancer therapy.
- Published
- 2021
29. Zika virus oncolytic activity requires CD8+ T cells and is boosted by immune checkpoint blockade
- Author
-
Tong Hu, Luciano Mazzoccoli, Jeremy N. Rich, Sachendra S. Bais, Michael S. Diamond, Milan G. Chheda, Hideho Okada, Pei Yong Shi, Arijita Jash, Jennifer Govero, Camila R. Fontes-Garfias, Sujan Shresta, Sharmila Nair, and Chao Shan
- Subjects
0301 basic medicine ,medicine.medical_treatment ,Cancer immunotherapy ,CD8-Positive T-Lymphocytes ,Inbred C57BL ,Brain cancer ,Mice ,0302 clinical medicine ,Stem Cell Research - Nonembryonic - Human ,Neoplasms ,Tumor Microenvironment ,Cytotoxic T cell ,Medicine ,Immune Checkpoint Inhibitors ,Cancer ,Oncolytic Virotherapy ,Tumor ,Brain Neoplasms ,General Medicine ,Combined Modality Therapy ,Oncolytic Viruses ,Oncology ,5.1 Pharmaceuticals ,030220 oncology & carcinogenesis ,Female ,Stem Cell Research - Nonembryonic - Non-Human ,Stem cell ,Development of treatments and therapeutic interventions ,Research Article ,Immunology ,Cell Line ,Vaccine Related ,03 medical and health sciences ,Experimental ,Rare Diseases ,Cell Line, Tumor ,Animals ,Humans ,Tumor microenvironment ,business.industry ,Neurosciences ,Immunotherapy ,Neoplasms, Experimental ,Zika Virus ,Stem Cell Research ,Immune checkpoint ,Oncolytic virus ,Brain Disorders ,Mice, Inbred C57BL ,030104 developmental biology ,Orphan Drug ,Good Health and Well Being ,Cancer research ,Immunization ,business ,Glioblastoma ,CD8 - Abstract
Glioblastoma multiforme (GBM) is a fatal human cancer in part because GBM stem cells are resistant to therapy and recurrence is inevitable. Previously, we demonstrated Zika virus (ZIKV) targets GBM stem cells and prevents death of mice with gliomas. Here, we evaluated the immunological basis of ZIKV-mediated protection against GBM. Introduction of ZIKV into the brain tumor increased recruitment of CD8+ T and myeloid cells to the tumor microenvironment. CD8+ T cells were required for ZIKV-dependent tumor clearance because survival benefits were lost with CD8+ T cell depletion. Moreover, while anti-PD-1 antibody monotherapy moderately improved tumor survival, when coadministered with ZIKV, survival increased. ZIKV-mediated tumor clearance also resulted in durable protection against syngeneic tumor rechallenge, which also depended on CD8+ T cells. To address safety concerns, we generated an immune-sensitized ZIKV strain, which was effective alone or in combination with immunotherapy. Thus, oncolytic ZIKV treatment can be leveraged by immunotherapies, which may prompt combination treatment paradigms for adult patients with GBM.
- Published
- 2021
30. PDGF Signaling Promotes Mitophagy in Glioblastoma Stem Cells Through N 6-Methyladenosine
- Author
-
Qi Xie, Sameer Agnihotri, Cuiqing Zhong, Jeremy N. Rich, Deguan Lv, Linjie Zhao, Deobrat Dixit, Xiuxing Wang, Xiqing Li, Derrick Lee, Briana C. Prager, Ryan C. Gimple, Qiulian Wu, Zhixin Qiu, Jia Z. Shen, Kailin Yang, and Guoxin Zhang
- Subjects
endocrine system ,biology ,Growth factor ,medicine.medical_treatment ,Regulator ,EGR1 ,Cell biology ,Cancer stem cell ,Mitophagy ,medicine ,biology.protein ,Stem cell ,Platelet-derived growth factor receptor ,Optineurin - Abstract
Glioblastomas are lethal primary brain tumors that contain glioblastoma stem cells (GSCs) that promote tumor growth. N6-methyladenosine (m6A) is the most common internal modification of mRNA and maintains critical GSC phenotypes. Here, we interrogated upstream regulation of m6A modifications in GSCs, revealing platelet-derived growth factor (PDGF) induction of m6A levels mediated by the methyltransferase METTL3. PDGF signaling stimulated EGR1 transcription that induced METTL3 to promote GSC proliferation and self-renewal, while targeting the PDGF-METTL3 axis generated mitochondrial dysfunction via mitophagy inhibition by regulating mRNA of the mitophagy regulator optineurin (OPTN) through m6A modification. Forced OPTN expression phenocopied PDGF inhibition and OPTN levels portend longer survival of glioblastoma patients. Collectively, we describe novel upstream regulation of m6A manifesting in mitochondrial regulation to promote maintenance of GSC stemness and proliferation, highlighting PDGF-METTL3-OPTN signaling as a therapeutic target in glioblastoma.
- Published
- 2021
- Full Text
- View/download PDF
31. Zika virus is transmitted in neural progenitor cells via cell-to-cell spread and infection is inhibited by the autophagy inducer trehalose
- Author
-
Alex E. Clark, Deborah H. Spector, Gene W. Yeo, Florian Krach, Zhe Zhu, and Jeremy N. Rich
- Subjects
Human cytomegalovirus ,0303 health sciences ,Cell type ,biology ,Immunology ,Autophagy ,medicine.disease ,biology.organism_classification ,Microbiology ,Virology ,Neural stem cell ,Virus ,Zika virus ,03 medical and health sciences ,0302 clinical medicine ,Viral replication ,Insect Science ,Vaccines and Antiviral Agents ,medicine ,biology.protein ,Neutralizing antibody ,030217 neurology & neurosurgery ,030304 developmental biology - Abstract
Zika virus (ZIKV) is a mosquito-borne human pathogen that causes congenital Zika syndrome and neurological symptoms in some adults. There are currently no approved treatments or vaccines for ZIKV, and exploration of therapies targeting host processes could avoid viral development of drug resistance. The purpose of our study was to determine if the nontoxic and widely used disaccharide trehalose, which showed antiviral activity against human cytomegalovirus (HCMV) in our previous work, could restrict ZIKV infection in clinically relevant neural progenitor cells (NPCs). Trehalose is known to induce autophagy, the degradation and recycling of cellular components. Whether autophagy is proviral or antiviral for ZIKV is controversial and depends on cell type and specific conditions used to activate or inhibit autophagy. We show here that trehalose treatment of NPCs infected with recent ZIKV isolates from Panama and Puerto Rico significantly reduces viral replication and spread. In addition, we demonstrate that ZIKV infection in NPCs spreads primarily cell-to-cell as an expanding infectious center, and NPCs are infected via contact with infected cells far more efficiently than by cell-free virus. Importantly, ZIKV was able to spread in NPCs in the presence of neutralizing antibody. IMPORTANCE Zika virus causes birth defects and can lead to neurological disease in adults. While infection rates are currently low, Zika virus (ZIKV) remains a public health concern with no treatment or vaccine available. Targeting a cellular pathway to inhibit viral replication is a potential treatment strategy that avoids development of antiviral resistance. We demonstrate in this study that the nontoxic autophagy-inducing disaccharide trehalose reduces spread and output of ZIKV in infected neural progenitor cells (NPCs), the major cells infected in the fetus. We show that ZIKV spreads cell-to-cell in NPCs as an infectious center and that NPCs are more permissive to infection by contact with infected cells than by cell-free virus. We find that neutralizing antibody does not prevent the spread of the infection in NPCs. These results are significant in demonstrating anti-ZIKV activity of trehalose and in clarifying the primary means of Zika virus spread in clinically relevant target cells.
