21 results on '"Renate Kirschner"'
Search Results
2. The KRAS-G12D mutation induces metabolic vulnerability in B-cell acute lymphoblastic leukemia
- Author
-
Yan Xu, Houshun Fang, Yao Chen, Yabin Tang, Huiying Sun, Ziqing Kong, Fan Yang, Renate Kirschner-Schwabe, Liang Zhu, Alex Toker, Ning Xiao, Bin-Bing S. Zhou, and Hui Li
- Subjects
Biological sciences ,Biochemistry ,Molecular biology ,Science - Abstract
Summary: Mutations in RAS pathway genes are highly prevalent in acute lymphoblastic leukemia (ALL). However, the effects of RAS mutations on ALL cell growth have not been experimentally characterized, and effective RAS-targeting therapies are being sought after. Here, we found that Reh ALL cells bearing the KRAS-G12D mutation showed increased proliferation rates in vitro but displayed severely compromised growth in mice. Exploring this divergence, proliferation assays with multiple ALL cell lines revealed that the KRAS-G12D rewired methionine and arginine metabolism. Isotope tracing results showed that KRAS-G12D promotes catabolism of methionine and arginine to support anabolism of polyamines and proline, respectively. Chemical inhibition of polyamine biosynthesis selectively killed KRAS-G12D B-ALL cells. Finally, chemically inhibiting AKT/mTOR signaling abrogated the altered amino acid metabolism and strongly promoted the in vivo growth of KRAS-G12D cells in B-ALL xenograft. Our study thus illustrates how hyperactivated AKT/mTOR signaling exerts distinct impacts on hematological malignancies vs. solid tumors.
- Published
- 2022
- Full Text
- View/download PDF
3. Chromatin accessibility landscape of pediatric T‐lymphoblastic leukemia and human T‐cell precursors
- Author
-
Büşra Erarslan‐Uysal, Joachim B Kunz, Tobias Rausch, Paulina Richter‐Pechańska, Ianthe AEM van Belzen, Viktoras Frismantas, Beat Bornhauser, Diana Ordoñez‐Rueada, Malte Paulsen, Vladimir Benes, Martin Stanulla, Martin Schrappe, Gunnar Cario, Gabriele Escherich, Kseniya Bakharevich, Renate Kirschner‐Schwabe, Cornelia Eckert, Tsvetomir Loukanov, Matthias Gorenflo, Sebastian M Waszak, Jean‐Pierre Bourquin, Martina U Muckenthaler, Jan O Korbel, and Andreas E Kulozik
- Subjects
ATAC‐Seq ,chromatin accessibility ,T‐cell development ,T‐cell leukemia ,Medicine (General) ,R5-920 ,Genetics ,QH426-470 - Abstract
Abstract We aimed at identifying the developmental stage at which leukemic cells of pediatric T‐ALLs are arrested and at defining leukemogenic mechanisms based on ATAC‐Seq. Chromatin accessibility maps of seven developmental stages of human healthy T cells revealed progressive chromatin condensation during T‐cell maturation. Developmental stages were distinguished by 2,823 signature chromatin regions with 95% accuracy. Open chromatin surrounding SAE1 was identified to best distinguish thymic developmental stages suggesting a potential role of SUMOylation in T‐cell development. Deconvolution using signature regions revealed that T‐ALLs, including those with mature immunophenotypes, resemble the most immature populations, which was confirmed by TF‐binding motif profiles. We integrated ATAC‐Seq and RNA‐Seq and found DAB1, a gene not related to leukemia previously, to be overexpressed, abnormally spliced and hyper‐accessible in T‐ALLs. DAB1‐negative patients formed a distinct subgroup with particularly immature chromatin profiles and hyper‐accessible binding sites for SPI1 (PU.1), a TF crucial for normal T‐cell maturation. In conclusion, our analyses of chromatin accessibility and TF‐binding motifs showed that pediatric T‐ALL cells are most similar to immature thymic precursors, indicating an early developmental arrest.
