424 results on '"Eberhart CG"'
Search Results
2. ACVR1C/SMAD2 signaling promotes invasion and growth in retinoblastoma
- Author
-
Asnaghi L, White DT, Key N, Choi J, Mahale A, Alkatan H, Edward DP, Elkhamary SM, Al-Mesfer S, Maktabi A, Hurtado CG, Lee GY, Carcaboso AM, Mumm JS, Safieh LA, and Eberhart CG
- Subjects
eye diseases - Abstract
Retinoblastoma is the most common intraocular cancer in children. While the primary tumor can often be treated by local or systemic chemotherapy, metastatic dissemination is generally resistant to therapy and remains a leading cause of pediatric cancer death in much of the world. In order to identify new therapeutic targets in aggressive tumors, we sequenced RNA transcripts in five snap frozen retinoblastomas which invaded the optic nerve and five which did not. A three-fold increase was noted in mRNA levels of ACVR1C/ALK7, a type I receptor of the TGF-ß family, in invasive retinoblastomas, while downregulation of DACT2 and LEFTY2, negative modulators of the ACVR1C signaling, was observed in most invasive tumors. A two- to three-fold increase in ACVR1C mRNA was also found in invasive WERI Rb1 and Y79 cells as compared to non-invasive cells in vitro. Transcripts of ACVR1C receptor and its ligands (Nodal, Activin A/B, and GDF3) were expressed in six retinoblastoma lines, and evidence of downstream SMAD2 signaling was present in all these lines. Pharmacological inhibition of ACVR1C signaling using SB505124, or genetic downregulation of the receptor using shRNA potently suppressed invasion, growth, survival, and reduced the protein levels of the mesenchymal markers ZEB1 and Snail. The inhibitory effects on invasion, growth, and proliferation were recapitulated by knocking down SMAD2, but not SMAD3. Finally, in an orthotopic zebrafish model of retinoblastoma, a 55% decrease in tumor spread was noted (p = 0.0026) when larvae were treated with 3 µM of SB505124, as compared to DMSO. Similarly, knockdown of ACVR1C in injected tumor cells using shRNA also resulted in a 54% reduction in tumor dissemination in the zebrafish eye as compared to scrambled shRNA control (p = 0.0005). Our data support a role for the ACVR1C/SMAD2 pathway in promoting invasion and growth of retinoblastoma.
- Published
- 2019
3. Demoplastic/nodular medulloblastoma
- Author
-
Pietsch, T, Ellison, Dw, Haapasalo, H, Giangaspero, Felice, Wiestler, Od, Pfister, S, and Eberhart, Cg
- Published
- 2016
4. Medulloblastoma, classic
- Author
-
Ellison, Dw, Eberhart, Cg, Giangaspero, Felice, Haapasalo, H, Pietsch, T, Wiestler, Od, and Pfister, S.
- Published
- 2016
5. Novel inhibitor of the canonical WNT-pathway LGK974 impedes the in vitro growth of glioblastoma cell lines decreasing the tumor stem cell compartment
- Author
-
Suwala, A, Kahlert, U, Eberhart, CG, and Maciaczyk, J
- Subjects
ddc: 610 ,WNT pathway ,porcupine inhibitor ,glioblastoma ,610 Medical sciences ,Medicine - Abstract
Objective: Developmentally conserved WNT-signaling pathway plays a crucial role in a physiological process of cell proliferation and migration. Moreover, it is associated with carcinogenesis and epithelial-to-mesenchymal-transition (EMT) in various tumors, including malignant gliomas. High WNT-activation[for full text, please go to the a.m. URL], 66. Jahrestagung der Deutschen Gesellschaft für Neurochirurgie (DGNC)
- Published
- 2015
- Full Text
- View/download PDF
6. Cytogenetic prognostication within medulloblastoma subgroups
- Author
-
Shih, DJH, Northcott, PA, Remke, M, Korshunov, A, Ramaswamy, V, Kool, M, Luu, B, Yao, Y, Wang, X, Dubuc, AM, Garzia, L, Peacock, J, Mack, SC, Wu, X, Rolider, A, Morrissy, AS, Cavalli, FMG, Jones, DTW, Zitterbart, K, Faria, CC, Schüller, U, Kren, L, Kumabe, T, Tominaga, T, Ra, YS, Garami, M, Hauser, P, Chan, JA, Robinson, S, Bognár, L, Klekner, A, Saad, AG, Liau, LM, Albrecht, S, Fontebasso, A, Cinalli, G, De Antonellis, P, Zollo, M, Cooper, MK, Thompson, RC, Bailey, S, Lindsey, JC, Di Rocco, C, Massimi, L, Michiels, EMC, Scherer, SW, Phillips, JJ, Gupta, N, Fan, X, Muraszko, KM, Vibhakar, R, Eberhart, CG, Fouladi, M, Lach, B, Jung, S, Wechsler-Reya, RJ, Fèvre-Montange, M, Jouvet, A, Jabado, N, Pollack, IF, Weiss, WA, Lee, JY, Cho, BK, Kim, SK, Wang, KC, Leonard, JR, Rubin, JB, De Torres, C, Lavarino, C, Mora, J, Cho, YJ, Tabori, U, Olson, JM, Gajjar, A, Packer, RJ, Rutkowski, S, Pomeroy, SL, French, PJ, Kloosterhof, NK, Kros, JM, Van Meir, EG, Clifford, SC, Bourdeaut, F, Delattre, O, Doz, FF, Hawkins, CE, Malkin, D, and Grajkowska, WA
- Subjects
Male ,Adolescent ,Kruppel-Like Transcription Factors ,Zinc Finger Protein Gli2 ,Risk Assessment ,Proto-Oncogene Proteins c-myc ,Cytogenetics ,Young Adult ,Risk Factors ,Predictive Value of Tests ,Biomarkers, Tumor ,Humans ,Hedgehog Proteins ,Oncology & Carcinogenesis ,Child ,In Situ Hybridization, Fluorescence ,Proportional Hazards Models ,Chromosomes, Human, Pair 14 ,Oncology And Carcinogenesis ,Chromosomes, Human, Pair 11 ,Gene Expression Profiling ,Nuclear Proteins ,Reproducibility of Results ,Infant ,Prognosis ,Gene Expression Regulation, Neoplastic ,Wnt Proteins ,Tissue Array Analysis ,Child, Preschool ,Female ,Medulloblastoma - Abstract
Purpose: Medulloblastoma comprises four distinct molecular subgroups: WNT, SHH, Group 3, and Group 4. Current medulloblastoma protocols stratify patients based on clinical features: patient age, metastatic stage, extent of resection, and histologic variant. Stark prognostic and genetic differences among the four subgroups suggest that subgroup-specific molecular biomarkers could improve patient prognostication. Patients and Methods: Molecular biomarkers were identified from a discovery set of 673 medulloblastomas from 43 cities around the world. Combined risk stratification models were designed based on clinical and cytogenetic biomarkers identified by multivariable Cox proportional hazards analyses. Identified biomarkers were tested using fluorescent in situ hybridization (FISH) on a nonoverlapping medulloblastoma tissue microarray (n = 453), with subsequent validation of the risk stratification models. Results: Subgroup information improves the predictive accuracy of a multivariable survival model compared with clinical biomarkers alone. Most previously published cytogenetic biomarkers are only prognostic within a single medulloblastoma subgroup. Profiling six FISH biomarkers (GLI2, MYC, chromosome 11 [chr11], chr14, 17p, and 17q) on formalin-fixed paraffin-embedded tissues, we can reliably and reproducibly identify very low-risk and very high-risk patients within SHH, Group 3, and Group 4 medulloblastomas. Conclusion: Combining subgroup and cytogenetic biomarkers with established clinical biomarkers substantially improves patient prognostication, even in the context of heterogeneous clinical therapies. The prognostic significance of most molecular biomarkers is restricted to a specific subgroup. We have identified a small panel of cytogenetic biomarkers that reliably identifies very high-risk and very low-risk groups of patients, making it an excellent tool for selecting patients for therapy intensification and therapy de-escalation in future clinical trials. © 2014 by American Society of Clinical Oncology.
- Published
- 2014
7. Subgroup-specific structural variation across 1,000 medulloblastoma genomes
- Author
-
Northcott, PA, Shih, DJH, Peacock, J, Garzia, L, Sorana Morrissy, A, Zichner, T, Stútz, AM, Korshunov, A, Reimand, J, Schumacher, SE, Beroukhim, R, Ellison, DW, Marshall, CR, Lionel, AC, MacK, S, Dubuc, A, Yao, Y, Ramaswamy, V, Luu, B, Rolider, A, Cavalli, FMG, Wang, X, Remke, M, Wu, X, Chiu, RYB, Chu, A, Chuah, E, Corbett, RD, Hoad, GR, Jackman, SD, Li, Y, Lo, A, Mungall, KL, Ming Nip, K, Qian, JQ, Raymond, AGJ, Thiessen, N, Varhol, RJ, Birol, I, Moore, RA, Mungall, AJ, Holt, R, Kawauchi, D, Roussel, MF, Kool, M, Jones, DTW, Witt, H, Fernandez-L, A, Kenney, AM, Wechsler-Reya, RJ, Dirks, P, Aviv, T, Grajkowska, WA, Perek-Polnik, M, Haberler, CC, Delattre, O, Reynaud, SS, Doz, FF, Pernet-Fattet, SS, Cho, BK, Kim, SK, Wang, KC, Scheurlen, W, Eberhart, CG, Fèvre-Montange, M, Jouvet, A, Pollack, IF, Fan, X, Muraszko, KM, Yancey Gillespie, G, Di Rocco, C, Massimi, L, Michiels, EMC, Kloosterhof, NK, French, PJ, Kros, JM, Olson, JM, Ellenbogen, RG, Zitterbart, K, Kren, L, Thompson, RC, and Cooper, MK
- Subjects
Oncogene Proteins, Fusion ,DNA Copy Number Variations ,Genome, Human ,General Science & Technology ,NF-kappa B ,Genes, myc ,Proteins ,Nerve Tissue Proteins ,Genomics ,Translocation, Genetic ,Transforming Growth Factor beta ,Gene Duplication ,Genomic Structural Variation ,MD Multidisciplinary ,Humans ,Hedgehog Proteins ,RNA, Long Noncoding ,Cerebellar Neoplasms ,Carrier Proteins ,Child ,Medulloblastoma ,Signal Transduction - Abstract
Medulloblastoma, the most common malignant paediatric brain tumour, is currently treated with nonspecific cytotoxic therapies including surgery, whole-brain radiation, and aggressive chemotherapy. As medulloblastoma exhibits marked intertumoural heterogeneity, with at least four distinct molecular variants, previous attempts to identify targets for therapy have been underpowered because of small samples sizes. Here we report somatic copy number aberrations (SCNAs) in 1,087 unique medulloblastomas. SCNAs are common in medulloblastoma, and are predominantly subgroup-enriched. The most common region of focal copy number gain is a tandem duplication of SNCAIP, a gene associated with Parkinson's disease, which is exquisitely restricted to Group 4α. Recurrent translocations of PVT1, including PVT1-MYC and PVT1-NDRG1, that arise through chromothripsis are restricted to Group 3. Numerous targetable SCNAs, including recurrent events targeting TGF-β signalling in Group 3, and NF-κB signalling in Group 4, suggest future avenues for rational, targeted therapy. © 2012 Macmillan Publishers Limited. All rights reserved.
- Published
- 2012
8. WNT activation by lithium abrogates TP53 mutation associated radiation resistance in medulloblastoma
- Author
-
Zhukova, N, Ramaswamy, V, Remke, M, Martin, DC, Castelo-Branco, P, Zhang, CH, Fraser, M, Tse, K, Poon, R, Shih, DJH, Baskin, B, Ray, PN, Bouffet, E, Dirks, P, von Bueren, AO, Pfaff, E, Korshunov, A, Jones, DTW, Northcott, PA, Kool, M, Pugh, TJ, Pomeroy, SL, Cho, YJ, Pietsch, T, Gessi, M, Rutkowski, S, Bognár, L, Cho, BK, Eberhart, CG, Conter, CF, Fouladi, M, French, PJ, Grajkowska, WA, Gupta, N, Hauser, P, Jabado, N, Vasiljevic, A, Jung, S, Kim, SK, Klekner, A, Kumabe, T, Lach, B, Leonard, JR, Liau, LM, Massimi, L, Pollack, IF, Ra, YS, Rubin, JB, Van Meir, EG, Wang, KC, Weiss, WA, Zitterbart, K, Bristow, RG, Alman, B, Hawkins, CE, Malkin, D, Clifford, SC, Pfister, SM, Taylor, MD, Tabori, U, Zhukova, N, Ramaswamy, V, Remke, M, Martin, DC, Castelo-Branco, P, Zhang, CH, Fraser, M, Tse, K, Poon, R, Shih, DJH, Baskin, B, Ray, PN, Bouffet, E, Dirks, P, von Bueren, AO, Pfaff, E, Korshunov, A, Jones, DTW, Northcott, PA, Kool, M, Pugh, TJ, Pomeroy, SL, Cho, YJ, Pietsch, T, Gessi, M, Rutkowski, S, Bognár, L, Cho, BK, Eberhart, CG, Conter, CF, Fouladi, M, French, PJ, Grajkowska, WA, Gupta, N, Hauser, P, Jabado, N, Vasiljevic, A, Jung, S, Kim, SK, Klekner, A, Kumabe, T, Lach, B, Leonard, JR, Liau, LM, Massimi, L, Pollack, IF, Ra, YS, Rubin, JB, Van Meir, EG, Wang, KC, Weiss, WA, Zitterbart, K, Bristow, RG, Alman, B, Hawkins, CE, Malkin, D, Clifford, SC, Pfister, SM, Taylor, MD, and Tabori, U
- Abstract
TP53 mutations confer subgroup specific poor survival for children with medulloblastoma. We hypothesized that WNT activation which is associated with improved survival for such children abrogates TP53 related radioresistance and can be used to sensitize TP53 mutant tumors for radiation. We examined the subgroup-specific role of TP53 mutations in a cohort of 314 patients treated with radiation. TP53 wild-type or mutant human medulloblastoma cell-lines and normal neural stem cells were used to test radioresistance of TP53 mutations and the radiosensitizing effect of WNT activation on tumors and the developing brain. Children with WNT/TP53 mutant medulloblastoma had higher 5-year survival than those with SHH/TP53 mutant tumours (100% and 36.6% ± 8.7%, respectively (p < 0.001)). Introduction of TP53 mutation into medulloblastoma cells induced radioresistance (survival fractions at 2Gy (SF2) of 89% ± 2% vs. 57.4% ± 1.8% (p < 0.01)). In contrast, β-catenin mutation sensitized TP53 mutant cells to radiation (p < 0.05). Lithium, an activator of the WNT pathway, sensitized TP53 mutant medulloblastoma to radiation (SF2 of 43.5% ± 1.5% in lithium treated cells vs. 56.6 ± 3% (p < 0.01)) accompanied by increased number of γH2AX foci. Normal neural stem cells were protected from lithium induced radiation damage (SF2 of 33% ± 8% for lithium treated cells vs. 27% ± 3% for untreated controls (p = 0.05). Poor survival of patients with TP53 mutant medulloblastoma may be related to radiation resistance. Since constitutive activation of the WNT pathway by lithium sensitizes TP53 mutant medulloblastoma cells and protect normal neural stem cells from radiation, this oral drug may represent an attractive novel therapy for high-risk medulloblastomas.