- Published
- 2020
32. Meningioma epigenetic grouping reveals biologic drivers and therapeutic vulnerabilities
- Author
-
Harish N. Vasudevan, Javier Villanueva-Meyer, Kyounghee Seo, Zhixin Qiu, Michael Martin, David A. Solomon, David R. Raleigh, N. Butowski, Tai-Chung Lam, Abrar Choudhury, Michael W. McDermott, Gilberto Kai-Kit Leung, Arie Perry, Penny K. Sneed, Stephen T. Magill, Briana C. Prager, Siyuan Liu, Jessica Schulte, Matthew S. Susko, Calixto-Hope G Lucas, Jeremy N. Rich, Jason Y. Chang, Joseph F. Costello, Charlotte Eaton, Joanna J. Phillips, Steve Braunstein, Michael Zhang, Jenny Kan-Suen Pu, Nancy Ann Oberheim Bush, Lai-Fung Li, Mitchell S. Berger, and William C. Chen
- Subjects
business.industry ,Central nervous system ,Hla expression ,Cell cycle ,medicine.disease ,nervous system diseases ,Merlin (protein) ,Transcriptome ,Meningioma ,Lymphatic system ,medicine.anatomical_structure ,otorhinolaryngologic diseases ,Cancer research ,Medicine ,Epigenetics ,business ,neoplasms - Abstract
Meningiomas arising from the meningothelial central nervous system lining are the most common primary intracranial tumors, and a significant cause of neurologic morbidity and mortality1. There are no effective medical therapies for meningioma patients2,3, and new treatments have been encumbered by limited understanding of meningioma biology. DNA methylation profiling provides robust classification of central nervous system tumors4, and can elucidate targets for molecular therapy5. Here we use DNA methylation profiling on 565 meningiomas integrated with genetic, transcriptomic, biochemical, and single-cell approaches to show meningiomas are comprised of 3 epigenetic groups with distinct clinical outcomes and biological features informing new treatments for meningioma patients. Merlin-intact meningiomas (group A, 34%) have the best outcomes and are distinguished by a novel apoptotic tumor suppressor function of NF2/Merlin. Immune-enriched meningiomas (group B, 38%) have intermediate outcomes and are distinguished by immune cell infiltration, HLA expression, and lymphatic vessels. Hypermitotic meningiomas (group C, 28%) have the worst outcomes and are distinguished by convergent genetic mechanisms misactivating the cell cycle. Consistently, we find cell cycle inhibitors block meningioma growth in cell culture, organoids, xenografts, and patients. Our results establish a framework for understanding meningioma biology, and provide preclinical rationale for new therapies to treat meningioma patients.
- Published
- 2020
- Full Text
- View/download PDF
33. Zika Virus Targets Glioblastoma Stem Cells Through a SOX2-Integrin αvβ5 Axis
- Author
-
Zhe Zhu, Pinar Mesci, Jeremy N. Rich, Sanjay Dhawan, and Clark C. Chen
- Subjects
biology ,business.industry ,Integrin ,biology.organism_classification ,medicine.disease ,Chromatin ,Zika virus ,Oncolytic virus ,SOX2 ,Cancer research ,medicine ,biology.protein ,Gene silencing ,Surgery ,Neurology (clinical) ,Stem cell ,business ,Glioblastoma - Published
- 2020
- Full Text
- View/download PDF
34. STEM-04. PLATELETS DRIVE GLIOBLASTOMA ONCOGENESIS BY ENHANCING THE GLIOMA STEM CELL PHENOTYPE
- Author
-
Peggy L.R. Harris, John Willis, Alankrita Raghavan, Jeremy N. Rich, Anthony Sloan, Jill S. Barnholtz-Sloan, Amber Kerstetter-Fogle, Theresa A Elder, Eckhard Jankowsky, Gino Cioffi, Christine Lee-Poturalski, and Andrew E Sloan
- Subjects
Cancer Research ,endocrine system ,fungi ,Biology ,medicine.disease ,medicine.disease_cause ,Oncology ,Glioma ,Cancer Stem Cells ,medicine ,Cancer research ,Platelet ,Neurology (clinical) ,Carcinogenesis ,Stem cell phenotype ,Glioblastoma - Abstract
Glioblastoma (GBM) is recognized as one of the deadliest forms of cancer, despite aggressive therapy consisting of maximal surgical resection followed by concurrent radiation and chemotherapy, the median survival remains ~12 months. Glioma stem cells (GSCs) possess potent tumor-initiating properties and comprise a cellular hierarchy that is responsible for treatment resistance and progression. Specifically targeting GSCs has been considered a promising therapeutic approach, however no clear method has been identified. Histologically, it is known that GSCs are found in perivascular and pseudsopalisading regions of GBM. Similarly, platelet aggregates are often found in pseudsopalisading necrotic regions, suggesting a potential interaction between platelets and GSCs due to their spatial locations. High platelet counts have been associated with poor clinical outcome in many cancers including ovarian and endometrial cancer. While platelets are known to affect progression of other tumors, mechanisms by which platelets influence GBM oncogenesis are unknown. Our work aimed to understand the crosstalk between GSCs and platelets within GBM solid tumors that work to enhance disease progression and treatment resistance. Our clinical studies suggest elevated platelet counts positively correlate with tumor growth and negatively correlate to overall patient survival. We found platelets and GSC co-localization in GBM solid tissue; platelet exposure to GSCs results in increased proliferation of GSCs specifically, by increasing the self-renewing capacity of GSCs in a dose dependent manner, and resulted in an increased “Stem-like” transcriptional pattern. Inhibiting the GSC-platelet interaction results in a decrease in GSC renewal and stemness. These results introduce a novel interaction between GSCs and platelets and elucidate a novel therapeutic approach specifically targeting GSCs by disrupting the GSC-platelet interaction.
- Published
- 2020
35. EPCO-36. GENOMIC INSTABILITY AND TRANSCRIPTOMIC SIGNATURES UNDERLYING EPIGENETIC MENINGIOMA SUBGROUPS REVEALS MECHANISMS OF IMMUNE INFILTRATION AND THERAPEUTIC VULNERABILITIES
- Author
-
Geno Guerra, Stephen S. Francis, Lai Fung Li, Steve Braunstein, Matthew S. Susko, David R. Raleigh, Jake Wendt, Jenny Kan-Suen Pu, Gerald Leung, Tai-Chung Lam, Penny K. Sneed, Jason Chan, Michael W. McDermott, Harish N. Vasudevan, Briana C. Prager, Javier Villanueva-Meyer, Arie Perry, Calixto-Hope G Lucas, David A. Solomon, Charlotte Eaton, Jeremy N. Rich, Abrar Choudhury, Stephen T. Magill, Mitchel S. Berger, Joseph F. Costello, and Nancy Ann Oberheim Bush
- Subjects
Genome instability ,Cancer Research ,Computational biology ,Biology ,medicine.disease ,nervous system diseases ,(Epi)Genetics and Computational Omics ,Transcriptome ,Meningioma ,Oncology ,Immune infiltration ,medicine ,otorhinolaryngologic diseases ,Neurology (clinical) ,Epigenetics ,neoplasms - Abstract
BACKGROUND Meningioma treatments are limited due to incomplete understanding of meningioma biology. To address this, we performed multiplatform molecular profiling on 565 meningiomas with comprehensive clinical data to define genomic drivers and identify therapeutic vulnerabilities. METHODS DNA methylation profiling was performed on meningiomas from UCSF (n=200, discovery) and Hong Kong University (n=365, validation). Median follow-up was 5.6 years, and there were 388/142/35 WHO grade I/II/III meningiomas. Copy number variants (CNVs) were calculated for all meningiomas, and RNA sequencing was performed on UCSF meningiomas. Cell type deconvolution, metagenomics, CRISPR, and pharmacology were used for mechanistic and functional validation. RESULTS Unsupervised hierarchical clustering of differentially methylated DNA probes revealed that meningiomas were comprised of 3 epigenetic subgroups associated with good, intermediate, and poor outcomes, with representation from all WHO grades in each subgroup. Meningiomas from the subgroup with the best outcomes (52% WHO grade I) were distinguished by recurrent gain of Chr5. Meningiomas from the subgroup with intermediate outcomes (31% WHO grade II) were distinguished by genomic stability, enrichment of innate immune genes, and immune infiltration in the setting of endogenous retroviral gene re-expression, a mechanism of immune recruitment. The most aggressive subgroup of meningiomas (57% WHO grade III) was distinguished by genomic instability, including recurrent loss of Chr22q harboring NF2, and decreased immune infiltration. Consistently, NF2 suppression in primary meningioma cells derived from immunogenic meningiomas decreased expression of innate immune genes critical for immune recruitment, suggesting a novel immunostimulatory function of NF2. The most aggressive subgroup of meningiomas were further distinguished by activation of the mitogenic FOXM1 transcriptional program, and recurrent loss of Chr9p harboring CDKN2A/B, which rendered primary meningioma cells from this subgroup susceptible to CDK4/6 inhibitors. CONCLUSIONS Meningiomas are comprised of 3 epigenetic subgroups defined by genetic mechanisms driving immune infiltration in the tumor microenvironment and meningioma cell proliferation.