- Published
- 2020
- Full Text
- View/download PDF
4. Long non-coding RNAs defining major subtypes of B cell precursor acute lymphoblastic leukemia
- Author
-
Alva Rani James, Michael P. Schroeder, Martin Neumann, Lorenz Bastian, Cornelia Eckert, Nicola Gökbuget, Jutta Ortiz Tanchez, Cornelia Schlee, Konstandina Isaakidis, Stefan Schwartz, Thomas Burmeister, Arend von Stackelberg, Michael A. Rieger, Stefanie Göllner, Martin Horstman, Martin Schrappe, Renate Kirschner-Schwabe, Monika Brüggemann, Carsten Müller-Tidow, Hubert Serve, Altuna Akalin, and Claudia D. Baldus
- Subjects
BCP-ALL subtypes ,DUX4 ,Ph-like ,NH-HeH ,Subtype-specific lncRNAs ,Key signaling pathways ,Diseases of the blood and blood-forming organs ,RC633-647.5 ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Background Long non-coding RNAs (lncRNAs) have emerged as a novel class of RNA due to its diverse mechanism in cancer development and progression. However, the role and expression pattern of lncRNAs in molecular subtypes of B cell acute lymphoblastic leukemia (BCP-ALL) have not yet been investigated. Here, we assess to what extent lncRNA expression and DNA methylation is driving the progression of relapsed BCP-ALL subtypes and we determine if the expression and DNA methylation profile of lncRNAs correlates with established BCP-ALL subtypes. Methods We performed RNA sequencing and DNA methylation (Illumina Infinium microarray) of 40 diagnosis and 42 relapse samples from 45 BCP-ALL patients in a German cohort and quantified lncRNA expression. Unsupervised clustering was applied to ascertain and confirm that the lncRNA-based classification of the BCP-ALL molecular subtypes is present in both our cohort and an independent validation cohort of 47 patients. A differential expression and differential methylation analysis was applied to determine the subtype-specific, relapse-specific, and differentially methylated lncRNAs. Potential functions of subtype-specific lncRNAs were determined by using co-expression-based analysis on nearby (cis) and distally (trans) located protein-coding genes. Results Using an integrative Bioinformatics analysis, we developed a comprehensive catalog of 1235 aberrantly dysregulated BCP-ALL subtype-specific and 942 relapse-specific lncRNAs and the methylation profile of three subtypes of BCP-ALL. The 1235 subtype-specific lncRNA signature represented a similar classification of the molecular subtypes of BCP-ALL in the independent validation cohort. We identified a strong correlation between the DUX4-specific lncRNAs and genes involved in the activation of TGF-β and Hippo signaling pathways. Similarly, Ph-like-specific lncRNAs were correlated with genes involved in the activation of PI3K-AKT, mTOR, and JAK-STAT signaling pathways. Interestingly, the relapse-specific lncRNAs correlated with the activation of metabolic and signaling pathways. Finally, we found 23 promoter methylated lncRNAs epigenetically facilitating their expression levels. Conclusion Here, we describe a set of subtype-specific and relapse-specific lncRNAs from three major BCP-ALL subtypes and define their potential functions and epigenetic regulation. The subtype-specific lncRNAs are reproducible and can effectively stratify BCP-ALL subtypes. Our data uncover the diverse mechanism of action of lncRNAs in BCP-ALL subtypes defining which lncRNAs are involved in the pathogenesis of disease and are relevant for the stratification of BCP-ALL subtypes.