- Published
- 2014
9. Hypoxia-mediated induction of EMT activators increases in vitro invasion of glioblastoma-derived cell cultures
- Author
-
Kahlert, UD, Siebzehnrubl, FA, Bar, EE, Eberhart, CG, Maciaczyk, J, Kahlert, UD, Siebzehnrubl, FA, Bar, EE, Eberhart, CG, and Maciaczyk, J
- Published
- 2014
10. Medulloblastoma
- Author
-
Giangaspero, Felice, Eberhart, Cg, Haapasalo, H, Pietsch, T, Wiestler, Od, and Ellison, Dws
- Published
- 2007
11. TERT promoter mutations are highly recurrent in SHH subgroup medulloblastoma
- Author
-
Remke, M, Ramaswamy, V, Peacock, J, Shih, DJH, Koelsche, C, Northcott, PA, Hill, N, Cavalli, FMG, Kool, Mirjam, Wang, X, Mack, SC, Barszczyk, M, Morrissy, AS, Wu, XC, Agnihotri, S, Luu, B, Jones, DTW, Garzia, L, Dubuc, AM, Zhukova, N, Vanner, R, Kros, J.M., French, Pim, van Meir, EG, Vibhakar, R, Zitterbart, K, Chan, JA, Bognar, L, Klekner, A, Lach, B, Jung, S, Saad, AG, Albrecht, S, Liau, LM, Zollo, M, Cooper, MK, Thompson, RC, Delattre, OO, Bourdeaut, F, Doz, FF, Garami, M, Hauser, P, Carlotti, CG, Van Meter, TE, Massimi, L, Fults, D, Pomeroy, SL, Kumabe, T, Ra, YS, Leonard, JR, Elbabaa, SK, Mora, J, Rubin, JB, Cho, YJ, McLendon, RE, Bigner, DD, Eberhart, CG, Fouladi, M, Wechsler-Reya, RJ, Faria, CC, Croul, SE, Huang, A, Bouffet, E, Hawkins, CE, Dirks, PB, Weiss, WA, Schuller, U, Pollack, IF, Rutkowski, S, Meyronet, D, Jouvet, A, Fevre-Montange, M, Jabado, N, Perek-Polnik, M, Grajkowska, WA, Kim, SK, Rutka, JT, Malkin, D, Tabori, U, Pfister, SM, Korshunov, A, von Deimling, A, Taylor, MD, Remke, M, Ramaswamy, V, Peacock, J, Shih, DJH, Koelsche, C, Northcott, PA, Hill, N, Cavalli, FMG, Kool, Mirjam, Wang, X, Mack, SC, Barszczyk, M, Morrissy, AS, Wu, XC, Agnihotri, S, Luu, B, Jones, DTW, Garzia, L, Dubuc, AM, Zhukova, N, Vanner, R, Kros, J.M., French, Pim, van Meir, EG, Vibhakar, R, Zitterbart, K, Chan, JA, Bognar, L, Klekner, A, Lach, B, Jung, S, Saad, AG, Albrecht, S, Liau, LM, Zollo, M, Cooper, MK, Thompson, RC, Delattre, OO, Bourdeaut, F, Doz, FF, Garami, M, Hauser, P, Carlotti, CG, Van Meter, TE, Massimi, L, Fults, D, Pomeroy, SL, Kumabe, T, Ra, YS, Leonard, JR, Elbabaa, SK, Mora, J, Rubin, JB, Cho, YJ, McLendon, RE, Bigner, DD, Eberhart, CG, Fouladi, M, Wechsler-Reya, RJ, Faria, CC, Croul, SE, Huang, A, Bouffet, E, Hawkins, CE, Dirks, PB, Weiss, WA, Schuller, U, Pollack, IF, Rutkowski, S, Meyronet, D, Jouvet, A, Fevre-Montange, M, Jabado, N, Perek-Polnik, M, Grajkowska, WA, Kim, SK, Rutka, JT, Malkin, D, Tabori, U, Pfister, SM, Korshunov, A, von Deimling, A, and Taylor, MD
- Abstract
Telomerase reverse transcriptase (TERT) promoter mutations were recently shown to drive telomerase activity in various cancer types, including medulloblastoma. However, the clinical and biological implications of TERT mutations in medulloblastoma have not been described. Hence, we sought to describe these mutations and their impact in a subgroup-specific manner. We analyzed the TERT promoter by direct sequencing and genotyping in 466 medulloblastomas. The mutational distributions were determined according to subgroup affiliation, demographics, and clinical, prognostic, and molecular features. Integrated genomics approaches were used to identify specific somatic copy number alterations in TERT promoter-mutated and wild-type tumors. Overall, TERT promoter mutations were identified in 21 % of medulloblastomas. Strikingly, the highest frequencies of TERT mutations were observed in SHH (83 %; 55/66) and WNT (31 %; 4/13) medulloblastomas derived from adult patients. Group 3 and Group 4 harbored this alteration in < 5 % of cases and showed no association with increased patient age. The prognostic implications of these mutations were highly subgroup-specific. TERT mutations identified a subset with good and poor prognosis in SHH and Group 4 tumors, respectively. Monosomy 6 was mostly restricted to WNT tumors without TERT mutations. Hallmark SHH focal copy number aberrations and chromosome 10q deletion were mutually exclusive with TERT mutations within SHH tumors. TERT promoter mutations are the most common recurrent somatic point mutation in medulloblastoma, and are very highly enriched in adult SHH and WNT tumors. TERT mutations define a subset of SHH medulloblastoma with distinct demographics, cytogenetics, and outcomes.
- Published
- 2013
12. Podocalyxin-like protein is expressed in glioblastoma multiforme stem-like cells and is associated with poor outcome
- Author
-
Binder, Z, Siu, I, Eberhart, C, Ap Rhys, C, Bai, R, Staedtke, V, Zhang, H, Smoll, N, Piantadosi, S, Piccirillo, S, Dimeco, F, Weingart, J, Vescovi, A, Olivi, A, Riggins, G, Gallia, G, Binder, ZA, Siu, IM, Eberhart, CG, Bai, RY, Smoll, NR, Piccirillo, SG, Weingart, JD, Riggins, GJ, Gallia, GL, VESCOVI, ANGELO LUIGI, Binder, Z, Siu, I, Eberhart, C, Ap Rhys, C, Bai, R, Staedtke, V, Zhang, H, Smoll, N, Piantadosi, S, Piccirillo, S, Dimeco, F, Weingart, J, Vescovi, A, Olivi, A, Riggins, G, Gallia, G, Binder, ZA, Siu, IM, Eberhart, CG, Bai, RY, Smoll, NR, Piccirillo, SG, Weingart, JD, Riggins, GJ, Gallia, GL, and VESCOVI, ANGELO LUIGI
- Abstract
Glioblastoma multiforme (GBM) is the most common primary malignant adult brain tumor and is associated with poor survival. Recently, stem-like cell populations have been identified in numerous malignancies including GBM. To identify genes whose expression is changed with differentiation, we compared transcript profiles from a GBM oncosphere line before and after differentiation. Bioinformatic analysis of the gene expression profiles identified podocalyxin-like protein (PODXL), a protein highly expressed in human embryonic stem cells, as a potential marker of undifferentiated GBM stem-like cells. The loss of PODXL expression upon differentiation of GBM stem-like cell lines was confirmed by quantitative real-time PCR and flow cytometry. Analytical flow cytometry of numerous GBM oncosphere lines demonstrated PODXL expression in all lines examined. Knockdown studies and flow cytometric cell sorting experiments demonstrated that PODXL is involved in GBM stem-like cell proliferation and oncosphere formation. Compared to PODXL-negative cells, PODXL-positive cells had increased expression of the progenitor/stem cell markers Musashi1, SOX2, and BMI1. Finally, PODXL expression directly correlated with increasing glioma grade and was a marker for poor outcome in patients with GBM. In summary, we have demonstrated that PODXL is expressed in GBM stem-like cells and is involved in cell proliferation and oncosphere formation. Moreover, high PODXL expression correlates with increasing glioma grade and decreased overall survival in patients with GBM. © 2013 Binder et al.
- Published
- 2013
13. Krüppel-like family of transcription factor 9, a Differentiation-Associated Transcription Factor, Suppresses Notch1 Signaling and Inhibits Glioblastoma-Initiating Stem Cells
- Author
-
Ying, M, Sang, Y, Li, Y, Guerrero Cazares, H, Quinones Hinojosa, A, Vescovi, A, Eberhart, C, Xia, S, Laterra, J, Eberhart, CG, Laterra, J., VESCOVI, ANGELO LUIGI, Ying, M, Sang, Y, Li, Y, Guerrero Cazares, H, Quinones Hinojosa, A, Vescovi, A, Eberhart, C, Xia, S, Laterra, J, Eberhart, CG, Laterra, J., and VESCOVI, ANGELO LUIGI
- Abstract
Tumor-initiating stem cells (alternatively called cancer stem cells, CSCs) are a subpopulation of tumor cells that plays unique roles in tumor propagation, therapeutic resistance, and tumor recurrence. It is becoming increasingly important to understand the molecular signaling that regulates the self-renewal and differentiation of CSCs. Transcription factors are critical for the regulation of normal and neopolastic stem cells. Here, we examined the expression and function of the Krüppel-like family of transcription factors (KLFs) in human glioblastoma (GBM)-derived neurosphere lines and low-passage primary GBM-derived neurospheres that are enriched for tumor-initiating stem cells. We identify KLF9 as a relatively unique differentiation-induced transcription factor in GBM-derived neurospheres. KLF9 is shown to induce neurosphere cell differentiation, inhibit neurosphere formation, and inhibit neurosphere-derived xenograft growth in vivo. We also show that KLF9 regulates GBM neurosphere cells by binding to the Notch1 promoter and suppressing Notch1 expression and downstream signaling. Our results show for the first time that KLF9 has differentiating and tumor-suppressing functions in tumor-initiating stem cells. © AlphaMed Press.
- Published
- 2011
14. DNER, an epigenetically modulated gene, regulates glioblastoma-derived neurosphere cell differentiation and tumor propagation
- Author
-
Sun, P, Xia, S, Lal, B, Eberhart, C, Quinones Hinojosa, A, Maciaczyk, J, Matsui, W, Dimeco, F, Piccirillo, S, Vescovi, A, Laterra, J, Eberhart, CG, Piccirillo, SM, Laterra, J., VESCOVI, ANGELO LUIGI, Sun, P, Xia, S, Lal, B, Eberhart, C, Quinones Hinojosa, A, Maciaczyk, J, Matsui, W, Dimeco, F, Piccirillo, S, Vescovi, A, Laterra, J, Eberhart, CG, Piccirillo, SM, Laterra, J., and VESCOVI, ANGELO LUIGI
- Abstract
Neurospheres derived from glioblastoma (GBM) and other solid malignancies contain neoplastic stem-like cells that efficiently propagate tumor growth and resist cytotoxic therapeutics. The primary objective of this study was to use histone-modifying agents to elucidate mechanisms by which the phenotype and tumor-promoting capacity of GBM-derived neoplastic stem-like cells are regulated. Using established GBM-derived neurosphere lines and low passage primary GBM-derived neurospheres, we show that histone deacetylase (HDAC) inhibitors inhibit growth, induce differentiation, and induce apoptosis of neoplastic neurosphere cells. A specific gene product induced by HDAC inhibition, Delta/Notch-like epidermal growth factor-related receptor (DNER), inhibited the growth of GBM-derived neurospheres, induced their differentiation in vivo and in vitro, and inhibited their engraftment and growth as tumor xenografts. The differentiating and tumor suppressive effects of DNER, a noncanonical Notch ligand, contrast with the previously established tumor-promoting effects of canonical Notch signaling in brain cancer stem-like cells. Our findings are the first to implicate noncanonical Notch signaling in the regulation of neoplastic stem-like cells and suggest novel neoplastic stem cell targeting treatment strategies for GBM and potentially other solid malignancies.