- Published
- 2020
36. IMMU-43. ZIKA VIRUS TO TREAT GLIOMA: TURNING COLD TUMORS HOT
- Author
-
Pei Yong Shi, Michael P. Diamond, Luciano Mazzoccoli, Sujan Shresta, Sharmila Nair, Milan G. Chheda, Jeremy N. Rich, Tong Hu, Arijita Jash, and Jennifer Govero
- Subjects
Cancer Research ,Oncology ,biology ,business.industry ,Glioma ,Immunology ,medicine ,Neurology (clinical) ,medicine.disease ,biology.organism_classification ,business ,Virology ,Zika virus - Abstract
GBM is a hallmark example of an immunotherapy-resistant tumor. Previously, we discovered that Zika virus (ZIKV) has a natural honing capacity to specifically kill GBM stem cells (GSCs). Using two syngeneic glioma models, GL261 and CT2A, we found that in vivo intratumoral ZIKV treatment reduces tumor size and extends median survival far beyond that expected for its anti-GSC effects; additionally, approximately 50% of mice were long-term survivors. We now report that ZIKV treatment is associated with an increase in tumor-specific CD8+ T cells and activated myeloid cells expressing MHC-II and NOS2. Treatment with CD8-depleting antibody abrogates the survival advantage conferred by ZIKV, demonstrating that CD8+ T cells are required for ZIKV efficacy. Remarkably, ZIKV treatment also confers protection against tumor re-challenge 1.5 years after the original treatment, and this too is dependent on CD8+ T cells. Lastly, in a bilateral tumor implantation model, intratumoral ZIKV treatment of one tumor increases overall survival, suggesting ZIKV is efficacious because it generates a global anti-tumor response. Building on the observation that ZIKV treatment increases CD8+ T cells in the tumor bed, we combined ZIKV with α-PD1 immunotherapy. We found survival was dramatically increased compared to either treatment alone. Finally, to improve safety of potential ZIKV treatment for patients, we generated an immune-sensitized virus. This strain remains efficacious as a single agent and in combination with immunotherapy in vivo. Overall, our results suggest ZIKV treatment addresses two key problems in our current treatment of GBM: it specifically targets highly resistant GSC and initiates a potent anti-tumor immune response against the remaining tumor. Our work reveals the mechanistic basis of ZIKV efficacy and demonstrates the potential for ZIKV as a therapy for GBM.
- Published
- 2020
37. STEM-12. ZIKA VIRUS TARGETS GLIOBLASTOMA STEM CELLS THROUGH A SOX2-INTEGRIN α (v)β (5) AXIS
- Author
-
Clark C. Chen, Jeremy N. Rich, Sanjay Dhawan, Zhe Zhu, and Pinar Mesci
- Subjects
Cancer Research ,biology ,fungi ,Integrin α ,biology.organism_classification ,medicine.disease ,Zika virus ,Oncology ,SOX2 ,Cancer Stem Cells ,medicine ,Cancer research ,Neurology (clinical) ,Stem cell ,Glioblastoma - Abstract
INTRODUCTION Oncolytic virus hold great promise as a platform for glioblastoma therapeutic development. Zika virus (ZIKV) is an oncolytic virus with exquisite selectivity for infecting and killing glioblastoma stem cells (GSCs). Here, we delineate the molecular determinant of this selectivity. METHODS cell-based glioblastoma models, glioblastoma organoid assays, in vivo murine glioblastoma models, ZIKV infectivity assays, gene silencing, ChIP-seq studies. RESULTS In independent models, ZIKV preferentially infected and lysed SOX2+ GSCs. Silencing of SOX2 expression attenuated this preferential infectivity. Of note, ZIKV infection of GSCs was independent of AXL, its putative receptor in normal brain. ChIP-seq experiments revealed that SOX2 bound within the ITGAV locus (encoding the integrin av subunit), and this binding was associated with accumulation of the active chromatin mark H3K27ac. Silencing of SOX2 suppressed ITGAV expression as well as ZIKV infectivity against GSCs, indicating that integrin is required for ZIKV infection. Of integrin b units, only silencing of integrin b5 prevented the killing of GSCs by ZIKV infection, suggesting ZIKV infection required the avb5 integrin. Supporting this hypothesis, blockade of the avb5 integrin substantially reduced ZIKV infection of GSCs in glioblastoma organoid assays and in clinical glioblastoma specimens. Sox2 expression additionally suppress GSC expression of all members of the interferon-stimulated genes (ISG family), thereby suppressing innate anti-viral response to facilitate ZIKV infection. CONCLUSIONS Collectively, our results reveal that ZIKV infection of GSCs is mediated by integrin α vβ 5 leading to SOX2 expression which negatively regulates antiviral immunity thereby facilitating ZIKV infection.
- Published
- 2020
38. Oncogenic 3D genome conformations identify novel therapeutic targets in ependymoma
- Author
-
Katerina Kraft, Robert J. Wechsler-Reya, Abhijit Chakraborty, Jill P. Mesirov, Derek Reid, Michael D. Taylor, Jesse R. Dixon, Frank Buchholz, Rocio Acuna-Hidalgo, Hannah Carter, Kristian W. Pajtler, Sachin Kumar, John R. Crawford, James T. Robinson, Ferhat Ay, Edwin F. Juarez, Stefan Mundlos, Kulandaimanuvel Antony Michaelraj, Konstantin Okonechnikov, Lukas Chavez, Denise M. Malicki, Nicole G. Coufal, Jens-Martin Hübner, Jeremy N. Rich, Till Milde, Matija Snuderl, Stefan M. Pfister, Donglim Esther Park, Owen Chapman, Anthony P. Schmitt, Michael J. Levy, Aylin Camgoz, Sahaana Chandran, Marcel Kool, Rosalind Bump, Shareef Nahas, Monika Mauermann, and Meghana Pagadala
- Subjects
Ependymoma ,Text mining ,business.industry ,medicine ,Computational biology ,Biology ,medicine.disease ,business ,Genome - Abstract
Ependymoma is a tumor of the brain or spinal cord. The two most common and aggressive molecular groups of ependymoma are the supratentorial RELA-fusion associated group and the posterior fossa ependymoma group A. In both groups, tumors occur mainly in young children and frequently recur after treatment1. Although the molecular mechanisms underlying these diseases have recently been uncovered, they remain difficult to target and innovative therapeutic approaches are urgently needed. Here, we use genome-wide chromosome conformation capture (Hi-C), complemented with CTCF (insulators) and H3K27ac (active enhancers) ChIP-seq as well as gene expression and whole-genome DNA methylation profiling in primary and relapsed ependymoma tumors and cell lines to identify chromosomal rearrangements and regulatory mechanisms underlying aberrant expression of genes that are essential for ependymoma tumorigenesis. In particular, we observe the formation of new topologically associating domains (‘neo-TADs’) by intra- and inter-chromosomal structural variants, tumor-specific 3D chromatin complexes of regulatory elements, and the replacement of CTCF insulators by DNA hyper-methylation as novel oncogenic mechanisms in ependymoma. Through inhibition experiments we validated that the newly identified genes, including RCOR2, ITGA6, LAMC1, and ARL4C, are highly essential for the survival of patient-derived ependymoma models in a disease subtype-specific manner. Thus, this study identifies potential novel therapeutic vulnerabilities in ependymoma and extends our ability to reveal tumor-dependency genes and pathways by oncogenic 3D genome conformations even in tumors that lack known genetic alterations.