- Published
- 2019
- Full Text
- View/download PDF
5. PDX models recapitulate the genetic and epigenetic landscape of pediatric T‐cell leukemia
- Author
-
Paulina Richter‐Pechańska, Joachim B Kunz, Beat Bornhauser, Caroline von Knebel Doeberitz, Tobias Rausch, Büşra Erarslan‐Uysal, Yassen Assenov, Viktoras Frismantas, Blerim Marovca, Sebastian M Waszak, Martin Zimmermann, Julia Seemann, Margit Happich, Martin Stanulla, Martin Schrappe, Gunnar Cario, Gabriele Escherich, Kseniya Bakharevich, Renate Kirschner‐Schwabe, Cornelia Eckert, Martina U Muckenthaler, Jan O Korbel, Jean‐Pierre Bourquin, and Andreas E Kulozik
- Subjects
ATAC‐Seq ,PDX stability ,T‐ALL ,T‐cell leukemia ,Medicine (General) ,R5-920 ,Genetics ,QH426-470 - Abstract
Abstract We compared 24 primary pediatric T‐cell acute lymphoblastic leukemias (T‐ALL) collected at the time of initial diagnosis and relapse from 12 patients and 24 matched patient‐derived xenografts (PDXs). DNA methylation profile was preserved in PDX mice in 97.5% of the promoters (ρ = 0.99). Similarly, the genome‐wide chromatin accessibility (ATAC‐Seq) was preserved remarkably well (ρ = 0.96). Interestingly, both the ATAC regions, which showed a significant decrease in accessibility in PDXs and the regions hypermethylated in PDXs, were associated with immune response, which might reflect the immune deficiency of the mice and potentially the incomplete interaction between murine cytokines and human receptors. The longitudinal approach of this study allowed an observation that samples collected from patients who developed a type 1 relapse (clonal mutations maintained at relapse) preserved their genomic composition; whereas in patients who developed a type 2 relapse (subset of clonal mutations lost at relapse), the preservation of the leukemia's composition was more variable. In sum, this study underlines the remarkable genomic stability, and for the first time documents the preservation of the epigenomic landscape in T‐ALL‐derived PDX models.
- Published
- 2018
- Full Text
- View/download PDF
6. Intragenic amplification of PAX5: a novel subgroup in B-cell precursor acute lymphoblastic leukemia?
- Author
-
Claire Schwab, Karin Nebral, Lucy Chilton, Cristina Leschi, Esmé Waanders, Judith M. Boer, Markéta Žaliová, Rosemary Sutton, Ingegerd Ivanov Öfverholm, Kentaro Ohki, Yuka Yamashita, Stefanie Groeneveld-Krentz, Eva Froňková, Marleen Bakkus, Joelle Tchinda, Thayana da Conceição Barbosa, Grazia Fazio, Wojciech Mlynarski, Agata Pastorczak, Giovanni Cazzaniga, Maria S. Pombo-de-Oliveira, Jan Trka, Renate Kirschner-Schwabe, Toshihiko Imamura, Gisela Barbany, Martin Stanulla, Andishe Attarbaschi, Renate Panzer-Grümayer, Roland P. Kuiper, Monique L. den Boer, Hélène Cavé, Anthony V. Moorman, Christine J. Harrison, and Sabine Strehl
- Subjects
Specialties of internal medicine ,RC581-951 - Published
- 2017
- Full Text
- View/download PDF
7. NOTCH1 mutation, TP53 alteration and myeloid antigen expression predict outcome heterogeneity in children with first relapse of T-cell acute lymphoblastic leukemia
- Author
-
Jana Hof, Corinne Kox, Stefanie Groeneveld-Krentz, Obul R. Bandapalli, Leonid Karawajew, Katharina Schedel, Joachim B. Kunz, Cornelia Eckert, Wolf-Dieter Ludwig, Richard Ratei, Peter Rhein, Günter Henze, Martina U. Muckenthaler, Andreas E. Kulozik, Arend von Stackelberg, and Renate Kirschner-Schwabe
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Published
- 2017
- Full Text
- View/download PDF
8. Pediatric T-cell lymphoblastic leukemia evolves into relapse by clonal selection, acquisition of mutations and promoter hypomethylation
- Author
-
Joachim B. Kunz, Tobias Rausch, Obul R. Bandapalli, Juliane Eilers, Paulina Pechanska, Stephanie Schuessele, Yassen Assenov, Adrian M. Stütz, Renate Kirschner-Schwabe, Jana Hof, Cornelia Eckert, Arend von Stackelberg, Martin Schrappe, Martin Stanulla, Rolf Koehler, Smadar Avigad, Sarah Elitzur, Rupert Handgretinger, Vladimir Benes, Joachim Weischenfeldt, Jan O. Korbel, Martina U. Muckenthaler, and Andreas E. Kulozik
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Abstract