- Published
- 2009
15. Abnormal DNA methylation of CD133 in colorectal and glioblastoma tumors
- Author
-
Yi, J, Tsai, H, Glöckner, S, Lin, S, Ohm, J, Easwaran, H, James, C, Costello, J, Riggins, G, Eberhart, C, Laterra, J, Vescovi, A, Ahuja, N, Herman, J, Schuebel, K, Baylin, S, Yi, JM, Tsai, HC, Glöckner, SC, Ohm, JE, James, CD, Costello, JF, Eberhart, CG, Herman, JG, Schuebel, KE, Baylin, SB, VESCOVI, ANGELO LUIGI, Yi, J, Tsai, H, Glöckner, S, Lin, S, Ohm, J, Easwaran, H, James, C, Costello, J, Riggins, G, Eberhart, C, Laterra, J, Vescovi, A, Ahuja, N, Herman, J, Schuebel, K, Baylin, S, Yi, JM, Tsai, HC, Glöckner, SC, Ohm, JE, James, CD, Costello, JF, Eberhart, CG, Herman, JG, Schuebel, KE, Baylin, SB, and VESCOVI, ANGELO LUIGI
- Abstract
Much recent effort has focused on identifying and characterizing cellular markers that distinguish tumor propagating cells (TPC) from more differentiated progeny. We report here an unusual promoter DNA methylation pattern for one such marker, the cell surface antigen CD133 (Prominin 1). This protein has been extensively used to enrich putative cancer propagating stem-like cell populations in epithelial tumors and, especially, glioblastomas. We find that, within individual cell lines of cultured colon cancers and glioblastomas, the promoter CpG island of CD133 is DNA methylate d, primarily, in cells with absent or low expression of the marker protein, whereas lack of such methylation is evident in purely CD133+ cells. Differential histone modification marks of active versus repressed genes accompany these DNA methylation changes. This heterogeneous CpG island DNA methylation status in the tumors is unusual in that other DNA hypermethylate d genes tested in such cultures preserve their methylation patterns between separated CD133+ and CD133- cell populations. Furthermore, the CD133 DNA methylation seems to constitute an abnormal promoter signature because it is not found in normal brain and colon but only in cultured and primary tumors. Thus, the DNA methyl ation is imposed on the transition between the active versus repressed transcription state for CD133 only in tumors. Our findings provide additional insight for the dynamics of aberrant DNA methylation associated with aberrant gene silencing in human tumors. ©2008 American Association for Cancer Research.
- Published
- 2008
16. Targeting the hypoxia pathway in uveal melanoma cells
- Author
-
ASNAGHI, L, primary, LIN, MH, additional, LIM, KS, additional, LIM, KJ, additional, BAR, EE, additional, and EBERHART, CG, additional
- Published
- 2013
- Full Text
- View/download PDF
17. Medulloblastoma stem cells.
- Author
-
Fan X, Eberhart CG, Fan, Xing, and Eberhart, Charles G
- Published
- 2008
- Full Text
- View/download PDF
18. RNA interference-mediated c-MYC inhibition prevents cell growth and decreases sensitivity to radio- and chemotherapy in childhood medulloblastoma cells.
- Author
-
von Bueren AO, Shalaby T, Oehler-Jänne C, Arnold L, Stearns D, Eberhart CG, Arcaro A, Pruschy M, Grotzer MA, von Bueren, André O, Shalaby, Tarek, Oehler-Jänne, Christoph, Arnold, Lucia, Stearns, Duncan, Eberhart, Charles G, Arcaro, Alexandre, Pruschy, Martin, and Grotzer, Michael A
- Abstract
Background: With current treatment strategies, nearly half of all medulloblastoma (MB) patients die from progressive tumors. Accordingly, the identification of novel therapeutic strategies remains a major goal. Deregulation of c-MYC is evident in numerous human cancers. In MB, over-expression of c-MYC has been shown to cause anaplasia and correlate with unfavorable prognosis.Methods: To study the role of c-MYC in MB biology, we down-regulated c-MYC expression by using small interfering RNA (siRNA) and investigated changes in cellular proliferation, cell cycle analysis, apoptosis, telomere maintenance, and response to ionizing radiation (IR) and chemotherapeutics in a representative panel of human MB cell lines expressing different levels of c-MYC (DAOY wild-type, DAOY transfected with the empty vector, DAOY transfected with c-MYC, D341, and D425).Results: siRNA-mediated c-MYC down-regulation resulted in an inhibition of cellular proliferation and clonogenic growth, inhibition of G1-S phase cell cycle progression, and a decrease in human telomerase reverse transcriptase (hTERT) expression and telomerase activity. On the other hand, down-regulation of c-MYC reduced apoptosis and decreased the sensitivity of human MB cells to IR, cisplatin, and etoposide. This effect was more pronounced in DAOY cells expressing high levels of c-MYC when compared with DAOY wild-type or DAOY cells transfected with the empty vector.Conclusion: In human MB cells, in addition to its roles in growth and proliferation, c-MYC is also a potent inducer of apoptosis. Therefore, targeting c-MYC might be of therapeutic benefit when used sequentially with chemo- and radiotherapy rather than concomitantly. [ABSTRACT FROM AUTHOR]- Published
- 2009
- Full Text
- View/download PDF
19. Activation of canonical WNT/β-catenin signaling enhances in vitro motility of glioblastoma cells by activation of ZEB1 and other activators of epithelial-to-mesenchymal transition.
- Author
-
Kahlert UD, Maciaczyk D, Doostkam S, Orr BA, Simons B, Bogiel T, Reithmeier T, Prinz M, Schubert J, Niedermann G, Brabletz T, Eberhart CG, Nikkhah G, Maciaczyk J, Kahlert, Ulf D, Maciaczyk, Donata, Doostkam, Soroush, Orr, Brent A, Simons, Brian, and Bogiel, Tomasz
- Abstract
Here we show that activation of the canonical WNT/β-catenin pathway increases the expression of stem cell genes and promotes the migratory and invasive capacity of glioblastoma. Modulation of WNT signaling alters the expression of epithelial-to-mesenchymal transition activators, suggesting a role of this process in the regulation of glioma motility. Using immunohistochemistry in patient-derived glioblastoma samples we showed higher numbers of cells with intranuclear signal for β-catenin in the infiltrating edge of tumor compared to central tumor parenchyma. These findings suggest that canonical WNT/β-catenin pathway is a critical regulator of GBM invasion and may represent a potential therapeutic target. [ABSTRACT FROM AUTHOR]
- Published
- 2012
- Full Text
- View/download PDF
20. Intraorbital meningiomas: a pathologic review using current World Health Organization criteria.
- Author
-
Jain D, Ebrahimi KB, Miller NR, and Eberhart CG
- Published
- 2010
- Full Text
- View/download PDF
21. Intertumoral Heterogeneity within Medulloblastoma Subgroups
- Author
-
Luca Massimi, Caterina Giannini, Betty Luu, Cynthia Hawkins, Byung Kyu Cho, James T. Rutka, G. Yancey Gillespie, Timothy E. Van Meter, Almos Klekner, Young Shin Ra, Carolina Nor, Anna Goldenberg, Rajeev Vibhakar, Hideo Nakamura, Gaetano Finocchiaro, Massimo Zollo, John Peacock, Marta Perek-Polnik, Keren Isaev, Alvaro Lassaletta, Amulya A. Nageswara Rao, Florence M.G. Cavalli, Maura Massimino, Livia Garzia, Jüri Reimand, Charles G. Eberhart, Maryam Fouladi, Enrique López-Aguilar, Annie Huang, Cécile Faure-Conter, Gary D. Bader, Jaume Mora, Ho Keung Ng, James M. Olson, Jennifer A. Chan, Erwin G. Van Meir, Daniela Pretti da Cunha Tirapelli, Hamza Farooq, Fernando Chico Ponce de León, Linda M. Liau, Kay Ka Wai Li, Shenandoah Robinson, Anne Jouvet, Tomoko Shofuda, José Pimentel, Reid C. Thompson, Yuan Yao Thompson, Ian F. Pollack, Nada Jabado, Boleslaw Lach, Pim J. French, Joshua B. Rubin, Eric Bouffet, Alexandre Vasiljevic, Ali G. Saad, Michael K. Cooper, Satoru Osuka, Peter B. Dirks, Jaroslav Sterba, Johan M. Kros, Claudia C. Faria, Kyu-Chang Wang, Seung-Ki Kim, Shin Jung, Craig Daniels, A. Sorana Morrissy, Ladislav Rampášek, Andrew R. Hallahan, Sarah Leary, Wiesława Grajkowska, Uri Tabori, Marc Remke, Samer K. Elbabaa, Michael D. Taylor, Andrew S. Moore, Toshihiro Kumabe, Mario Perezpeña-Diazconti, Vijay Ramaswamy, Ronald L. Hamilton, Claudia M. Kuzan-Fischer, Marie Lise C. van Veelen, Helen Wheeler, Michal Zapotocky, Ji Yeoun Lee, David Shih, Jeffrey R. Leonard, Karel Zitterbart, Carlos Gilberto Carlotti, Steffen Albrecht, Jonathon Torchia, Peter Hauser, Ute Bartels, William A. Weiss, Sameer Agnihotri, Lola B. Chambless, Neurosurgery, Pathology, Neurology, Cavalli, Fmg, Remke, M, Rampasek, L, Peacock, J, Shih, Djh, Luu, B, Garzia, L, Torchia, J, Nor, C, Morrissy, A, Agnihotri, S, Thompson, Yy, Kuzan-Fischer, Cm, Farooq, H, Isaev, K, Daniels, C, Cho, Bk, Kim, Sk, Wang, Kc, Lee, Jy, Grajkowska, Wa, Perek-Polnik, M, Vasiljevic, A, Faure-Conter, C, Jouvet, A, Giannini, C, Nageswara Rao, Aa, Li, Kkw, Ng, Hk, Eberhart, Cg, Pollack, If, Hamilton, Rl, Gillespie, Gy, Olson, Jm, Leary, S, Weiss, Wa, Lach, B, Chambless, Lb, Thompson, Rc, Cooper, Mk, Vibhakar, R, Hauser, P, van Veelen, Mc, Kros, Jm, French, Pj, Ra, Y, Kumabe, T, López-Aguilar, E, Zitterbart, K, Sterba, J, Finocchiaro, G, Massimino, M, Van Meir, Eg, Osuka, S, Shofuda, T, Klekner, A, Zollo, M, Leonard, Jr, Rubin, Jb, Jabado, N, Albrecht, S, Mora, J, Van Meter, Te, Jung, S, Moore, A, Hallahan, Ar, Chan, Ja, Tirapelli, Dpc, Carlotti, Cg, Fouladi, M, Pimentel, J, Faria, Cc, Saad, Ag, Massimi, L, Liau, Lm, Wheeler, H, Nakamura, H, Elbabaa, Sk, Perezpeña-Diazconti, M, Chico Ponce de León, F, Robinson, S, Zapotocky, M, Lassaletta, A, Huang, Weiping, Hawkins, Ce, Tabori, U, Bouffet, E, Bartels, U, Dirks, Pb, Rutka, Jt, Bader, Gd, Reimand, J, Goldenberg, A, Ramaswamy, V, Taylor, Md, Cavalli F.M.G., Remke M., Rampasek L., Peacock J., Shih D.J.H., Luu B., Garzia L., Torchia J., Nor C., Morrissy A.S., Agnihotri S., Thompson Y.Y., Kuzan-Fischer C.M., Farooq H., Isaev K., Daniels C., Cho B.-K., Kim S.-K., Wang K.-C., Lee J.Y., Grajkowska W.A., Perek-Polnik M., Vasiljevic A., Faure-Conter C., Jouvet A., Giannini C., Nageswara Rao A.A., Li K.K.W., Ng H.-K., Eberhart C.G., Pollack I.F., Hamilton R.L., Gillespie G.Y., Olson J.M., Leary S., Weiss W.A., Lach B., Chambless L.B., Thompson R.C., Cooper M.K., Vibhakar R., Hauser P., van Veelen M.-L.C., Kros J.M., French P.J., Ra Y.S., Kumabe T., Lopez-Aguilar E., Zitterbart K., Sterba J., Finocchiaro G., Massimino M., Van Meir E.G., Osuka S., Shofuda T., Klekner A., Zollo M., Leonard J.R., Rubin J.B., Jabado N., Albrecht S., Mora J., Van Meter T.E., Jung S., Moore A.S., Hallahan A.R., Chan J.A., Tirapelli D.P.C., Carlotti C.G., Fouladi M., Pimentel J., Faria C.C., Saad A.G., Massimi L., Liau L.M., Wheeler H., Nakamura H., Elbabaa S.K., Perezpena-Diazconti M., Chico Ponce de Leon F., Robinson S., Zapotocky M., Lassaletta A., Huang A., Hawkins C.E., Tabori U., Bouffet E., Bartels U., Dirks P.B., Rutka J.T., Bader G.D., Reimand J., Goldenberg A., Ramaswamy V., and Taylor M.D.
- Subjects
0301 basic medicine ,Cancer Research ,medulloblastoma ,CURRENT CONSENSUS ,copy number ,Bioinformatics ,subgroups ,Cohort Studies ,Individual data ,Cluster Analysis ,R PACKAGE ,Precision Medicine ,GENECHIP DATA ,Sonic hedgehog ,ADULT MEDULLOBLASTOMA ,DNA Copy Number Variation ,Cancer ,Pediatric ,Genomics ,methylation ,CANCER ,3. Good health ,Oncology ,DNA methylation ,Human ,Pediatric Research Initiative ,DNA Copy Number Variations ,Pediatric Cancer ,Oncology and Carcinogenesis ,Computational biology ,Biology ,Article ,CLASSIFICATION ,Homogeneous clusters ,03 medical and health sciences ,Rare Diseases ,Genetics ,medicine ,Humans ,Oncology & Carcinogenesis ,Cerebellar Neoplasms ,RECURRENCE ,neoplasms ,Medulloblastoma ,Cluster Analysi ,gene expression ,MUTATIONS ,subgroup ,Gene Expression Profiling ,Human Genome ,Neurosciences ,integrative clustering ,DNA Methylation ,medicine.disease ,Precision medicine ,MEDULOBLASTOMA ,Brain Disorders ,nervous system diseases ,Brain Cancer ,Gene expression profiling ,stomatognathic diseases ,030104 developmental biology ,Genomic ,biology.protein ,MOLECULAR SUBGROUPS ,GENOMIC DATA ,Cohort Studie - Abstract
While molecular subgrouping has revolutionized medulloblastoma classification, the extent of heterogeneity within subgroups is unknown. Similarity network fusion (SNF) applied to genome-wide DNA methylation and gene expression data across 763 primary samples identifies very homogeneous clusters of patients, supporting the presence of medulloblastoma subtypes. After integration of somatic copy-number alterations, and clinical features specific to each cluster, we identify 12 different subtypes of medulloblastoma. Integrative analysis using SNF further delineates group 3 from group 4 medulloblastoma, which is not as readily apparent through analyses of individual data types. Two clear subtypes of infants with Sonic Hedgehog medulloblastoma with disparate outcomes and biology are identified. Medulloblastoma subtypes identified through integrative clustering have important implications for stratification of future clinical trials.