- Published
- 2020
- Full Text
- View/download PDF
39. Treatment of glioblastoma using multicomponent silica nanoparticles
- Author
-
Kathleen Tong, Taylor Moon, Pubudu M. Peiris, Aaron Yun, S. Raghunathan, Youngjun Park, Vindya S. Perera, Gil Covarrubias, Morgan E Lorkowski, Mark A. Griswold, Oguz Turan, Deobrat Dixit, Jeremy N. Rich, Georgia Loutrianakis, Efstathios Karathanasis, Shane Cooley, Peter Bielecki, and Ketan B. Ghaghada
- Subjects
Pharmacology ,Drug ,education.field_of_study ,biology ,Chemistry ,media_common.quotation_subject ,Biochemistry (medical) ,Population ,Brain tumor ,Pharmaceutical Science ,Medicine (miscellaneous) ,Nanoparticle ,Mesoporous silica ,medicine.disease ,Article ,Nitric oxide synthase ,In vivo ,Drug delivery ,medicine ,biology.protein ,Cancer research ,Pharmacology (medical) ,education ,Genetics (clinical) ,media_common - Abstract
Glioblastomas (GBMs) remain highly lethal. This partially stems from the presence of brain tumor initiating cells (BTICs), a highly plastic cellular subpopulation that is resistant to current therapies. In addition to resistance, the blood-brain barrier limits the penetration of most drugs into GBMs. To effectively deliver a BTIC-specific inhibitor to brain tumors, we developed a multicomponent nanoparticle, termed Fe@MSN, which contains a mesoporous silica shell and an iron oxide core. Fibronectin-targeting ligands directed the nanoparticle to the near-perivascular areas of GBM. After Fe@MSN particles deposited in the tumor, an external low-power radiofrequency (RF) field triggered rapid drug release due to mechanical tumbling of the particle resulting in penetration of high amounts of drug across the blood-brain tumor interface and widespread drug delivery into the GBM. We loaded the nanoparticle with the drug 1400W, which is a potent inhibitor of the inducible nitric oxide synthase (iNOS). It has been shown that iNOS is preferentially expressed in BTICs and is required for their maintenance. Using the 1400W-loaded Fe@MSN and RF-triggered release, in vivo studies indicated that the treatment disrupted the BTIC population in hypoxic niches, suppressed tumor growth and significantly increased survival in BTIC-derived GBM xenografts.
- Published
- 2020
40. A Randomized Trial of a Multifactorial Strategy to Prevent Serious Fall Injuries
- Author
-
Thomas M. Gill, Peter Peduzzi, Rich Eder, Denise Esserman, Susan L. Greenspan, Heather G. Allore, Lori Goehring, Dorothy I. Baker, Robert B. Wallace, David A. Ganz, Thomas G. Travison, Martha B. Carnie, Jerry H. Gurwitz, Neil B. Alexander, Siobhan K McMahon, Charles Lu, David B. Reuben, Can Meng, Joanne M. McGloin, Fred C. Ko, Patricia C. Dykes, Katy L. B. Araujo, Michael E. Miller, Carri Casteel, Peter Charpentier, Jay Magaziner, Albert L. Siu, Erich J. Greene, Eleni A. Skokos, Thomas W. Storer, James Dziura, Priscilla K. Gazarian, Nancy K. Latham, Stephen C. Waring, Albert W. Wu, Rosaly Correa-de-Araujo, Jeremy N. Rich, Shalender Bhasin, Shehzad Basaria, Chad Boult, Neil M. Resnick, Elena Volpi, Brooke Brawley, Haseena Rajeevan, Jocelyn Wiggins, Maureen Fagan, and Pamela W. Duncan
- Subjects
Male ,Aging ,Comparative Effectiveness Research ,STRIDE Trial Investigators ,Poison control ,030204 cardiovascular system & hematology ,Suicide prevention ,Medical and Health Sciences ,Occupational safety and health ,law.invention ,0302 clinical medicine ,Randomized controlled trial ,law ,Risk Factors ,80 and over ,030212 general & internal medicine ,Precision Medicine ,Aged, 80 and over ,Accidental Injuries ,Incidence ,food and beverages ,Human factors and ergonomics ,Injuries and accidents ,General Medicine ,Health Services ,fractures ,mobility ,Hospitalization ,Female ,Patient Safety ,Independent Living ,Risk assessment ,medicine.medical_specialty ,Physical Injury - Accidents and Adverse Effects ,Clinical Trials and Supportive Activities ,frailty ,Risk Assessment ,Article ,03 medical and health sciences ,Clinical Research ,General & Internal Medicine ,Injury prevention ,medicine ,Humans ,Intensive care medicine ,Aged ,business.industry ,Prevention ,fungi ,aging ,Patient Care Management ,Good Health and Well Being ,Multicenter study ,Accidental Falls ,business - Abstract
BackgroundInjuries from falls are major contributors to complications and death in older adults. Despite evidence from efficacy trials that many falls can be prevented, rates of falls resulting in injury have not declined.MethodsWe conducted a pragmatic, cluster-randomized trial to evaluate the effectiveness of a multifactorial intervention that included risk assessment and individualized plans, administered by specially trained nurses, to prevent fall injuries. A total of 86 primary care practices across 10 health care systems were randomly assigned to the intervention or to enhanced usual care (the control) (43 practices each). The participants were community-dwelling adults, 70 years of age or older, who were at increased risk for fall injuries. The primary outcome, assessed in a time-to-event analysis, was the first serious fall injury, adjudicated with the use of participant report, electronic health records, and claims data. We hypothesized that the event rate would be lower by 20% in the intervention group than in the control group.ResultsThe demographic and baseline characteristics of the participants were similar in the intervention group (2802 participants) and the control group (2649 participants); the mean age was 80 years, and 62.0% of the participants were women. The rate of a first adjudicated serious fall injury did not differ significantly between the groups, as assessed in a time-to-first-event analysis (events per 100 person-years of follow-up, 4.9 in the intervention group and 5.3 in the control group; hazard ratio, 0.92; 95% confidence interval [CI], 0.80 to 1.06; P = 0.25). The rate of a first participant-reported fall injury was 25.6 events per 100 person-years of follow-up in the intervention group and 28.6 events per 100 person-years of follow-up in the control group (hazard ratio, 0.90; 95% CI, 0.83 to 0.99; P = 0.004). The rates of hospitalization or death were similar in the two groups.ConclusionsA multifactorial intervention, administered by nurses, did not result in a significantly lower rate of a first adjudicated serious fall injury than enhanced usual care. (Funded by the Patient-Centered Outcomes Research Institute and others; STRIDE ClinicalTrials.gov number, NCT02475850.).
- Published
- 2020
41. Dual Role of WISP1 in maintaining glioma stem cells and tumor-supportive macrophages in glioblastoma
- Author
-
Andrew E. Sloan, Xiaoguang Fang, Xiuxing Wang, George R. Stark, Zhi Huang, Haidong Huang, Xiaoxia Li, Wenchao Zhou, Chengwei Chu, Qian Huang, Jeremy N. Rich, Weiwei Tao, Xingjiang Yu, Aili Zhang, Kui Zhai, Qiulian Wu, Shideng Bao, and Jennifer S. Yu
- Subjects
0301 basic medicine ,General Physics and Astronomy ,Kaplan-Meier Estimate ,Mice, SCID ,0302 clinical medicine ,RNA interference ,Mice, Inbred NOD ,Tumor Microenvironment ,lcsh:Science ,Mice, Knockout ,Multidisciplinary ,Brain Neoplasms ,Wnt signaling pathway ,Glioma ,U937 Cells ,030220 oncology & carcinogenesis ,Doxycycline ,Neoplastic Stem Cells ,RNA Interference ,Stem cell ,Signal Transduction ,Cancer microenvironment ,endocrine system ,Science ,Biology ,General Biochemistry, Genetics and Molecular Biology ,Article ,CCN Intercellular Signaling Proteins ,03 medical and health sciences ,Paracrine signalling ,Cell Line, Tumor ,Proto-Oncogene Proteins ,medicine ,Gene silencing ,Animals ,Humans ,Autocrine signalling ,neoplasms ,Cell Proliferation ,Tumor microenvironment ,Macrophages ,fungi ,General Chemistry ,medicine.disease ,Xenograft Model Antitumor Assays ,nervous system diseases ,CNS cancer ,030104 developmental biology ,Cancer research ,lcsh:Q ,Proto-Oncogene Proteins c-akt - Abstract
The interplay between glioma stem cells (GSCs) and the tumor microenvironment plays crucial roles in promoting malignant growth of glioblastoma (GBM), the most lethal brain tumor. However, the molecular mechanisms underlying this crosstalk are incompletely understood. Here, we show that GSCs secrete the Wnt‐induced signaling protein 1 (WISP1) to facilitate a pro-tumor microenvironment by promoting the survival of both GSCs and tumor-associated macrophages (TAMs). WISP1 is preferentially expressed and secreted by GSCs. Silencing WISP1 markedly disrupts GSC maintenance, reduces tumor-supportive TAMs (M2), and potently inhibits GBM growth. WISP1 signals through Integrin α6β1-Akt to maintain GSCs by an autocrine mechanism and M2 TAMs through a paracrine manner. Importantly, inhibition of Wnt/β-catenin-WISP1 signaling by carnosic acid (CA) suppresses GBM tumor growth. Collectively, these data demonstrate that WISP1 plays critical roles in maintaining GSCs and tumor-supportive TAMs in GBM, indicating that targeting Wnt/β-catenin-WISP1 signaling may effectively improve GBM treatment and the patient survival., The tumour microenvironment plays an important role in promoting glioblastoma. Here, the authors show that glioma stem cells secrete WISP1, which promotes both the survival of the stem cells and tumour-associated macrophages.
- Published
- 2020
42. Metabolic Regulation of the Epigenome Drives Lethal Infantile Ependymoma
- Author
-
Samuel Weiss, Leo J.Y. Kim, Xiaochong Wu, Randy Van Ommeren, Yanqing Jiang, Kaitlin Kharas, Evgeny Kanshin, Moloud Ahmadi, Alberto Delaidelli, Geneviève Deblois, David Przelicki, Stephane Angers, Hiromichi Suzuki, Sameer Agnihotri, Bradly G. Wouters, Graham MacLeod, Ricky Tsai, Pasqualino De Antonellis, Michelle Ly, Stacey L. Krumholtz, Paul Guilhamon, James Loukides, Ravi N. Vellanki, Alex Rasnitsyn, Hamza Farooq, Daniel Schramek, Nada Jabado, María Sánchez-Osuna, Laura K. Donovan, Vijay Ramaswamy, Ibrahim El-Hamamy, Joonas Haapasalo, Jeremy N. Rich, Michael D. Taylor, Benjamin A. Garcia, Mike Tyers, Kyle Juraschka, Winnie Ong, Olivier Saulnier, Panagiotis Prinos, John J.Y. Lee, Borja L. Holgado, Olga Sirbu, Craig Daniels, Cheryl H. Arrowsmith, Cory Richman, Poul H. Sorensen, Kulandaimanuvel Antony Michealraj, Sheila K. Singh, Andrea Bajic, Polina Balin, Stephen C. Mack, Betty Luu, Fiona J. Coutinho, Dilakshan Srikanthan, Florence M.G. Cavalli, Sachin Kumar, Evan Y. Wang, Mathieu Lupien, Peter B. Dirks, Maria C. Vladoiu, Lincoln Stein, Livia Garzia, Ahmad Malik, John Wojcik, Avesta Rastan, Michealraj, K. A., Kumar, S. A., Kim, L. J. Y., Cavalli, F. M. G., Przelicki, D., Wojcik, J. B., Delaidelli, A., Bajic, A., Saulnier, O., Macleod, G., Vellanki, R. N., Vladoiu, M. C., Guilhamon, P., Ong, W., Lee, J. J. Y., Jiang, Y., Holgado, B. L., Rasnitsyn, A., Malik, A. A., Tsai, R., Richman, C. M., Juraschka, K., Haapasalo, J., Wang, E. Y., De Antonellis, P., Suzuki, H., Farooq, H., Balin, P., Kharas, K., Van Ommeren, R., Sirbu, O., Rastan, A., Krumholtz, S. L., Ly, M., Ahmadi, M., Deblois, G., Srikanthan, D., Luu, B., Loukides, J., Wu, X., Garzia, L., Ramaswamy, V., Kanshin, E., Sanchez-Osuna, M., El-Hamamy, I., Coutinho, F. J., Prinos, P., Singh, S., Donovan, L. K., Daniels, C., Schramek, D., Tyers, M., Weiss, S., Stein, L. D., Lupien, M., Wouters, B. G., Garcia, B. A., Arrowsmith, C. H., Sorensen, P. H., Angers, S., Jabado, N., Dirks, P. B., Mack, S. C., Agnihotri, S., Rich, J. N., and Taylor, M. D.
- Subjects
Epigenomics ,Ependymoma ,Male ,ependymoma ,Epigenomic ,Somatic cell ,cancer metabolism ,Infratentorial Neoplasms ,Biology ,General Biochemistry, Genetics and Molecular Biology ,Cell Line ,Histones ,Brain Neoplasm ,03 medical and health sciences ,Epigenome ,0302 clinical medicine ,Histone demethylation ,Histone methylation ,medicine ,Animals ,Humans ,Epigenetics ,030304 developmental biology ,hindbrain development ,Cell Proliferation ,Infratentorial Neoplasm ,0303 health sciences ,Brain Neoplasms ,Animal ,Lysine ,Infant ,DNA Methylation ,medicine.disease ,microenvironment ,Mice, Inbred C57BL ,Histone ,Acetylation ,paediatric cancer ,Mutation ,biology.protein ,Cancer research ,030217 neurology & neurosurgery ,epigenetic ,Human - Abstract
Posterior fossa A (PFA) ependymomas are lethal malignancies of the hindbrain in infants and toddlers. Lacking highly recurrent somatic mutations, PFA ependymomas are proposed to be epigenetically driven tumors for which model systems are lacking. Here we demonstrate that PFA ependymomas are maintained under hypoxia, associated with restricted availability of specific metabolites to diminish histone methylation, and increase histone demethylation and acetylation at histone 3 lysine 27 (H3K27). PFA ependymomas initiate from a cell lineage in the first trimester of human development that resides in restricted oxygen. Unlike other ependymomas, transient exposure of PFA cells to ambient oxygen induces irreversible cellular toxicity. PFA tumors exhibit a low basal level of H3K27me3, and, paradoxically, inhibition of H3K27 methylation specifically disrupts PFA tumor growth. Targeting metabolism and/or the epigenome presents a unique opportunity for rational therapy for infants with PFA ependymoma. Hypoxia reprograms the cellular metabolome and epigenome to promote growth of the most lethal ependymomas.
- Published
- 2020
43. Three-dimensional bioprinted glioblastoma microenvironments model cellular dependencies and immune interactions
- Author
-
Jacob Schimelman, Tyler E. Miller, Benjamin F. Cravatt, Alysson R. Muotri, Shruti Bhargava, Zheng Zhong, Deobrat Dixit, Derrick Lee, Shaochen Chen, Xueyi Wan, Bingjie Sun, Michael H. Lorenzini, Qi Xie, Jing Tian, Aaron Yu, Jeremy N. Rich, Reilly L. Kidwell, Zhixin Qiu, Linjie Zhao, Ryan C. Gimple, Hui Jing, Briana C. Prager, Zhe Zhu, Jing Tang, Pengrui Wang, Pinar Mesci, Min Tang, Trevor Tam, and Qiulian Wu
- Subjects
Cancer microenvironment ,Cell type ,Clinical Sciences ,Brain tumor ,Biology ,Article ,Cell Line ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Immune system ,Rare Diseases ,Neural Stem Cells ,Stem Cell Research - Nonembryonic - Human ,Precursor cell ,Cell Line, Tumor ,medicine ,Tumor Microenvironment ,CRISPR ,Animals ,Humans ,Molecular Biology ,030304 developmental biology ,Cancer ,Cell Proliferation ,0303 health sciences ,Tumor ,Microglia ,Tissue Scaffolds ,Cancer stem cells ,Neurosciences ,Bioprinting ,Cell Biology ,medicine.disease ,Stem Cell Research ,Cell biology ,Brain Disorders ,Brain Cancer ,CNS cancer ,Crosstalk (biology) ,medicine.anatomical_structure ,Biochemistry and Cell Biology ,Stem cell ,Glioblastoma ,030217 neurology & neurosurgery ,Biotechnology ,Developmental Biology - Abstract
Brain tumors are dynamic complex ecosystems with multiple cell types. To model the brain tumor microenvironment in a reproducible and scalable system, we developed a rapid three-dimensional (3D) bioprinting method to construct clinically relevant biomimetic tissue models. In recurrent glioblastoma, macrophages/microglia prominently contribute to the tumor mass. To parse the function of macrophages in 3D, we compared the growth of glioblastoma stem cells (GSCs) alone or with astrocytes and neural precursor cells in a hyaluronic acid-rich hydrogel, with or without macrophage. Bioprinted constructs integrating macrophage recapitulate patient-derived transcriptional profiles predictive of patient survival, maintenance of stemness, invasion, and drug resistance. Whole-genome CRISPR screening with bioprinted complex systems identified unique molecular dependencies in GSCs, relative to sphere culture. Multicellular bioprinted models serve as a scalable and physiologic platform to interrogate drug sensitivity, cellular crosstalk, invasion, context-specific functional dependencies, as well as immunologic interactions in a species-matched neural environment.