- Published
- 2017
22. Therapeutic impact of cytoreductive surgery and irradiation of posterior fossa ependymoma in the molecular era: A retrospective multicohort analysis
- Author
-
Uri Tabori, Marta M. Alonso, Frank S. Lieberman, James T. Rutka, Xing Fan, Kevin Petrecca, Michael A. Grotzer, Lyndsey Emery, Jennifer A. Chan, Luca Massimi, Elizabeth Vera-Bolanos, Antonio Omuro, Richard Everson, Stewart Goldman, Sarah Leary, Alvaro Lassaletta, Sofia Nunes, Ralph P. Ermoian, Betty Luu, Cynthia Hawkins, Amulya A. Nageswara Rao, Jeffrey R. Leonard, Maura Massimino, Teresa Tuñón, Massimo Zollo, James M. Olson, Almos Klekner, Andrey Korshunov, Ute Bartels, Tong Lin, Roger J. Packer, Matthias A. Karajannis, David W. Ellison, Christopher Dunham, David D. Eisenstat, Jaume Mora, Martin Mynarek, Erwin G. Van Meir, Roger E. McLendon, Karel Zitterbart, Corrine Gardner, Giuseppe Cinalli, Amar Gajjar, Kenneth Aldape, Karin M. Muraszko, Vijay Ramaswamy, Thomas Hielscher, Ronald L. Hamilton, Lola B. Chambless, Michael D. Prados, David T.W. Jones, Harshad Ladha, Horacio Soto, Wiesława Grajkowska, Jason E. Cain, Felice Giangaspero, Marcel Kool, Eric S. Lipp, William H. Yong, Andrew J. Grossbach, Tibor Hortobágyi, Tarek Shalaby, Helen Wheeler, Peter Hauser, Marie Lise C. van Veelen, Kristian W. Pajtler, Dorcas Fulton, Mariarita Santi, László Bognár, Jaroslav Sterba, Jing Wu, Ji Yeoun Lee, Carlos Gilberto Carlotti, Katja von Hoff, Stefan Rutkowski, Michael D. Taylor, Daniela Pretti da Cunha Tirapelli, Phillipe Metellus, Stephen C. Mack, Thomas E. Merchant, Samer K. Elbabaa, Marc Remke, Girish Dhall, Riccardo Soffietti, Eric Bouffet, Miguel A. Guzman, Khalida Wani, Charles G. Eberhart, Terri S. Armstrong, Tom Mikkelsen, Francesca R. Buttarelli, Nada Jabado, Paul G. Fisher, Juliette Hukin, Maryam Fouladi, Sama Ahsan, Sueli Mieko Oba-Shinjo, Linda M. Liau, Satoru Osuka, Sridharan Gururangan, Peter B. Dirks, Eugene Hwang, Howard Colman, H. Ian Robins, Caterina Giannini, Suely Kazue Nagahashi Marie, Frank van Landeghem, Mark R. Gilbert, Ulrich Schüller, Florence M.G. Cavalli, David Zagzag, Ian F. Pollack, Claudia C. Faria, Stefan M. Pfister, Shin Jung, Ramaswamy, V, Hielscher, T, Mack, Sc, Lassaletta, A, Lin, T, Pajtler, Kw, Jones, Dt, Luu, B, Cavalli, Fm, Aldape, K, Remke, M, Mynarek, M, Rutkowski, S, Gururangan, S, Mclendon, Re, Lipp, E, Dunham, C, Hukin, J, Eisenstat, Dd, Fulton, D, van Landeghem, Fk, Santi, M, van Veelen, Ml, Van Meir, Eg, Osuka, S, Fan, X, Muraszko, Km, Tirapelli, Dp, Oba Shinjo, Sm, Marie, Sk, Carlotti, Cg, Lee, Jy, Rao, Aa, Giannini, C, Faria, Cc, Nunes, S, Mora, J, Hamilton, Rl, Hauser, P, Jabado, N, Petrecca, K, Jung, S, Massimi, L, Zollo, Massimo, Cinalli, G, Bognár, L, Klekner, A, Hortobágyi, T, Leary, S, Ermoian, Rp, Olson, Jm, Leonard, Jr, Gardner, C, Grajkowska, Wa, Chambless, Lb, Cain, J, Eberhart, Cg, Ahsan, S, Massimino, M, Giangaspero, F, Buttarelli, Fr, Packer, Rj, Emery, L, Yong, Wh, Soto, H, Liau, Lm, Everson, R, Grossbach, A, Shalaby, T, Grotzer, M, Karajannis, Ma, Zagzag, D, Wheeler, H, von Hoff, K, Alonso, Mm, Tuñon, T, Schüller, U, Zitterbart, K, Sterba, J, Chan, Ja, Guzman, M, Elbabaa, Sk, Colman, H, Dhall, G, Fisher, Pg, Fouladi, M, Gajjar, A, Goldman, S, Hwang, E, Kool, M, Ladha, H, Vera Bolanos, E, Wani, K, Lieberman, F, Mikkelsen, T, Omuro, Am, Pollack, If, Prados, M, Robins, Hi, Soffietti, R, Wu, J, Metellus, P, Tabori, U, Bartels, U, Bouffet, E, Hawkins, Ce, Rutka, Jt, Dirks, P, Pfister, Sm, Merchant, Te, Gilbert, Mr, Armstrong, T, Korshunov, A, Ellison, Dw, Taylor, M. d., Ramaswamy V., Hielscher T., Mack S.C., Lassaletta A., Lin T., Pajtler K.W., Jones D.T.W., Luu B., Cavalli F.M.G., Aldape K., Remke M., Mynarek M., Rutkowski S., Gururangan S., McLendon R.E., Lipp E.S., Dunham C., Hukin J., Eisenstat D.D., Fulton D., Van Landeghem F.K.H., Santi M., Van Veelen M.-L.C., Van Meir E.G., Osuka S., Fan X., Muraszko K.M., Tirapelli D.P.C., Oba-Shinjo S.M., Marie S.K.N., Carlotti C.G., Lee J.Y., Rao A.A.N., Giannini C., Faria C.C., Nunes S., Mora J., Hamilton R.L., Hauser P., Jabado N., Petrecca K., Jung S., Massimi L., Zollo M., Cinalli G., Bognar L., Klekner A., Hortobagyi T., Leary S., Ermoian R.P., Olson J.M., Leonard J.R., Gardner C., Grajkowska W.A., Chambless L.B., Cain J., Eberhart C.G., Ahsan S., Massimino M., Giangaspero F., Buttarelli F.R., Packer R.J., Emery L., Yong W.H., Soto H., Liau L.M., Everson R., Grossbach A., Shalaby T., Grotzer M., Karajannis M.A., Zagzag D., Wheeler H., Von Hoff K., Alonso M.M., Tunon T., Schuller U., Zitterbart K., Sterba J., Chan J.A., Guzman M., Elbabaa S.K., Colman H., Dhall G., Fisher P.G., Fouladi M., Gajjar A., Goldman S., Hwang E., Kool M., Ladha H., Vera-Bolanos E., Wani K., Lieberman F., Mikkelsen T., Omuro A.M., Pollack I.F., Prados M., Robins H.I., Soffietti R., Wu J., Metellus P., Tabori U., Bartels U., Bouffet E., Hawkins C.E., Rutka J.T., Dirks P., Pfister S.M., Merchant T.E., Gilbert M.R., Armstrong T.S., Korshunov A., Ellison D.W., Taylor M.D., and Neurosurgery
- Subjects
Male ,Ependymoma ,Cancer Research ,medicine.medical_treatment ,cancer research ,oncology ,posterior fossa ependymoma ,cytoreductive surgery ,radiation therapy ,Infratentorial Neoplasms ,Cohort Studies ,0302 clinical medicine ,Retrospective Studie ,Medicine ,Pediatric ependymoma ,Child ,10. No inequality ,Infratentorial Neoplasm ,biology ,Cytoreduction Surgical Procedures ,Combined Modality Therapy ,3. Good health ,Oncology ,Child, Preschool ,030220 oncology & carcinogenesis ,Female ,Radiology ,Human ,Adult ,medicine.medical_specialty ,Adolescent ,Fossa ,Ependymoma, biomarkers ,03 medical and health sciences ,Original Reports ,Humans ,Retrospective Studies ,Chemotherapy ,business.industry ,Proportional hazards model ,Infant ,Retrospective cohort study ,biology.organism_classification ,medicine.disease ,Surgery ,Radiation therapy ,Cohort Studie ,business ,030217 neurology & neurosurgery - Abstract
Purpose Posterior fossa ependymoma comprises two distinct molecular variants termed EPN_PFA and EPN_PFB that have a distinct biology and natural history. The therapeutic value of cytoreductive surgery and radiation therapy for posterior fossa ependymoma after accounting for molecular subgroup is not known. Methods Four independent nonoverlapping retrospective cohorts of posterior fossa ependymomas (n = 820) were profiled using genome-wide methylation arrays. Risk stratification models were designed based on known clinical and newly described molecular biomarkers identified by multivariable Cox proportional hazards analyses. Results Molecular subgroup is a powerful independent predictor of outcome even when accounting for age or treatment regimen. Incompletely resected EPN_PFA ependymomas have a dismal prognosis, with a 5-year progression-free survival ranging from 26.1% to 56.8% across all four cohorts. Although first-line (adjuvant) radiation is clearly beneficial for completely resected EPN_PFA, a substantial proportion of patients with EPN_PFB can be cured with surgery alone, and patients with relapsed EPN_PFB can often be treated successfully with delayed external-beam irradiation. Conclusion The most impactful biomarker for posterior fossa ependymoma is molecular subgroup affiliation, independent of other demographic or treatment variables. However, both EPN_PFA and EPN_PFB still benefit from increased extent of resection, with the survival rates being particularly poor for subtotally resected EPN_PFA, even with adjuvant radiation therapy. Patients with EPN_PFB who undergo gross total resection are at lower risk for relapse and should be considered for inclusion in a randomized clinical trial of observation alone with radiation reserved for those who experience recurrence.
- Published
- 2016
23. Cytogenetic Prognostication Within Medulloblastoma Subgroups
- Author
-
Franck Bourdeaut, Michael K. Cooper, Almos Klekner, Stefan Rutkowski, David Shih, Jeffrey R. Leonard, Simon Bailey, Concezio Di Rocco, Ulrich Schüller, Giuseppe Cinalli, Pim J. French, Cinzia Lavarino, Young Shin Ra, John Peacock, Yuan Yao, A. Sorana Morrissy, William A. Weiss, Nanne K. Kloosterhof, László Bognár, Pasqualino De Antonellis, Teiji Tominaga, Stephen W. Scherer, Paul A. Northcott, David T.W. Jones, Reid C. Thompson, Shenandoah Robinson, Marta Perek-Polnik, Boleslaw Lach, Massimo Zollo, Olivier Delattre, Jennifer A. Chan, Janet C. Lindsey, Xin Wang, Nada Jabado, Karel Zitterbart, David Malkin, Adi Rolider, Roger J. Packer, James M. Olson, Steffen Albrecht, Ji Yeoun Lee, Wiesława Grajkowska, Charles G. Eberhart, Marcel Kool, Vijay Ramaswamy, Seung-Ki Kim, Joanna J. Phillips, Andrey Korshunov, Michael D. Taylor, Florence M.G. Cavalli, Luca Massimi, Xiaochong Wu, Maryam Fouladi, Peter Hauser, Xing Fan, Steven C. Clifford, Leos Kren, Carmen de Torres, Erna M.C. Michiels, Adrian M. Dubuc, Amar Gajjar, Livia Garzia, Eric Bouffet, Ian F. Pollack, Marc Remke, Jaume Mora, Claudia C. Faria, Miklós Garami, Erwin G. Van Meir, Byung Kyu Cho, Karin M. Muraszko, Joshua B. Rubin, Anne Jouvet, Stefan M. Pfister, Nalin Gupta, Johan M. Kros, Shin Jung, Yoon Jae Cho, Rajeev Vibhakar, Linda M. Liau, Stephen C. Mack, Scott L. Pomeroy, Betty Luu, Cynthia Hawkins, Ali G. Saad, Kyu-Chang Wang, Uri Tabori, Michelle Fèvre-Montange, Adam M. Fontebasso, Robert J. Wechsler-Reya, Toshihiro Kumabe, James T. Rutka, François Doz, Shih, Dj, Northcott, Pa, Remke, M, Korshunov, A, Ramaswamy, V, Kool, M, Luu, B, Yao, Y, Wang, X, Dubuc, Am, Garzia, L, Peacock, J, Mack, Sc, Wu, X, Rolider, A, Morrissy, A, Cavalli, Fm, Jones, Dt, Zitterbart, K, Faria, Cc, Schüller, U, Kren, L, Kumabe, T, Tominaga, T, Shin Ra, Y, Garami, M, Hauser, P, Chan, Ja, Robinson, S, Bognár, L, Klekner, A, Saad, Ag, Liau, Lm, Albrecht, S, Fontebasso, A, Cinalli, G, DE ANTONELLIS, Pasqualino, Zollo, Massimo, Cooper, Mk, Thompson, Rc, Bailey, S, Lindsey, Jc, Di Rocco, C, Massimi, L, Michiels, Em, Scherer, Sw, Phillips, Jj, Gupta, N, Fan, X, Muraszko, Km, Vibhakar, R, Eberhart, Cg, Fouladi, M, Lach, B, Jung, S, Wechsler Reya, Rj, Fèvre Montange, M, Jouvet, A, Jabado, N, Pollack, If, Weiss, Wa, Lee, Jy, Cho, Bk, Kim, Sk, Wang, Kc, Leonard, Jr, Rubin, Jb, de Torres, C, Lavarino, C, Mora, J, Cho, Yj, Tabori, U, Olson, Jm, Gajjar, A, Packer, Rj, Rutkowski, S, Pomeroy, Sl, French, Pj, Kloosterhof, Nk, Kros, Jm, Van Meir, Eg, Clifford, Sc, Bourdeaut, F, Delattre, O, Doz, Ff, Hawkins, Ce, Malkin, D, Grajkowska, Wa, Perek Polnik, M, Bouffet, E, Rutka, Jt, Pfister, Sm, Taylor, M. d., Pulmonary Medicine, Pediatrics, Neurology, and Pathology
- Subjects
Male ,Cancer Research ,Pathology ,Risk Factors ,Young adult ,Stage (cooking) ,Pair 11 ,Child ,In Situ Hybridization, Fluorescence ,In Situ Hybridization ,Cancer ,Pediatric ,screening and diagnosis ,Tumor ,Nuclear Proteins ,ORIGINAL REPORTS ,Orvostudományok ,Prognosis ,3. Good health ,Gene Expression Regulation, Neoplastic ,Detection ,Oncology ,Child, Preschool ,Predictive value of tests ,Female ,Patient Safety ,Biotechnology ,4.2 Evaluation of markers and technologies ,Human ,medicine.medical_specialty ,Pediatric Research Initiative ,Adolescent ,Pediatric Cancer ,Clinical Sciences ,Oncology and Carcinogenesis ,Kruppel-Like Transcription Factors ,Zinc Finger Protein Gli2 ,Extent of resection ,Klinikai orvostudományok ,Risk Assessment ,Chromosomes ,Fluorescence ,Proto-Oncogene Proteins c-myc ,Cytogenetics ,Young Adult ,Rare Diseases ,Patient age ,Clinical Research ,Predictive Value of Tests ,Biomarkers, Tumor ,medicine ,Humans ,Hedgehog Proteins ,Oncology & Carcinogenesis ,Preschool ,Proportional Hazards Models ,Chromosomes, Human, Pair 14 ,Medulloblastoma ,Neoplastic ,business.industry ,Proportional hazards model ,Chromosomes, Human, Pair 11 ,Gene Expression Profiling ,Pair 14 ,Reproducibility of Results ,Infant ,medicine.disease ,Brain Disorders ,4.1 Discovery and preclinical testing of markers and technologies ,Brain Cancer ,Wnt Proteins ,Gene Expression Regulation ,Tissue Array Analysis ,business ,Biomarkers - Abstract
Purpose Medulloblastoma comprises four distinct molecular subgroups: WNT, SHH, Group 3, and Group 4. Current medulloblastoma protocols stratify patients based on clinical features: patient age, metastatic stage, extent of resection, and histologic variant. Stark prognostic and genetic differences among the four subgroups suggest that subgroup-specific molecular biomarkers could improve patient prognostication. Patients and Methods Molecular biomarkers were identified from a discovery set of 673 medulloblastomas from 43 cities around the world. Combined risk stratification models were designed based on clinical and cytogenetic biomarkers identified by multivariable Cox proportional hazards analyses. Identified biomarkers were tested using fluorescent in situ hybridization (FISH) on a nonoverlapping medulloblastoma tissue microarray (n = 453), with subsequent validation of the risk stratification models. Results Subgroup information improves the predictive accuracy of a multivariable survival model compared with clinical biomarkers alone. Most previously published cytogenetic biomarkers are only prognostic within a single medulloblastoma subgroup. Profiling six FISH biomarkers (GLI2, MYC, chromosome 11 [chr11], chr14, 17p, and 17q) on formalin-fixed paraffin-embedded tissues, we can reliably and reproducibly identify very low-risk and very high-risk patients within SHH, Group 3, and Group 4 medulloblastomas. Conclusion Combining subgroup and cytogenetic biomarkers with established clinical biomarkers substantially improves patient prognostication, even in the context of heterogeneous clinical therapies. The prognostic significance of most molecular biomarkers is restricted to a specific subgroup. We have identified a small panel of cytogenetic biomarkers that reliably identifies very high-risk and very low-risk groups of patients, making it an excellent tool for selecting patients for therapy intensification and therapy de-escalation in future clinical trials.