- Published
- 2019
44. Therapeutic targeting of ependymoma as informed by oncogenic enhancer profiling
- Author
-
Anne Song, Alexander J. Federation, Leo J.Y. Kim, David T.W. Jones, Ana Fernandez Miñan, Laura McDonald, Mathieu Lupien, Susan Q. Ke, Lukas Chavez, Briana C. Prager, Sheila K. Singh, Peter B. Dirks, Borja L. Holgado, Kristian W. Pajtler, Yan Li, Till Milde, Marc Zapatka, Angel M. Carcaboso, Livia Garzia, Xiuxing Wang, Chao Jun Li, Kenneth Aldape, Christine Lee, Ian C. Scott, Xin Wang, Laura K. Donovan, Xiu-Wu Bian, Sylvia Doan, Stephen M. Dombrowski, Betty Luu, Michael D. Taylor, Adam Tropper, Vaidehi Mahadev, James E. Bradner, Ryan C. Gimple, Tyler E. Miller, Serap Erkek, Christopher G. Hubert, Daniel C. Factor, Kulandaimanuvel Antony Michaelraj, Stefan M. Pfister, Kelsey C. Bertrand, Jennifer Zuccaro, Zhiqin Huang, Yuan Yao Thompson, Hendrik Witt, Nada Jabado, Konstantin Okonechnikov, Paul A. Northcott, James J. Morrow, Senthuran Vijayarajah, Jeremy N. Rich, Susanne Gröbner, Andrey Korshunov, Vijay Ramaswamy, Sisi Lai, Stephen C. Mack, Alina Saiakhova, Annie Huang, Claudia L.L. Valentim, James T. Rutka, Eric Bouffet, Xiaochong Wu, Matthias Lienhard, Qiulian Wu, Jüri Reimand, Peter J. Houghton, Andrew R. Morton, Peter C. Scacheri, John J.Y. Lee, Marina Ryzhova, Patrick Sin-Chan, Peter Lichter, Stephen T. Keir, Marcel Kool, Alex's Lemonade Stand Foundation for Childhood Cancer, Cancer Prevention and Research Institute of Texas, Ministry of Science, Technology and Space (Israel), James S. McDonnell Foundation, and National Institutes of Health (US)
- Subjects
0301 basic medicine ,Ependymoma ,Biology ,Small hairpin RNA ,Mice ,03 medical and health sciences ,Cancer epigenetics ,RNA interference ,Cancer genomics ,medicine ,Animals ,Humans ,Gene Regulatory Networks ,Molecular Targeted Therapy ,Precision Medicine ,Enhancer ,Gene ,Regulation of gene expression ,Multidisciplinary ,Base Sequence ,Oncogenes ,medicine.disease ,Xenograft Model Antitumor Assays ,Chromatin ,Gene Expression Regulation, Neoplastic ,CNS cancer ,Enhancer Elements, Genetic ,030104 developmental biology ,Cancer research ,Female ,RNA Interference ,Transcription Factors - Abstract
Genomic sequencing has driven precision-based oncology therapy; however, the genetic drivers of many malignancies remain unknown or non-targetable, so alternative approaches to the identification of therapeutic leads are necessary. Ependymomas are chemotherapy-resistant brain tumours, which, despite genomic sequencing, lack effective molecular targets. Intracranial ependymomas are segregated on the basis of anatomical location (supratentorial region or posterior fossa) and further divided into distinct molecular subgroups that reflect differences in the age of onset, gender predominance and response to therapy1,2,3. The most common and aggressive subgroup, posterior fossa ependymoma group A (PF-EPN-A), occurs in young children and appears to lack recurrent somatic mutations2. Conversely, posterior fossa ependymoma group B (PF-EPN-B) tumours display frequent large-scale copy number gains and losses but have favourable clinical outcomes1,3. More than 70% of supratentorial ependymomas are defined by highly recurrent gene fusions in the NF-κB subunit gene RELA (ST-EPN-RELA), and a smaller number involve fusion of the gene encoding the transcriptional activator YAP1 (ST-EPN-YAP1)1,3,4. Subependymomas, a distinct histologic variant, can also be found within the supratetorial and posterior fossa compartments, and account for the majority of tumours in the molecular subgroups ST-EPN-SE and PF-EPN-SE. Here we describe mapping of active chromatin landscapes in 42 primary ependymomas in two non-overlapping primary ependymoma cohorts, with the goal of identifying essential super-enhancer-associated genes on which tumour cells depend. Enhancer regions revealed putative oncogenes, molecular targets and pathways; inhibition of these targets with small molecule inhibitors or short hairpin RNA diminished the proliferation of patient-derived neurospheres and increased survival in mouse models of ependymomas. Through profiling of transcriptional enhancers, our study provides a framework for target and drug discovery in other cancers that lack known genetic drivers and are therefore difficult to treat., This work was supported by an Alex's Lemonade Stand Young Investigator Award (S.C.M.), The CIHR Banting Fellowship (S.C.M.), The Cancer Prevention Research Institute of Texas (S.C.M., RR170023), Sibylle Assmus Award for Neurooncology (K.W.P.), the DKFZ-MOST (Ministry of Science, Technology & Space, Israel) program in cancer research (H.W.), James S. McDonnell Foundation (J.N.R.) and NIH grants: CA154130 (J.N.R.), R01 CA169117 (J.N.R.), R01 CA171652 (J.N.R.), R01 NS087913 (J.N.R.) and R01 NS089272 (J.N.R.). R.C.G. is supported by NIH grants T32GM00725 and F30CA217065. M.D.T. is supported by The Garron Family Chair in Childhood Cancer Research, and grants from the Pediatric Brain Tumour Foundation, Grand Challenge Award from CureSearch for Children’s Cancer, the National Institutes of Health (R01CA148699, R01CA159859), The Terry Fox Research Institute and Brainchild. M.D.T. is also supported by a Stand Up To Cancer St. Baldrick’s Pediatric Dream Team Translational Research Grant (SU2C-AACR-DT1113).
- Published
- 2017
- Full Text
- View/download PDF
45. STEM-17. THE GLIOMA STEM CELL PLATELET INTERACTION DRIVES GBM ONCOGENESIS IDENTIFYING A NOVEL THERAPEUTIC APPROACH
- Author
-
Amber Kerstetter-Fogle, Eckhard Jankowsky, Andrew E. Sloan, Harry Hoffman, Anirban Sen Gupta, Ansh Desai, Jill S. Barnholtz-Sloan, Anthony Sloan, Jeremy N. Rich, Theresa Elder, Peggy L.R. Harris, Christine Lee-Poturalski, and Gino Cioffi
- Subjects
endocrine system ,Cancer Research ,Cancer ,Biology ,medicine.disease_cause ,medicine.disease ,Therapeutic approach ,Oncology ,Cell culture ,Glioma ,medicine ,Cancer research ,Platelet ,Neurology (clinical) ,Platelet activation ,Stem cell ,Carcinogenesis - Abstract
The effect of platelets on oncogenesis has been studied extensively in cancer metastasis, but not in glioblastoma (GBM), where metastasis is rare. Here we identify the unique crosstalk between glioma stem cells (GSCs) and platelets within GBM solid tumors that enhance disease progression. Targeting GSCs is considered a promising therapeutic approach; however, no clear method has been identified. High platelet counts have been associated with poor clinical outcome in many cancers including ovarian and endometrial cancer. While platelets are known to affect progression of other tumors, mechanisms by which platelets influence GBM oncogenesis are unknown. Immunofluorescence, qPCR, and western blot were used to evaluate the presence of GSCs and platelets and their colocalization in GBM patient tissue at University Hospitals-Seidman Cancer Center. Functional assays followed by RNA sequencing were conducted to determine the functional effect of healthy and GBM platelets on growth of patient derived, autologous GSCs. Our clinical studies suggest elevated platelet counts positively correlate with GSC proliferation and negatively correlate with overall survival in patients with GBM. Patients with high platelet counts ( >350k/µl) had a median survival time of 9 months compared to 16 months median survival for patients with normal platelet count (150-350/µl) (p
- Published
- 2021
- Full Text
- View/download PDF
46. Constitutive Ras signaling and Ink4a/Arf inactivation cooperate during the development of B-ALL in mice
- Author
-
Jessie Xiong, Brian Lawney, Sanjay S. Patel, David M. Weinstock, Charles G. Mullighan, Peter S. Dennis, Zeyuan Song, Ruiyang Liu, Madeleine E. Lemieux, Ahmad Alduaij, Ruben D. Carrasco, Meng Jiang, Kumar Sukhdeo, Yunyu Zhang, Norman E. Sharpless, Kathryn G. Roberts, Geraldine S. Pinkus, Tomasz Sewastianik, Yue Kang, and Jeremy N. Rich
- Subjects
0301 basic medicine ,Somatic cell ,Hematology ,Biology ,medicine.disease ,medicine.disease_cause ,Phenotype ,Lymphoplasmacytic Lymphoma ,Lymphoma ,Pathogenesis ,03 medical and health sciences ,Leukemia ,030104 developmental biology ,Immunology ,medicine ,Cancer research ,KRAS ,Carcinogenesis - Abstract
Despite recent advances in treatment, human precursor B-cell acute lymphoblastic leukemia (B-ALL) remains a challenging clinical entity. Recent genome-wide studies have uncovered frequent genetic alterations involving RAS pathway mutations and loss of the INK4A/ARF locus, suggesting their important role in the pathogenesis, relapse, and chemotherapy resistance of B-ALL. To better understand the oncogenic mechanisms by which these alterations might promote B-ALL and to develop an in vivo preclinical model of relapsed B-ALL, we engineered mouse strains with induced somatic KrasG12D pathway activation and/or loss of Ink4a/Arf during early stages of B-cell development. Although constitutive activation of KrasG12D in B cells induced prominent transcriptional changes that resulted in enhanced proliferation, it was not sufficient by itself to induce development of a high-grade leukemia/lymphoma. Instead, in 40% of mice, these engineered mutations promoted development of a clonal low-grade lymphoproliferative disorder resembling human extranodal marginal-zone lymphoma of mucosa-associated lymphoid tissue or lymphoplasmacytic lymphoma. Interestingly, loss of the Ink4a/Arf locus, apart from reducing the number of apoptotic B cells broadly attenuated KrasG12D-induced transcriptional signatures. However, combined Kras activation and Ink4a/Arf inactivation cooperated functionally to induce a fully penetrant, highly aggressive B-ALL phenotype resembling high-risk subtypes of human B-ALL such as BCR-ABL and CRFL2-rearranged. Ninety percent of examined murine B-ALL tumors showed loss of the wild-type Ink4a/Arf locus without acquisition of highly recurrent cooperating events, underscoring the role of Ink4a/Arf in restraining Kras-driven oncogenesis in the lymphoid compartment. These data highlight the importance of functional cooperation between mutated Kras and Ink4a/Arf loss on B-ALL.
- Published
- 2017
- Full Text
- View/download PDF
47. Strategies to Reduce Injuries and Develop Confidence in Elders (STRIDE): A Cluster-Randomized Pragmatic Trial of a Multifactorial Fall Injury Prevention Strategy: Design and Methods
- Author
-
Lawrence Garber, Peter Peduzzi, Shehzad Basaria, Elena Volpi, Denise Esserman, Catherine Hanson, James Dziura, David A. Ganz, Peter Charpentier, Thomas G. Travison, Martha B. Carnie, Jerry H. Gurwitz, Thomas M. Gill, Patricia C. Dykes, Charles Lu, Haseena Rajeevan, Neil B. Alexander, Fred C. Ko, Richard Eder, Heather G. Allore, Stephen C. Waring, David B. Reuben, Dorothy I. Baker, Shalender Bhasin, Albert W. Wu, Steven B. Clauser, Katy L. B. Araujo, Rosaly Correa-de-Araujo, Jocelyn Wiggins, Albert L. Siu, Carri Casteel, Maureen Fagan, Thomas W. Storer, Eleni A. Skokos, Jeremy N. Rich, Erich J. Greene, Michael E. Miller, Jay Magaziner, Pamela W. Duncan, Scott Margolis, Susan L. Greenspan, Siobhan K McMahon, Joanne M. McGloin, Nancy K. Latham, and Robert B. Wallace
- Subjects
Male ,Gerontology ,and promotion of well-being ,Comparative Effectiveness Research ,Aging ,Nurse falls care managers ,Poison control ,01 natural sciences ,Suicide prevention ,law.invention ,010104 statistics & probability ,0302 clinical medicine ,Randomized controlled trial ,law ,Health care ,80 and over ,Fall prevention ,030212 general & internal medicine ,Injuries and Accidents ,Aged, 80 and over ,Rehabilitation ,Head injury ,Health Services ,The Journal of Gerontology: Medical Sciences ,Female ,Patient Safety ,medicine.symptom ,medicine.medical_specialty ,Clinical Trials and Supportive Activities ,Clinical Sciences ,and over ,Motivational Interviewing ,Risk Assessment ,03 medical and health sciences ,Clinical Research ,Injury prevention ,medicine ,Humans ,Patient and stakeholders in fall injury prevention research ,0101 mathematics ,Aged ,business.industry ,Prevention ,Prevention of disease and conditions ,medicine.disease ,Good Health and Well Being ,Clinical effectiveness ,Falling (accident) ,Injury (total) Accidents/Adverse Effects ,Physical therapy ,3.1 Primary prevention interventions to modify behaviours or promote wellbeing ,Wounds and Injuries ,Accidental Falls ,Geriatrics and Gerontology ,business ,3.1 Primary prevention interventions to modify behaviours or promote well-being - Abstract
Author(s): Bhasin, Shalender; Gill, Thomas M; Reuben, David B; Latham, Nancy K; Gurwitz, Jerry H; Dykes, Patricia; McMahon, Siobhan; Storer, Thomas W; Duncan, Pamela W; Ganz, David A; Basaria, Shehzad; Miller, Michael E; Travison, Thomas G; Greene, Erich J; Dziura, James; Esserman, Denise; Allore, Heather; Carnie, Martha B; Fagan, Maureen; Hanson, Catherine; Baker, Dorothy; Greenspan, Susan L; Alexander, Neil; Ko, Fred; Siu, Albert L; Volpi, Elena; Wu, Albert W; Rich, Jeremy; Waring, Stephen C; Wallace, Robert; Casteel, Carri; Magaziner, Jay; Charpentier, Peter; Lu, Charles; Araujo, Katy; Rajeevan, Haseena; Margolis, Scott; Eder, Richard; McGloin, Joanne M; Skokos, Eleni; Wiggins, Jocelyn; Garber, Lawrence; Clauser, Steven B; Correa-De-Araujo, Rosaly; Peduzzi, Peter | Abstract: Background:Fall injuries are a major cause of morbidity and mortality among older adults. We describe the design of a pragmatic trial to compare the effectiveness of an evidence-based, patient-centered multifactorial fall injury prevention strategy to an enhanced usual care. Methods:Strategies to Reduce Injuries and Develop Confidence in Elders (STRIDE) is a 40-month cluster-randomized, parallel-group, superiority, pragmatic trial being conducted at 86 primary care practices in 10 health care systems across United States. The 86 practices were randomized to intervention or control group using covariate-based constrained randomization, stratified by health care system. Participants are community-living persons, ≥70 years, at increased risk for serious fall injuries. The intervention is a comanagement model in which a nurse Falls Care Manager performs multifactorial risk assessments, develops individualized care plans, which include surveillance, follow-up evaluation, and intervention strategies. Control group receives enhanced usual care, with clinicians and patients receiving evidence-based information on falls prevention. Primary outcome is serious fall injuries, operationalized as those leading to medical attention (nonvertebral fractures, joint dislocation, head injury, lacerations, and other major sequelae). Secondary outcomes include all fall injuries, all falls, and well-being (concern for falling; anxiety and depressive symptoms; physical function and disability). Target sample size was 5,322 participants to provide 90% power to detect 20% reduction in primary outcome rate relative to control. Results:Trial enrolled 5,451 subjects in 20 months. Intervention and follow-up are ongoing. Conclusions:The findings of the STRIDE study will have important clinical and policy implications for the prevention of fall injuries in older adults.