- Published
- 2014
24. Subgroup-specific structural variation across 1,000 medulloblastoma genomes
- Author
-
Jenny Q. Qian, Darell D. Bigner, Miklós Garami, Shaun D. Jackman, Wiesława Grajkowska, Nalin Gupta, Johan M. Kros, Poul H. Sorensen, Anna Kenney, Stéphanie Reynaud, Byung Kyu Cho, Ian F. Pollack, Marcel Kool, Steven C. Clifford, Kyu-Chang Wang, Inanc Birol, Tzvi Aviv, Hendrick Witt, Gemma Hoad, Martine F. Roussel, Christine Haberler, Pim J. French, Betty Luu, Cynthia Hawkins, Claudia C. Faria, Richard A. Moore, Karin M. Muraszko, Yuan Yao, Nanne K. Kloosterhof, Rameen Beroukhim, Leos Kren, Erna M.C. Michiels, Jan O. Korbel, Paul A. Northcott, Stefan M. Pfister, Marc Remke, Nina Thiessen, Jennifer A. Chan, Adam M. Fontebasso, Maryam Fouladi, Shin Jung, Richard G. Ellenbogen, Richard Corbett, László Bognár, Yoon Jae Cho, Massimo Zollo, Robert J. Wechsler-Reya, Steven E. Schumacher, Xing Fan, Michael J. Levy, Wolfram Scheurlen, Young Shin Ra, Adrian M. Stütz, William A. Weiss, Simon Bailey, Rajeev Vibhakar, Giuseppe Cinalli, Toshihiro Kumabe, Marco A. Marra, Christian R. Marshall, Eric Bouffet, Luca Massimi, Scott L. Pomeroy, Sarah S. Pernet-Fattet, Andrew J. Mungall, James T. Rutka, G. Yancey Gillespie, Charles G. Eberhart, Peter Hauser, Andy Chu, Jüri Reimand, Xiaochong Wu, Adi Rolider, Xin Wang, Stephen W. Scherer, Reid C. Thompson, Ka Ming Nip, Anne Jouvet, Timothy E. Van Meter, Robert A. Holt, Anthony Raymond, Livia Garzia, Teiji Tominaga, Erwin G. Van Meir, John Peacock, Michael D. Taylor, Achille Iolascon, Roger E. McLendon, Andrey Korshunov, Stephen C. Mack, Nada Jabado, Readman Chiu, Africa Fernandez-L, Eric Chuah, Richard Varhol, Hideo Nakamura, Samer K. Elbabaa, Uri Tabori, Peter B. Dirks, Gary D. Bader, Linda M. Liau, François Doz, Allan Lo, Janet C. Lindsey, Adrian M. Dubuc, Michelle Fèvre-Montange, David T.W. Jones, Carlos Gilberto Carlotti, Ali G. Saad, Steffen Albrecht, Michael K. Cooper, Karen Mungall, Daisuke Kawauchi, A. Sorana Morrissy, Boleslaw Lach, Karel Zitterbart, Joshua B. Rubin, Matthew Meyerson, Florence M.G. Cavalli, Yisu Li, Shenandoah Robinson, Marta Perek-Polnik, Olivier Delattre, David Malkin, Almos Klekner, James M. Olson, Steven J.M. Jones, Thomas Zichner, David W. Ellison, Seung-Ki Kim, Vijay Ramaswamy, Anath C. Lionel, David Shih, Jeffrey R. Leonard, Concezio Di Rocco, Pulmonary Medicine, Pediatrics, Neurology, Pathology, Northcott, Pa, Shih, Dj, Peacock, J, Garzia, L, Morrissy, A, Zichner, T, Stütz, Am, Korshunov, A, Reimand, J, Schumacher, Se, Beroukhim, R, Ellison, Dw, Marshall, Cr, Lionel, Ac, Mack, S, Dubuc, A, Yao, Y, Ramaswamy, V, Luu, B, Rolider, A, Cavalli, Fm, Wang, X, Remke, M, Wu, X, Chiu, Ry, Chu, A, Chuah, E, Corbett, Rd, Hoad, Gr, Jackman, Sd, Li, Y, Lo, A, Mungall, Kl, Nip, Km, Qian, Jq, Raymond, Ag, Thiessen, Nt, Varhol, Rj, Birol, I, Moore, Ra, Mungall, Aj, Holt, R, Kawauchi, D, Roussel, Mf, Kool, M, Jones, Dt, Witt, H, Fernandez L., A, Kenney, Am, Wechsler Reya, Rj, Dirks, P, Aviv, T, Grajkowska, Wa, Perek Polnik, M, Haberler, Cc, Delattre, O, Reynaud, S, Doz, Ff, Pernet Fattet, S, Cho, Bk, Kim, Sk, Wang, Kc, Scheurlen, W, Eberhart, Cg, Fèvre Montange, M, Jouvet, A, Pollack, If, Fan, X, Muraszko, Km, Gillespie, Gy, Di Rocco, C, Massimi, L, Michiels, Em, Kloosterhof, Nk, French, Pj, Kros, Jm, Olson, Jm, Ellenbogen, Rg, Zitterbart, K, Kren, L, Thompson, Rc, Cooper, Mk, Lach, B, Mclendon, Re, Bigner, Dd, Fontebasso, A, Albrecht, S, Jabado, N, Lindsey, Jc, Bailey, S, Gupta, N, Weiss, Wa, Bognár, L, Klekner, A, Van Meter, Te, Kumabe, T, Tominaga, T, Elbabaa, Sk, Leonard, Jr, Rubin, Jb, Liau, Lm, Van Meir, Eg, Fouladi, M, Nakamura, H, Cinalli, G, Garami, M, Hauser, P, Saad, Ag, Iolascon, Achille, Jung, S, Carlotti, Cg, Vibhakar, R, Ra, Y, Robinson, S, Zollo, Massimo, Faria, Cc, Chan, Ja, Levy, Ml, Sorensen, Ph, Meyerson, M, Pomeroy, Sl, Cho, Yj, Bader, Gd, Tabori, U, Hawkins, Ce, Bouffet, E, Scherer, Sw, Rutka, Jt, Malkin, D, Clifford, Sc, Jones, Sj, Korbel, Jo, Pfister, Sm, Marra, Ma, and Taylor, M. D.
- Subjects
DNA Copy Number Variations ,Oncogene Proteins, Fusion ,medicine.medical_treatment ,Genes, myc ,Nerve Tissue Proteins ,Biology ,Bioinformatics ,medulloblastoma ,Article ,Translocation, Genetic ,Targeted therapy ,Structural variation ,03 medical and health sciences ,0302 clinical medicine ,Transforming Growth Factor beta ,Gene Duplication ,Gene duplication ,medicine ,Humans ,Hedgehog Proteins ,Cerebellar Neoplasms ,Child ,030304 developmental biology ,Medulloblastoma ,0303 health sciences ,Multidisciplinary ,Chromothripsis ,PROTEÍNAS DE TRANSPORTE (GENÉTICA) ,Genome, Human ,NF-kappa B ,Cancer ,Proteins ,Genomics ,medicine.disease ,Human genetics ,3. Good health ,PVT1 ,030220 oncology & carcinogenesis ,Genomic Structural Variation ,RNA, Long Noncoding ,Carrier Proteins ,Signal Transduction - Abstract
Medulloblastoma, the most common malignant paediatric brain tumour, is currently treated with nonspecific cytotoxic therapies including surgery, whole-brain radiation, and aggressive chemotherapy. As medulloblastoma exhibits marked intertumoural heterogeneity, with at least four distinct molecular variants, previous attempts to identify targets for therapy have been underpowered because of small samples sizes. Here we report somatic copy number aberrations (SCNAs) in 1,087 unique medulloblastomas. SCNAs are common in medulloblastoma, and are predominantly subgroup-enriched. The most common region of focal copy number gain is a tandem duplication of SNCAIP, a gene associated with Parkinson's disease, which is exquisitely restricted to Group 4 alpha. Recurrent translocations of PVT1, including PVT1-MYC and PVT1-NDRG1, that arise through chromothripsis are restricted to Group 3. Numerous targetable SCNAs, including recurrent events targeting TGF-beta signalling in Group 3, and NF-kappa B signalling in Group 4, suggest future avenues for rational, targeted therapy.
- Published
- 2012
25. Transforming growth factor-β signaling pathway activation in Keratoconus.
- Author
-
Engler C, Chakravarti S, Doyle J, Eberhart CG, Meng H, Stark WJ, Kelliher C, Jun AS, Engler, Christoph, Chakravarti, Shukti, Doyle, Jefferson, Eberhart, Charles G, Meng, Huan, Stark, Walter J, Kelliher, Clare, and Jun, Albert S
- Abstract
Purpose: To assess the presence of transforming growth factor-β (TGFβ) pathway markers in the epithelium of keratoconus patient corneas.Design: Retrospective, comparative case series of laboratory specimens.Methods: Immunohistochemistry results for TGFβ2, total TGFβ, mothers against decacentaplegic homolog (Smad) 2, and phosphorylated Smad2 was performed on formalin-fixed, paraffin-embedded sections of keratoconus patient corneas and normal corneas from human autopsy eyes. Keratoconus patient corneas were divided in two groups, depending on their severity based on keratometer readings and pachymetry. Autopsy controls were age-matched with the keratoconus cases. Immunohistochemistry signal quantification was performed using automated software. Real-time reverse-transcriptase polymerase chain reaction was performed on total ribonucleic acid of epithelium of keratoconus patient corneas and autopsy control corneas.Results: Immunohistochemistry quantification showed a significant increase in mean signal in the group of severe keratoconus cases compared with normal corneas for TGFβ2 and phosphorylated Smad2 (P < .05). Immunohistochemistry analysis using antibodies against total TGFβ and Smad2 did not show any significant increase in the keratoconus cases versus the autopsy controls. Reverse-transcriptase polymerase chain reaction exhibited elevated messenger ribonucleic acid levels of Smad2 and TGFβ2 in severe keratoconus corneal epithelium.Conclusions: This work shows increased TGFβ pathway markers in severe keratoconus cases and provides the rationale for investigating TGFβ signaling further in the pathophysiology of keratoconus. [ABSTRACT FROM AUTHOR]- Published
- 2011
- Full Text
- View/download PDF
26. Neurobrucellosis presenting as leukoencephalopathy.
- Author
-
Seidel G, Pardo CA, Newman-Toker D, Olivi A, and Eberhart CG
- Abstract
A 65-year-old man developed a leukoencephalopathy associated with neurobrucellosis. The disease followed a 15-month progressive course with neurologic symptoms, and magnetic resonance imaging revealed bilateral symmetrical T2 signal hyperintensities in the white matter. Biopsy of the cerebral cortex and white matter was significant for nongranulomatous meningoencephalitis with reactive microgliosis and astrogliosis. The inflammatory infiltrate was predominantly composed of T lymphocytes, including numerous cytotoxic T cells. There was no evidence of significant myelin destruction. No organisms were detected microscopically, but elevated immunoglobulin G titers to Brucella were found in the cerebrospinal fluid. An abscess formed at the biopsy site, and Brucella melitensiswas cultured from abscess contents. Neurobrucellosis is difficult to diagnose outside endemic regions and is associated with leukoencephalopathy-like pathology. Cytotoxic T lymphocytes and microglia activation play an immunopathogenic role in this rare disease. [ABSTRACT FROM AUTHOR]
- Published
- 2003
- Full Text
- View/download PDF
27. Establishment and Characterization of Three Human Ocular Adnexal Sebaceous Carcinoma Cell Lines.
- Author
-
Lee SC, Peterson C, Wang K, Alaali L, Eshleman J, Mahoney NR, Li E, Eberhart CG, and Campbell AA
- Subjects
- Humans, Female, Cell Line, Tumor, Male, Cell Proliferation drug effects, Adenocarcinoma, Sebaceous pathology, Adenocarcinoma, Sebaceous metabolism, Adenocarcinoma, Sebaceous genetics, Apoptosis, Mitomycin pharmacology, Cell Differentiation, Aged, Eye Neoplasms pathology, Eye Neoplasms metabolism, Eye Neoplasms genetics, Fluorouracil pharmacology, Middle Aged, Sebaceous Gland Neoplasms pathology, Sebaceous Gland Neoplasms metabolism, Sebaceous Gland Neoplasms genetics
- Abstract
Ocular adnexal sebaceous carcinoma (SebCA) represents one of the most clinically problematic periocular tumors, often requiring aggressive surgical resection. The pathobiology of this tumor remains poorly understood, and few models exist that are suitable for preclinical testing. The aim of this study was to establish new cell lines to serve as models for pathobiological and drug testing. With patient consent, freshly resected tumor tissue was cultured using conditional reprogramming cell conditions. Standard techniques were used to characterize the cell lines in terms of overall growth, clonogenicity, apoptosis, and differentiation in vitro. Additional analyses including Western blotting, short tandem repeat (STR) profiling, and next-generation sequencing (NGS) were performed. Drug screening using mitomycin-C (MMC), 5-fluorouricil (5-FU), and 6-Diazo-5-oxo-L-norleucine (DON) were performed. JHH-SebCA01, JHH-SebCA02, and JHH-SebCA03 cell lines were established from two women and one man undergoing surgical resection of eyelid tumors. At passage 15, they each showed a doubling time of two to three days, and all could form colonies in anchorage-dependent conditions, but not in soft agar. The cells contained cytoplasmic vacuoles consistent with sebaceous differentiation, and adipophilin protein was present in all three lines. STR profiling confirmed that all lines were derived from their respective patients. NGS of the primary tumors and their matched cell lines identified numerous shared mutations, including alterations similar to those previously described in SebCA. Treatment with MMC or 5-FU resulted in dose-dependent growth inhibition and the induction of both apoptosis and differentiation. MYC protein was abundant in all three lines, and the glutamine metabolism inhibitor DON, previously shown to target high MYC tumors, slowed the growth of all our SebCA models. Ocular adnexal SebCA cell lines can be established using conditional reprogramming cell conditions, and our three new models are useful for testing therapies and interrogating the functional role of MYC and other possible molecular drivers. Current topical chemotherapies promote both apoptosis and differentiation in SebCA cells, and these tumors appear sensitive to inhibition or MYC-associated metabolic changes.
- Published
- 2024
- Full Text
- View/download PDF
28. Brain Nucleic Acid Delivery and Genome Editing via Focused Ultrasound-Mediated Blood-Brain Barrier Opening and Long-Circulating Nanoparticles.
- Author
-
Kwak G, Grewal A, Slika H, Mess G, Li H, Kwatra M, Poulopoulos A, Woodworth GF, Eberhart CG, Ko HS, Manbachi A, Caplan J, Price RJ, Tyler B, and Suk JS
- Subjects
- Animals, Brain metabolism, Mice, Ultrasonic Waves, Astrocytes metabolism, DNA chemistry, DNA administration & dosage, Polymers chemistry, RNA, Messenger metabolism, RNA, Messenger genetics, Neurons metabolism, Gene Transfer Techniques, Plasmids administration & dosage, Plasmids genetics, Nucleic Acids chemistry, Nucleic Acids administration & dosage, Nucleic Acids metabolism, Humans, Blood-Brain Barrier metabolism, Nanoparticles chemistry, Gene Editing methods
- Abstract
We introduce a two-pronged strategy comprising focused ultrasound (FUS)-mediated blood-brain barrier (BBB) opening and long-circulating biodegradable nanoparticles (NPs) for systemic delivery of nucleic acids to the brain. Biodegradable poly(β-amino ester) polymer-based NPs were engineered to stably package various types of nucleic acid payloads and enable prolonged systemic circulation while retaining excellent serum stability. FUS was applied to a predetermined coordinate within the brain to transiently open the BBB, thereby allowing the systemically administered long-circulating NPs to traverse the BBB and accumulate in the FUS-treated brain region, where plasmid DNA or mRNA payloads produced reporter proteins in astrocytes and neurons. In contrast, poorly circulating and/or serum-unstable NPs, including the lipid NP analogous to a platform used in clinic, were unable to provide efficient nucleic acid delivery to the brain regardless of the BBB-opening FUS. The marriage of FUS-mediated BBB opening and the long-circulating NPs engineered to copackage mRNA encoding CRISPR-associated protein 9 and single-guide RNA resulted in genome editing in astrocytes and neurons precisely in the FUS-treated brain region. The combined delivery strategy provides a versatile means to achieve efficient and site-specific therapeutic nucleic acid delivery to and genome editing in the brain via a systemic route.
- Published
- 2024
- Full Text
- View/download PDF
29. Systematic transcriptomic analysis of childhood medulloblastoma identifies N6- methyladenosine-dependent lncRNA signatures associated with molecular subtype, immune cell in fi ltration, and prognosis.
- Author
-
Joshi K, Yuan M, Katsushima K, Saulnier O, Ray A, Amankwah E, Stapleton S, Jallo G, Taylor MD, Eberhart CG, and Perera RJ
- Abstract
Medulloblastoma, the most common malignant pediatric brain tumor, is classified into four main molecular subgroups, but group 3 and group 4 tumors are difficult to subclassify and have a poor prognosis. Rapid point-of-care diagnostic and prognostic assays are needed to improve medulloblastoma risk stratification and management. N6-methyladenosine (m6A) is a common RNA modification and long non-coding RNAs (lncRNAs) play a central role in tumor progression, but their impact on gene expression and associated clinical outcomes in medulloblastoma are unknown. Here we analyzed 469 medulloblastoma tumor transcriptomes to identify lncRNAs co-expressed with m6A regulators. Using LASSO-Cox analysis, we identified a five-gene m6A-associated lncRNA signature (M6LSig) significantly associated with overall survival, which was combined in a prognostic clinical nomogram. Using expression of the 67 m6A-associated lncRNAs, a subgroup classification model was generated using the XGBoost machine learning algorithm, which had a classification accuracy > 90%, including for group 3 and 4 samples. All M6LSig genes were significantly correlated with at least one immune cell type abundance in the tumor microenvironment, and the risk score was positively correlated with CD4
+ naïve T cell abundance and negatively correlated with follicular helper T cells and eosinophils. Knockdown of key m6A writer genes METTL3 and METTL14 in a group 3 medulloblastoma cell line (D425-Med) decreased cell proliferation and upregulated many M6LSig genes identified in our in silico analysis, suggesting that the signature genes are functional in medulloblastoma. This study highlights a crucial role for m6A-dependent lncRNAs in medulloblastoma prognosis and immune responses and provides the foundation for practical clinical tools that can be rapidly deployed in clinical settings., Competing Interests: Competing interests The authors declare no competing interests.- Published
- 2024
- Full Text
- View/download PDF
30. Systematic transcriptomic analysis of childhood medulloblastoma identifies N6-methyladenosine-dependent lncRNA signatures associated with molecular subtype, immune cell infiltration, and prognosis.
- Author
-
Joshi K, Yuan M, Katsushima K, Saulnier O, Ray A, Amankwah E, Stapleton S, Jallo G, Taylor MD, Eberhart CG, and Perera RJ
- Subjects
- Humans, Prognosis, Child, Gene Expression Profiling methods, Male, Female, Tumor Microenvironment genetics, Tumor Microenvironment immunology, Methyltransferases, Medulloblastoma genetics, Medulloblastoma pathology, Medulloblastoma metabolism, RNA, Long Noncoding genetics, RNA, Long Noncoding metabolism, Adenosine analogs & derivatives, Adenosine metabolism, Cerebellar Neoplasms genetics, Cerebellar Neoplasms pathology, Transcriptome
- Abstract
Medulloblastoma, the most common malignant pediatric brain tumor, is classified into four main molecular subgroups, but group 3 and group 4 tumors are difficult to subclassify and have a poor prognosis. Rapid point-of-care diagnostic and prognostic assays are needed to improve medulloblastoma risk stratification and management. N6-methyladenosine (m6A) is a common RNA modification and long non-coding RNAs (lncRNAs) play a central role in tumor progression, but their impact on gene expression and associated clinical outcomes in medulloblastoma are unknown. Here we analyzed 469 medulloblastoma tumor transcriptomes to identify lncRNAs co-expressed with m6A regulators. Using LASSO-Cox analysis, we identified a five-gene m6A-associated lncRNA signature (M6LSig) significantly associated with overall survival, which was combined in a prognostic clinical nomogram. Using expression of the 67 m6A-associated lncRNAs, a subgroup classification model was generated using the XGBoost machine learning algorithm, which had a classification accuracy > 90%, including for group 3 and 4 samples. All M6LSig genes were significantly correlated with at least one immune cell type abundance in the tumor microenvironment, and the risk score was positively correlated with CD4
+ naïve T cell abundance and negatively correlated with follicular helper T cells and eosinophils. Knockdown of key m6A writer genes METTL3 and METTL14 in a group 3 medulloblastoma cell line (D425-Med) decreased cell proliferation and upregulated many M6LSig genes identified in our in silico analysis, suggesting that the signature genes are functional in medulloblastoma. This study highlights a crucial role for m6A-dependent lncRNAs in medulloblastoma prognosis and immune responses and provides the foundation for practical clinical tools that can be rapidly deployed in clinical settings., (© 2024. The Author(s).)- Published
- 2024
- Full Text
- View/download PDF
31. Teaching NeuroImage: Giant Cell Arteritis With Optic Perineuritis.
- Author
-
Taga A, Wali A, Eberhart CG, and Green KE
- Subjects
- Humans, Magnetic Resonance Imaging, Female, Aged, Male, Giant Cell Arteritis complications, Giant Cell Arteritis diagnostic imaging, Optic Neuritis diagnostic imaging
- Published
- 2024
- Full Text
- View/download PDF
32. ASXL1 inactivation and reduced H3K27me3 across central nervous system tumors.
- Author
-
Zhang KY, Parker M, Weber-Levine C, Kalluri A, Gonzalez-Gomez I, Raabe E, Dudley JC, Gocke C, Lin MT, Zou Y, Sherief M, Kamson DO, Holdhoff M, Mukherjee D, Croog V, Schreck KC, Rincon-Torroella J, Bettegowda C, Eberhart CG, Bale T, and Lucas CG
- Subjects
- Humans, Central Nervous System Neoplasms metabolism, Central Nervous System Neoplasms pathology, Histones metabolism, Histones genetics, Repressor Proteins genetics, Repressor Proteins metabolism
- Published
- 2024
- Full Text
- View/download PDF
33. Ocular adnexal sebaceous carcinoma in a patient with Li-Fraumeni syndrome.
- Author
-
Hsu CW, Peterson CW, Eberhart CG, Meyer CF, Armstrong DK, Fiallos K, and Campbell AA
- Abstract
Li-Fraumeni syndrome (LFS) is caused by a pathogenic germline variant at the TP53 locus and is associated with an increased predisposition to a variety of cancers. The neoplasms most frequently associated with LFS are sarcomas, breast cancer, brain tumors, and adrenocortical carcinomas. In this case report, we present a 43-year-old male diagnosed with an ocular adnexal sebaceous carcinoma of the right upper eyelid who was confirmed to have LFS with subsequent genetic testing. The mutational profile of both the patient's genetic screen and tumor sequencing were congruent, demonstrating the same pathogenic loss-of-function TP53 variant. This case report highlights the importance of pursuing genetic testing in patients with a history of multiple tumor types, particularly those with uncommon diagnoses. In this case, confirmation of LFS had important implications for personalized patient care, including identification of contraindicated treatment interventions and the imaging modalities necessary for vigilant follow-up screening.