- Published
- 2017
- Full Text
- View/download PDF
48. Targeting glioma stem cells through combined BMI1 and EZH2 inhibition
- Author
-
Ryan C. Gimple, Xiuxing Wang, Leo J.Y. Kim, Xun Jin, Andrew E. Sloan, Shideng Bao, Qiulian Wu, Ping Huang, Jeremy N. Rich, Jill S. Barnholtz-Sloan, Briana C. Prager, Lisa C. Wallace, Claudia L.L. Valentim, Tyler E. Miller, Tanwarat Sanvoranart, Stephen C. Mack, and Qi Gang Zhou
- Subjects
0301 basic medicine ,cancer stem cell ,endocrine system ,glioma stem cell ,Angiogenesis ,macromolecular substances ,Biology ,Article ,General Biochemistry, Genetics and Molecular Biology ,Epigenesis, Genetic ,Mice ,03 medical and health sciences ,Downregulation and upregulation ,Cancer stem cell ,Glioma ,medicine ,Animals ,Humans ,Enhancer of Zeste Homolog 2 Protein ,EZH2 ,Polycomb Repressive Complex 1 ,Brain Neoplasms ,Reverse Transcriptase Polymerase Chain Reaction ,fungi ,Mesenchymal stem cell ,General Medicine ,medicine.disease ,BMI1 ,Cell biology ,030104 developmental biology ,Neoplastic Stem Cells ,Cancer research ,Stem cell ,Glioblastoma - Abstract
Glioblastomas are lethal cancers defined by angiogenesis and pseudopalisading necrosis. Here, we demonstrate that these histological features are associated with distinct transcriptional programs, with vascular regions showing a proneural profile and hypoxic regions a mesenchymal pattern. As these regions harbor glioma stem cells (GSCs), we investigated the epigenetic regulation of these two niches. Proneural, perivascular GSCs activated EZH2, whereas mesenchymal GSCs in hypoxic regions expressed BMI1 protein, which promoted cellular survival under stress, due to downregulation of the E3 ligase, RNF144A. Using both genetic and pharmacologic inhibition, we found that proneural GSCs are preferentially sensitive to EZH2 disruption, whereas mesenchymal GSCs are preferentially sensitive to BMI1 inhibition. Given that glioblastomas contain both proneural and mesenchymal GSCs, combined EZH2 and BMI1 targeting proved more effective than either agent alone both in culture and in vivo, suggesting that strategies that simultaneously target multiple epigenetic regulators within glioblastomas may be necessary to overcome resistance to therapies caused by intratumoral heterogeneity.
- Published
- 2017
- Full Text
- View/download PDF
49. A Multilevel Analysis of Patient Engagement and Patient-Reported Outcomes in Primary Care Practices of Accountable Care Organizations
- Author
-
Stephen M. Shortell, Thomas P. Huber, Jeremy N. Rich, Tom Summerfelt, Bing Ying Poon, Hector P. Rodriguez, Patricia P. Ramsay, and Susan L. Ivey
- Subjects
Male ,Outcome Assessment ,Disease ,Cardiovascular ,0302 clinical medicine ,Patient-Centered Care ,Surveys and Questionnaires ,Outcome Assessment, Health Care ,80 and over ,030212 general & internal medicine ,Depression (differential diagnoses) ,Aged, 80 and over ,Response rate (survey) ,patient engagement ,Depression ,030503 health policy & services ,Diabetes ,Middle Aged ,Heart Disease ,Mental Health ,patient-reported outcomes ,Cardiovascular Diseases ,Multilevel Analysis ,Female ,0305 other medical science ,Adult ,medicine.medical_specialty ,Adolescent ,Clinical Sciences ,accountable care organizations ,7.3 Management and decision making ,Young Adult ,03 medical and health sciences ,Clinical Research ,General & Internal Medicine ,Behavioral and Social Science ,Diabetes Mellitus ,Internal Medicine ,medicine ,Humans ,Patient Reported Outcome Measures ,Social determinants of health ,Aged ,Patient Activation Measure ,Accountable Care Organizations ,business.industry ,Prevention ,Capsule Commentary ,Odds ratio ,medicine.disease ,Comorbidity ,Health Care ,Good Health and Well Being ,Cross-Sectional Studies ,Logistic Models ,Family medicine ,Observational study ,Management of diseases and conditions ,Patient Participation ,business - Abstract
BackgroundThe growing movement toward more accountable care delivery and the increasing number of people with chronic illnesses underscores the need for primary care practices to engage patients in their own care.ObjectiveFor adult primary care practices seeing patients with diabetes and/or cardiovascular disease, we examined the relationship between selected practice characteristics, patient engagement, and patient-reported outcomes of care.DesignCross-sectional multilevel observational study of 16 randomly selected practices in two large accountable care organizations (ACOs).ParticipantsPatients with diabetes and/or cardiovascular disease (CVD) who met study eligibility criteria (n = 4368) and received care in 2014 were randomly selected to complete a patient activation and PRO survey (51% response rate; n = 2176). Primary care team members of the 16 practices completed surveys that assessed practice culture, relational coordination, and teamwork (86% response rate; n = 411).Main measuresPatient-reported outcomes included depression (PHQ-4), physical functioning (PROMIS SF12a), and social functioning (PROMIS SF8a), the Patient Assessment of Chronic Illness Care instrument (PACIC-11), and the Patient Activation Measure instrument (PAM-13). Patient-level covariates included patient age, gender, education, insurance coverage, limited English language proficiency, blood pressure, HbA1c, LDL-cholesterol, and disease comorbidity burden. For each of the 16 practices, patient-centered culture and the degree of relational coordination among team members were measured using a clinician and staff survey. The implementation of shared decision-making activities in each practice was assessed using an operational leader survey.Key resultsHaving a patient-centered culture was positively associated with fewer depression symptoms (odds ratio [OR] = 1.51; confidence interval [CI] 1.04, 2.19) and better physical function scores (OR = 1.85; CI 1.25, 2.73). Patient activation was positively associated with fewer depression symptoms (OR = 2.26; CI 1.79, 2.86), better physical health (OR = 2.56; CI 2.00, 3.27), and better social health functioning (OR = 4.12; CI 3.21, 5.29). Patient activation (PAM-13) mediated the positive association between patients' experience of chronic illness care and each of the three patient-reported outcome measures-fewer depression symptoms, better physical health, and better social health. Relational coordination and shared decision-making activities reported by practices were not significantly associated with higher patient-reported outcome scores.ConclusionsDiabetic and CVD patients who received care from ACO-affiliated practices with more developed patient-centered cultures reported lower PHQ-4 depression symptom scores and better physical functioning. Diabetic and CVD patients who were more highly activated to participate in their care reported lower PHQ-4 scores and better physical and social outcomes of care.
- Published
- 2017
- Full Text
- View/download PDF
50. TMIC-01. THE DYSTROGLYCAN RECEPTOR MAINTAINS GLIOMA STEM CELLS IN THE VASCULAR NICHE
- Author
-
Terrance Grant Johns, Zara C. Bruce, Jeremy N. Rich, Krishna Bhat, Kevin P. Campbell, Justin D. Lathia, Yi Chieh Lim, Ulrich Baumgartner, Kathleen S. Ensbey, Fiona M. Smith, Brett W. Stringer, Paul R. Jamieson, Carolin Offenhäuser, Courtney L.R. Jurd, Thomas Robertson, Bryan W. Day, Rochelle C.J. D’Souza, Andrew W. Boyd, Rosalind L. Jeffree, Seckin Akgul, and Yuchen Li
- Subjects
Cancer Research ,biology ,Central nervous system ,Tumor initiation ,medicine.disease ,Phenotype ,Extracellular matrix ,medicine.anatomical_structure ,Oncology ,Glioma ,Dystroglycan ,biology.protein ,medicine ,Cancer research ,Tumor Microenvironment ,Neurology (clinical) ,Stem cell ,Receptor - Abstract
Glioblastomas (GBMs) are malignant central nervous system (CNS) neoplasms with a very poor prognosis. They display cellular hierarchies containing self-renewing tumourigenic glioma stem cells (GSCs) in a complex heterogeneous microenvironment. One proposed GSC niche is the extracellular matrix (ECM)-rich perivascular bed of the tumour. Here, we report that the ECM binding alpha (α) subunit of the dystroglycan (DG) receptor is expressed and functionally glycosylated on GSCs residing in the vascular niche. Glycosylated αDG is also expressed highly on the most aggressive mesenchymal-like GBM tumour tissue. Furthermore, we found that DG acts to maintain a de-differentiated stem cell-like phenotype via tight control of MAPK activation. Antibody-based blockade of αDG induces robust ERK-mediated differentiation leading to reduced GSC potential. DG was shown to be required for tumour initiation, with constitutive loss significantly delaying or preventing tumourigenic potential in-vivo. These findings reveal a central role of the DG receptor not only as a structural element but also as a critical factor in the maintenance of GSCs in the GBM vascular niche.
- Published
- 2019
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.