- Published
- 2024
- Full Text
- View/download PDF
34. VISTA Emerges as a Promising Target against Immune Evasion Mechanisms in Medulloblastoma.
- Author
-
Muñoz Perez N, Pensabene JM, Galbo PM Jr, Sadeghipour N, Xiu J, Moziak K, Yazejian RM, Welch RL, Bell WR, Sengupta S, Aulakh S, Eberhart CG, Loeb DM, Eskandar E, Zheng D, Zang X, and Martin AM
- Abstract
Background: Relapsed medulloblastoma (MB) poses a significant therapeutic challenge due to its highly immunosuppressive tumor microenvironment. Immune checkpoint inhibitors (ICIs) have struggled to mitigate this challenge, largely due to low T-cell infiltration and minimal PD-L1 expression. Identifying the mechanisms driving low T-cell infiltration is crucial for developing more effective immunotherapies., Methods: We utilize a syngeneic mouse model to investigate the tumor immune microenvironment of MB and compare our findings to transcriptomic and proteomic data from human MB., Results: Flow cytometry reveals a notable presence of CD45
hi /CD11bhi macrophage-like and CD45int /CD11bint microglia-like tumor-associated macrophages (TAMs), alongside regulatory T-cells (Tregs ), expressing high levels of the inhibitory checkpoint molecule VISTA. Compared to sham control mice, the CD45hi /CD11bhi compartment significantly expands in tumor-bearing mice and exhibits a myeloid-specific signature composed of VISTA, CD80, PD-L1, CTLA-4, MHCII, CD40, and CD68. These findings are corroborated by proteomic and transcriptomic analyses of human MB samples. Immunohistochemistry highlights an abundance of VISTA-expressing myeloid cells clustering at the tumor-cerebellar border, while T-cells are scarce and express FOXP3. Additionally, tumor cells exhibit immunosuppressive properties, inhibiting CD4 T-cell proliferation in vitro. Identification of VISTA's binding partner, VSIG8, on tumor cells, and its correlation with increased VISTA expression in human transcriptomic analyses suggests a potential therapeutic target., Conclusions: This study underscores the multifaceted mechanisms of immune evasion in MB and highlights the therapeutic potential of targeting the VISTA-VSIG axis to enhance anti-tumor responses.- Published
- 2024
- Full Text
- View/download PDF
35. An unusual finding of an anaplastic meningioma in NF2-related schwannomatosis.
- Author
-
Adelhoefer SJ, Feghali J, Rajan S, Eberhart CG, Staedtke V, and Cohen AR
- Subjects
- Humans, Male, Child, Neurofibromatosis 2 complications, Neurofibromatosis 2 surgery, Neurofibromatosis 2 diagnostic imaging, Neurilemmoma surgery, Neurilemmoma complications, Neurilemmoma diagnostic imaging, Neurilemmoma pathology, Skin Neoplasms surgery, Skin Neoplasms pathology, Skin Neoplasms complications, Magnetic Resonance Imaging, Meningioma surgery, Meningioma complications, Meningioma diagnostic imaging, Meningioma pathology, Meningeal Neoplasms surgery, Meningeal Neoplasms complications, Meningeal Neoplasms diagnostic imaging, Meningeal Neoplasms pathology, Neurofibromatoses complications, Neurofibromatoses surgery, Neurofibromatoses diagnostic imaging
- Abstract
NF2-related schwannomatosis (NF2) is a rare autosomal-dominant genetic disorder characterized by bilateral vestibular schwannomas and multiple meningiomas. This case report presents the extremely rare occurrence of an anaplastic meningioma in a 12-year-old male with previously undiagnosed NF2. The patient presented with a history of abdominal pain and episodic emesis, gait unsteadiness, right upper and lower extremity weakness, and facial weakness. He had sensorineural hearing loss and wore bilateral hearing aids. MR imaging revealed a sizable left frontoparietal, dural-based meningioma with heterogeneous enhancement with mass effect on the brain and midline shift. Multiple additional CNS lesions were noted including a homogenous lesion at the level of T5 indicative of compression of the spinal cord. The patient underwent a frontotemporoparietal craniotomy for the removal of his large dural-based meningioma, utilizing neuronavigation and transdural ultrasonography for precise en bloc resection of the mass. Histopathology revealed an anaplastic meningioma, WHO grade 3, characterized by brisk mitotic activity, small-cell changes, high Ki-67 proliferation rate, and significant loss of P16. We report an anaplastic meningioma associated with an underlying diagnosis of NF2 for which we describe clinical and histopathological features., (© 2024. The Author(s), under exclusive licence to Springer-Verlag GmbH Germany, part of Springer Nature.)
- Published
- 2024
- Full Text
- View/download PDF
36. Postradiation platinum-etoposide in adult medulloblastomas: retrospective analysis of hematological toxicity.
- Author
-
Komlodi-Pasztor E, Munjapara V, Eberhart CG, Bazer DA, Sherief MR, Kamson DO, Ye X, Ozer BH, and Holdhoff M
- Subjects
- Humans, Male, Retrospective Studies, Adult, Female, Young Adult, Middle Aged, Hematologic Diseases chemically induced, Hematologic Diseases etiology, Antineoplastic Combined Chemotherapy Protocols adverse effects, Follow-Up Studies, Adolescent, Craniospinal Irradiation adverse effects, Platinum therapeutic use, Medulloblastoma radiotherapy, Medulloblastoma drug therapy, Etoposide adverse effects, Etoposide administration & dosage, Cerebellar Neoplasms radiotherapy, Cerebellar Neoplasms drug therapy, Cerebellar Neoplasms therapy
- Abstract
Aim: Adult medulloblastomas (MB) are rare, and optimal post-craniospinal irradiation (CSI) chemotherapy is not yet defined. We investigated hematological toxicity in patients treated with platinum-etoposide (EP) post-CSI. Methods: Retrospective, single-institution study to determine hematological toxicity in adult MB patients treated with EP (1995-2022). Results: Thirteen patients with a median follow-up of 50 months (range, 10-233) were analyzed. Four discontinued treatment due to toxicity, one after 1, 3 after 3 cycles. Hematological toxicities included grade 3 (5 patients) and grade 4 (6 patients). Two patients experienced post-treatment progression and died 16 and 37 months from diagnosis. Conclusion: Post-CSI EP demonstrates acceptable hematological toxicity in adult MB. However, the small cohort precludes definitive survival outcome conclusions. Prospective studies for comprehensive comparisons with other regimens are needed in this context.
- Published
- 2024
- Full Text
- View/download PDF
37. A therapeutically targetable positive feedback loop between lnc-HLX-2-7, HLX, and MYC that promotes group 3 medulloblastoma.
- Author
-
Katsushima K, Joshi K, Yuan M, Romero B, Batish M, Stapleton S, Jallo G, Kolanthai E, Seal S, Saulnier O, Taylor MD, Wechsler-Reya RJ, Eberhart CG, and Perera RJ
- Subjects
- Humans, Mice, Animals, Feedback, Oncogenes, Cell Proliferation genetics, Gene Expression Regulation, Neoplastic, Cell Line, Tumor, Transcription Factors metabolism, Homeodomain Proteins genetics, Homeodomain Proteins metabolism, Medulloblastoma genetics, Medulloblastoma pathology, Cerebellar Neoplasms drug therapy, Cerebellar Neoplasms genetics, RNA, Long Noncoding genetics, RNA, Long Noncoding metabolism
- Abstract
Recent studies suggest that long non-coding RNAs (lncRNAs) contribute to medulloblastoma (MB) formation and progression. We have identified an lncRNA, lnc-HLX-2-7, as a potential therapeutic target in group 3 (G3) MBs. lnc-HLX-2-7 RNA specifically accumulates in the promoter region of HLX, a sense-overlapping gene of lnc-HLX-2-7, which activates HLX expression by recruiting multiple factors, including enhancer elements. RNA sequencing and chromatin immunoprecipitation reveal that HLX binds to and activates the promoters of several oncogenes, including TBX2, LIN9, HOXM1, and MYC. Intravenous treatment with cerium-oxide-nanoparticle-coated antisense oligonucleotides targeting lnc-HLX-2-7 (CNP-lnc-HLX-2-7) inhibits tumor growth by 40%-50% in an intracranial MB xenograft mouse model. Combining CNP-lnc-HLX-2-7 with standard-of-care cisplatin further inhibits tumor growth and significantly prolongs mouse survival compared with CNP-lnc-HLX-2-7 monotherapy. Thus, the lnc-HLX-2-7-HLX-MYC axis is important for regulating G3 MB progression, providing a strong rationale for using lnc-HLX-2-7 as a therapeutic target for G3 MBs., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2024 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
38. Collagen type XII is undetectable in keratoconus Bowman's layer.
- Author
-
Rigi M, Son HS, Moon L, Matthaei M, Srikumaran D, Jun AS, Eberhart CG, and Soiberman US
- Subjects
- Humans, Collagen Type XII genetics, Collagen Type XII metabolism, HEK293 Cells, Cornea pathology, Keratoconus metabolism, Epithelium, Corneal pathology
- Abstract
Purpose: Corneal biomechanical failure is the hallmark of keratoconus (KC); however, the cause of this failure remains elusive. Collagen type XII ( COL12A1 ), which localises to Bowman's layer (BL), is thought to function in stress-bearing areas, such as BL. Given the putative protective role of COL12A1 in biomechanical stability, this study aims to characterise COL12A1 expression in all corneal layers involved in KC., Methods: TaqMan quantitative PCR was performed on 31 corneal epithelium samples of progressive KC and myopic control eyes. Tissue microarrays were constructed using full-thickness corneas from 61 KC cases during keratoplasty and 18 non-KC autopsy eyes and stained with an antibody specific to COL12A1. Additionally, COL12A1 was knocked out in vitro in immortalised HEK293 cells., Results: COL12A1 expression was reduced at transcript levels in KC epithelium compared with controls (ratio: 0.58, p<0.03). Immunohistochemical studies demonstrated that COL12A1 protein expression in BL was undetectable, with reduced expression in KC epithelium, basement membrane and stroma., Conclusions: The apparent absence of COL12A1 in KC BL, together with the functional importance that COL12A1 is thought to have in stress bearing areas, suggests that COL12A1 may play a role in the pathogenesis of KC. Further studies are necessary to investigate the mechanisms that lead to COL12A1 dysregulation in KC., Competing Interests: Competing interests: None declared., (© Author(s) (or their employer(s)) 2024. No commercial re-use. See rights and permissions. Published by BMJ.)
- Published
- 2024
- Full Text
- View/download PDF
39. DNA methylation landscapes in DIPG reveal methylome variability that can be modified pharmacologically.
- Author
-
Tetens AR, Martin AM, Arnold A, Novak OV, Idrizi A, Tryggvadottir R, Craig-Schwartz J, Liapodimitri A, Lunsford K, Barbato MI, Eberhart CG, Resnick AC, Raabe EH, and Koldobskiy MA
- Abstract
Background: Diffuse intrinsic pontine glioma (DIPG) is a uniformly lethal brainstem tumor of childhood, driven by histone H3 K27M mutation and resultant epigenetic dysregulation. Epigenomic analyses of DIPG have shown global loss of repressive chromatin marks accompanied by DNA hypomethylation. However, studies providing a static view of the epigenome do not adequately capture the regulatory underpinnings of DIPG cellular heterogeneity and plasticity., Methods: To address this, we performed whole-genome bisulfite sequencing on a large panel of primary DIPG specimens and applied a novel framework for analysis of DNA methylation variability, permitting the derivation of comprehensive genome-wide DNA methylation potential energy landscapes that capture intrinsic epigenetic variation., Results: We show that DIPG has a markedly disordered epigenome with increasingly stochastic DNA methylation at genes regulating pluripotency and developmental identity, potentially enabling cells to sample diverse transcriptional programs and differentiation states. The DIPG epigenetic landscape was responsive to treatment with the hypomethylating agent decitabine, which produced genome-wide demethylation and reduced the stochasticity of DNA methylation at active enhancers and bivalent promoters. Decitabine treatment elicited changes in gene expression, including upregulation of immune signaling such as the interferon response, STING, and MHC class I expression, and sensitized cells to the effects of histone deacetylase inhibition., Conclusions: This study provides a resource for understanding the epigenetic instability that underlies DIPG heterogeneity. It suggests the application of epigenetic therapies to constrain the range of epigenetic states available to DIPG cells, as well as the use of decitabine in priming for immune-based therapies., Competing Interests: The authors declare that they have no competing interests., (© The Author(s) 2024. Published by Oxford University Press, the Society for Neuro-Oncology and the European Association of Neuro-Oncology.)
- Published
- 2024
- Full Text
- View/download PDF
40. Frontal lobe mass in a 46-year-old woman.
- Author
-
Zhang KY, Ramlal B, Schreck KC, Eberhart CG, and Lucas CG
- Subjects
- Female, Humans, Middle Aged, Mutation, Glioma, Brain Neoplasms diagnostic imaging
- Published
- 2024
- Full Text
- View/download PDF
41. miRNA-211 maintains metabolic homeostasis in medulloblastoma through its target gene long-chain acyl-CoA synthetase 4.
- Author
-
Yuan M, Mahmud I, Katsushima K, Joshi K, Saulnier O, Pokhrel R, Lee B, Liyanage W, Kunhiraman H, Stapleton S, Gonzalez-Gomez I, Kannan RM, Eisemann T, Kolanthai E, Seal S, Garrett TJ, Abbasi S, Bockley K, Hanes J, Chapagain P, Jallo G, Wechsler-Reya RJ, Taylor MD, Eberhart CG, Ray A, and Perera RJ
- Subjects
- Animals, Humans, Mice, Cell Line, Tumor, Cell Proliferation, Gene Expression Regulation, Neoplastic, Homeostasis, Ligases genetics, Ligases metabolism, Quality of Life, Cerebellar Neoplasms metabolism, Medulloblastoma metabolism, MicroRNAs genetics, MicroRNAs metabolism
- Abstract
The prognosis of childhood medulloblastoma (MB) is often poor, and it usually requires aggressive therapy that adversely affects quality of life. microRNA-211 (miR-211) was previously identified as an important regulator of cells that descend from neural cells. Since medulloblastomas primarily affect cells with similar ontogeny, we investigated the role and mechanism of miR-211 in MB. Here we showed that miR-211 expression was highly downregulated in cell lines, PDXs, and clinical samples of different MB subgroups (SHH, Group 3, and Group 4) compared to normal cerebellum. miR-211 gene was ectopically expressed in transgenic cells from MB subgroups, and they were subjected to molecular and phenotypic investigations. Monoclonal cells stably expressing miR-211 were injected into the mouse cerebellum. miR-211 forced expression acts as a tumor suppressor in MB both in vitro and in vivo, attenuating growth, promoting apoptosis, and inhibiting invasion. In support of emerging regulatory roles of metabolism in various forms of cancer, we identified the acyl-CoA synthetase long-chain family member (ACSL4) as a direct miR-211 target. Furthermore, lipid nanoparticle-coated, dendrimer-coated, and cerium oxide-coated miR-211 nanoparticles were applied to deliver synthetic miR-211 into MB cell lines and cellular responses were assayed. Synthesizing nanoparticle-miR-211 conjugates can suppress MB cell viability and invasion in vitro. Our findings reveal miR-211 as a tumor suppressor and a potential therapeutic agent in MB. This proof-of-concept paves the way for further pre-clinical and clinical development., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
42. Pathologic vs. protective roles of hypoxia-inducible factor 1 in RPE and photoreceptors in wet vs. dry age-related macular degeneration.
- Author
-
Babapoor-Farrokhran S, Qin Y, Flores-Bellver M, Niu Y, Bhutto IA, Aparicio-Domingo S, Guo C, Rodrigues M, Domashevich T, Deshpande M, Megarity H, Chopde R, Eberhart CG, Canto-Soler V, Montaner S, and Sodhi A
- Subjects
- Mice, Animals, Humans, Aged, Retinal Pigment Epithelium metabolism, Hypoxia-Inducible Factor 1 metabolism, Angiogenesis Inhibitors, Vascular Endothelial Growth Factor A metabolism, Visual Acuity, Oxidants metabolism, Hypoxia metabolism, Geographic Atrophy, Wet Macular Degeneration metabolism, Choroidal Neovascularization genetics, Choroidal Neovascularization prevention & control, Choroidal Neovascularization metabolism
- Abstract
It has previously been reported that antioxidant vitamins can help reduce the risk of vision loss associated with progression to advanced age-related macular degeneration (AMD), a leading cause of visual impairment among the elderly. Nonetheless, how oxidative stress contributes to the development of choroidal neovascularization (CNV) in some AMD patients and geographic atrophy (GA) in others is poorly understood. Here, we provide evidence demonstrating that oxidative stress cooperates with hypoxia to synergistically stimulate the accumulation of hypoxia-inducible factor (HIF)-1α in the retinal pigment epithelium (RPE), resulting in increased expression of the HIF-1-dependent angiogenic mediators that promote CNV. HIF-1 inhibition blocked the expression of these angiogenic mediators and prevented CNV development in an animal model of ocular oxidative stress, demonstrating the pathological role of HIF-1 in response to oxidative stress stimulation in neovascular AMD. While human-induced pluripotent stem cell (hiPSC)-derived RPE monolayers exposed to chemical oxidants resulted in disorganization and disruption of their normal architecture, RPE cells proved remarkably resistant to oxidative stress. Conversely, equivalent doses of chemical oxidants resulted in apoptosis of hiPSC-derived retinal photoreceptors. Pharmacologic inhibition of HIF-1 in the mouse retina enhanced-while HIF-1 augmentation reduced-photoreceptor apoptosis in two mouse models for oxidative stress, consistent with a protective role for HIF-1 in photoreceptors in patients with advanced dry AMD. Collectively, these results suggest that in patients with AMD, increased expression of HIF-1α in RPE exposed to oxidative stress promotes the development of CNV, but inadequate HIF-1α expression in photoreceptors contributes to the development of GA., Competing Interests: Competing interests statement: A.S. is the cofounder of, holds equity in, is a member of the Board of Directors, and serves as the Chief Executive Officer of HIF Therapeutics, Inc. This arrangement has been reviewed and approved by the Johns Hopkins University in accordance with its conflict of interest policies.
- Published
- 2023
- Full Text
- View/download PDF
43. Transition to Chronic Fibrosis in an Animal Model of Retinal Detachment With Features of Proliferative Vitreoretinopathy.
- Author
-
Peterson C, Lu Y, Santiago CP, Price AC, McNally MM, Schubert W, Nassar K, Zollner T, Blackshaw S, Eberhart CG, and Singh MS
- Subjects
- Adult, Animals, Humans, Rabbits, Vascular Endothelial Growth Factor A, Models, Animal, Fibrosis, Fibrillar Collagens, Vitreoretinopathy, Proliferative etiology, Retinal Detachment etiology
- Abstract
Purpose: Proliferative vitreoretinopathy (PVR) is the most common cause of failure of surgically repaired rhegmatogenous retinal detachment (RRD). Chemically induced and cell injection PVR models do not fully simulate the clinical characteristics of PVR in the post-RRD context. There is an unmet need for translational models in which to study mechanisms and treatments specific to RRD-PVR., Methods: RRD was induced in adult Dutch Belted rabbits. Posterior segments were fixed or processed for RNA sequencing at 6 hours and 2, 7, 14, and 35 days after induction. Histochemical staining and immunolabeling for glial fibrillary acidic protein, alpha smooth muscle actin, vascular endothelial growth factor receptor 2, CD68, and RPE 65 kDa protein were performed, and labeling intensity was scored. Single cell RNA sequencing was performed., Results: Acute histopathological changes included intravitreal and intraretinal hemorrhage, leukocytic vitritis, chorioretinitis, and retinal rarefaction. Chronic lesions showed retinal atrophy, gliosis, fibrotic subretinal membranes, and epiretinal fibrovascular proliferation. Fibrillar collagen was present in the fibrocellular and fibrovascular membranes in chronic lesions. Moderate to strong labeling of glia and vasculature was detected in chronic lesions. At day 14, most cells profiled by single cell sequencing were identified as Mϋller glia and microglia, consistent with immunolabeling. Expression of several fibrillar collagen genes was upregulated in chronic lesions., Conclusions: Histological and transcriptional features of this rabbit model simulate important features of human RRD-PVR, including the transition to chronic intraretinal and periretinal fibrosis. This animal model of RRD with features of PVR will enable further research on targeted treatment interventions.
- Published
- 2023
- Full Text
- View/download PDF
44. Severe Mpox Infection of the Eye and Periocular Region.
- Author
-
Bacorn C, Majidi S, Schultz H, Sulewski ME, Eberhart CG, and Mahoney NR
- Subjects
- Humans, Face, Eyelids, Mpox (monkeypox)
- Abstract
Mpox is an emerging zoonotic infection with potentially severe ocular and periocular consequences, particularly in immunocompromised patients. This report summarizes 2 cases of fulminant mpox presenting in patients with AIDS. In the first case, confluent lesions resulted in orbital compartment syndrome and total eyelid necrosis. In the second case, eyelid involvement was accompanied by corneal melt and perforation. Despite aggressive medical and surgical treatment, both patients developed permanent loss of vision and ultimately expired., Competing Interests: The authors have no financial or conflicts of interest to disclose., (Copyright © 2023 The American Society of Ophthalmic Plastic and Reconstructive Surgery, Inc.)
- Published
- 2023
- Full Text
- View/download PDF
45. Orbital SOX10-mutant schwannoma with plexiform growth: Expanding the histopathological spectrum of a new molecular group.
- Author
-
Wali AA, Yang R, Merbs SL, Rodriguez FJ, Eberhart CG, and Lucas CG
- Subjects
- Humans, SOXE Transcription Factors genetics, Neurilemmoma diagnostic imaging, Neurilemmoma genetics, Neurilemmoma pathology
- Published
- 2023
- Full Text
- View/download PDF
46. The 5th Edition of the World Health Organization Classification of Tumours of the Eye and Orbit.
- Author
-
Milman T, Grossniklaus HE, Goldman-Levy G, Kivelä TT, Coupland SE, White VA, Mudhar HS, Eberhart CG, Verdijk RM, Heegaard S, Gill AJ, Jager MJ, Rodríguez-Reyes AA, Esmaeli B, Hodge JC, and Cree IA
- Abstract
Competing Interests: The authors have no conflicts of interest to declare.
- Published
- 2023
- Full Text
- View/download PDF
47. Bioreducible Gene Delivery Platform that Promotes Intracellular Payload Release and Widespread Brain Dispersion.
- Author
-
Rao D, Kwak G, Wang H, Eberhart CG, Hanes J, and Suk JS
- Subjects
- Genetic Therapy, Brain metabolism, Gene Transfer Techniques, Polymers metabolism
- Abstract
We here introduce a novel bioreducible polymer-based gene delivery platform enabling widespread transgene expression in multiple brain regions with therapeutic relevance following intracranial convection-enhanced delivery. Our bioreducible nanoparticles provide markedly enhanced gene delivery efficacy in vitro and in vivo compared to nonbiodegradable nanoparticles primarily due to the ability to release gene payloads preferentially inside cells. Remarkably, our platform exhibits competitive gene delivery efficacy in a neuron-rich brain region compared to a viral vector under previous and current clinical investigations with demonstrated positive outcomes. Thus, our platform may serve as an attractive alternative for the intracranial gene therapy of neurological disorders.
- Published
- 2023
- Full Text
- View/download PDF
48. Histological Comparative Analysis of Bowman Layer Grafts Procured Using 3 Different Techniques.
- Author
-
Son HS, Moon L, Wang J, Eberhart CG, Jun AS, Srikumaran D, and Soiberman US
- Subjects
- Humans, Descemet Membrane, Tissue Donors, Tissue and Organ Harvesting, Endothelium, Corneal pathology, Descemet Stripping Endothelial Keratoplasty methods
- Abstract
Purpose: The objective of this study is to perform a histological analysis of Bowman layer (BL) grafts., Methods: BL grafts were procured from 13 human cadaver corneal tissues using 3 different donor preparation techniques. Subsequently, the grafts were fixed in 10% buffered formalin phosphate and embedded in paraffin. Hematoxylin and eosin sections of BL grafts were obtained and analyzed under a light microscope. BL and full graft thickness were measured using an image-processing software., Results: All 13 BL grafts contained residual anterior stromal tissue. BL stripping using Kelman-McPherson and Moorfield forceps (technique 3) achieved the thinnest graft thickness with a mean full graft thickness of 18.7 μm (95% confidence interval [CI], -9.8 to 47.2) at the thinnest point of the graft, whereas BL procurement using the Melles lamellar dissector (technique 2) led to the highest mean full graft thickness of 279.9 μm (95% CI, 251.4-308.5) even at the thinnest area of the graft. By contrast, BL dissection using a blunt dissector (technique 1) provided a mean full graft thickness of 70.2 μm (95% CI, 40.4-100.1) at the graft's thinnest point. Although peripheral graft tears occurred in 50%, 50%, and 100% of techniques 1, 2, and 3, respectively, intact 6.25-mm diameter BL grafts could be secured in 50%, 100%, and 80% of techniques 1, 2, and 3, respectively., Conclusions: None of the techniques used led to the procurement of pure BL grafts devoid of the anterior stroma. Peripheral scoring with a thin needle and tissue manipulation with Kelman-McPherson and Moorfield forceps led to the thinnest grafts in this study., Competing Interests: The authors have no funding or conflicts of interest to disclose., (Copyright © 2023 Wolters Kluwer Health, Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
49. Applications of chemical exchange saturation transfer magnetic resonance imaging in identifying genetic markers in gliomas.
- Author
-
Jiang S, Wen Z, Ahn SS, Cai K, Paech D, Eberhart CG, and Zhou J
- Subjects
- Humans, Genetic Markers, Magnetic Resonance Imaging methods, Protons, Isocitrate Dehydrogenase genetics, Glioma diagnostic imaging, Glioma genetics, Glioma chemistry, Brain Neoplasms diagnostic imaging, Brain Neoplasms genetics, Brain Neoplasms chemistry
- Abstract
Chemical exchange saturation transfer (CEST) imaging is an important molecular magnetic resonance imaging technique that can image numerous low-concentration biomolecules with water-exchangeable protons (such as cellular proteins) and tissue pH. CEST, or more specially amide proton transfer-weighted imaging, has been widely used for the detection, diagnosis, and response assessment of brain tumors, and its feasibility in identifying molecular markers in gliomas has also been explored in recent years. In this paper, after briefing on the basic principles and quantification methods of CEST imaging, we review its early applications in identifying isocitrate dehydrogenase mutation status, MGMT methylation status, 1p/19q deletion status, and H3K27M mutation status in gliomas. Finally, we discuss the limitations or weaknesses in these studies., (© 2022 The Authors. NMR in Biomedicine published by John Wiley & Sons Ltd.)
- Published
- 2023
- Full Text
- View/download PDF
50. Epigenetic upregulation of Schlafen11 renders WNT- and SHH-activated medulloblastomas sensitive to cisplatin.
- Author
-
Nakata S, Murai J, Okada M, Takahashi H, Findlay TH, Malebranche K, Parthasarathy A, Miyashita S, Gabdulkhaev R, Benkimoun I, Druillennec S, Chabi S, Hawkins E, Miyahara H, Tateishi K, Yamashita S, Yamada S, Saito T, On J, Watanabe J, Tsukamoto Y, Yoshimura J, Oishi M, Nakano T, Imamura M, Imai C, Yamamoto T, Takeshima H, Sasaki AT, Rodriguez FJ, Nobusawa S, Varlet P, Pouponnot C, Osuka S, Pommier Y, Kakita A, Fujii Y, Raabe EH, Eberhart CG, and Natsumeda M
- Subjects
- Humans, Cisplatin pharmacology, Up-Regulation, Irinotecan, Epigenesis, Genetic, Hedgehog Proteins genetics, Hedgehog Proteins metabolism, Nuclear Proteins metabolism, Medulloblastoma drug therapy, Medulloblastoma genetics, Cerebellar Neoplasms drug therapy, Cerebellar Neoplasms genetics
- Abstract
Background: Intensive chemotherapeutic regimens with craniospinal irradiation have greatly improved survival in medulloblastoma patients. However, survival markedly differs among molecular subgroups and their biomarkers are unknown. Through unbiased screening, we found Schlafen family member 11 (SLFN11), which is known to improve response to DNA damaging agents in various cancers, to be one of the top prognostic markers in medulloblastomas. Hence, we explored the expression and functions of SLFN11 in medulloblastoma., Methods: SLFN11 expression for each subgroup was assessed by immunohistochemistry in 98 medulloblastoma patient samples and by analyzing transcriptomic databases. We genetically or epigenetically modulated SLFN11 expression in medulloblastoma cell lines and determined cytotoxic response to the DNA damaging agents cisplatin and topoisomerase I inhibitor SN-38 in vitro and in vivo., Results: High SLFN11 expressing cases exhibited significantly longer survival than low expressing cases. SLFN11 was highly expressed in the WNT-activated subgroup and in a proportion of the SHH-activated subgroup. While WNT activation was not a direct cause of the high expression of SLFN11, a specific hypomethylation locus on the SLFN11 promoter was significantly correlated with high SLFN11 expression. Overexpression or deletion of SLFN11 made medulloblastoma cells sensitive and resistant to cisplatin and SN-38, respectively. Pharmacological upregulation of SLFN11 by the brain-penetrant histone deacetylase-inhibitor RG2833 markedly increased sensitivity to cisplatin and SN-38 in SLFN11-negative medulloblastoma cells. Intracranial xenograft studies also showed marked sensitivity to cisplatin by SLFN11-overexpression in medulloblastoma cells., Conclusions: High SLFN11 expression is one factor which renders favorable outcomes in WNT-activated and a subset of SHH-activated medulloblastoma possibly through enhancing response to cisplatin., (© The Author(s) 2022. Published by Oxford University Press on behalf of the Society for Neuro-Oncology. All rights reserved. For permissions, please e-mail: journals.permissions@oup.com.)
- Published
- 2023
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.