Relapsed precursor T-cell acute lymphoblastic leukemia is characterized by resistance against chemotherapy and is frequently fatal. We aimed at understanding the molecular mechanisms resulting in relapse of T-cell acute lymphoblastic leukemia and analyzed 13 patients at first diagnosis, remission and relapse by whole exome sequencing, targeted ultra-deep sequencing, multiplex ligation dependent probe amplification and DNA methylation array. Compared to primary T-cell acute lymphoblastic leukemia, in relapse the number of single nucleotide variants and small insertions and deletions approximately doubled from 11.5 to 26. Targeted ultra-deep sequencing sensitively detected subclones that were selected for in relapse. The mutational pattern defined two types of relapses. While both are characterized by selection of subclones and acquisition of novel mutations, ‘type 1’ relapse derives from the primary leukemia whereas ‘type 2’ relapse originates from a common pre-leukemic ancestor. Relapse-specific changes included activation of the nucleotidase NT5C2 resulting in resistance to chemotherapy and mutations of epigenetic modulators, exemplified by SUZ12, WHSC1 and SMARCA4. While mutations present in primary leukemia and in relapse were enriched for known drivers of leukemia, relapse-specific changes revealed an association with general cancer-promoting mechanisms. This study thus identifies mechanisms that drive progression of pediatric T-cell acute lymphoblastic leukemia to relapse and may explain the characteristic treatment resistance of this condition.
- Published
- 2015
- Full Text
- View/download PDF
9. The activating STAT5B N642H mutation is a common abnormality in pediatric T-cell acute lymphoblastic leukemia and confers a higher risk of relapse
- Author
-
Obul R. Bandapalli, Stephanie Schuessele, Joachim B. Kunz, Tobias Rausch, Adrian M. Stütz, Noa Tal, Ifat Geron, Nava Gershman, Shai Izraeli, Juliane Eilers, Nina Vaezipour, Renate Kirschner-Schwabe, Jana Hof, Arend von Stackelberg, Martin Schrappe, Martin Stanulla, Martin Zimmermann, Rolf Koehler, Smadar Avigad, Rupert Handgretinger, Viktoras Frismantas, Jean Pierre Bourquin, Beat Bornhauser, Jan O. Korbel, Martina U. Muckenthaler, and Andreas E. Kulozik
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Published
- 2014
- Full Text
- View/download PDF
10. High VLA-4 expression is associated with adverse outcome and distinct gene expression changes in childhood B-cell precursor acute lymphoblastic leukemia at first relapse
- Author
-
Shabnam Shalapour, Jana Hof, Renate Kirschner-Schwabe, Lorenz Bastian, Cornelia Eckert, Javier Prada, Günter Henze, Arend von Stackelberg, and Karl Seeger
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Abstract
Background Resistance to therapy and subsequent relapse remain major challenges in the clinical management of relapsed childhood acute lymphoblastic leukemia. As the bone marrow environment plays an important role in survival and chemotherapy resistance of leukemia cells by activating different signaling pathways, such as the VLA-4 and PI3K/Akt pathways, we studied the prognostic and biological impact of VLA-4 expression in leukemia cells from children with relapsed B-cell precursor acute lymphoblastic leukemia and its influence on the sensitivity of the leukemia cells to drugs.Design and Methods VLA-4 expression was quantified by real-time polymerase chain reaction in leukemia cells from 56 patients with relapsed acute lymphoblastic leukemia enrolled in the ALL-REZ BFM 2002 trial of the Berlin-Frankfurt-Münster study group. Gene expression changes related to VLA-4 expression were investigated by microarray-based mRNA profiling. The effect of VLA-4 signaling on proliferation and drug resistance was studied in co-cultures of leukemia and stromal cells.Results High expression of VLA-4 at first relapse was associated with adverse prognostic factors, poor molecular response to therapy and significantly worse probabilities of event-free and overall survival. VLA-4 expression was an independent prognostic parameter. Comparing gene expression profiles of leukemia cells with high versus low VLA-4 expression, we identified 27 differentially expressed genes primarily involved in the PI3K/Akt, ephrin and Rho GTPase pathways. Blocking of VLA-4 signaling in combination with cytarabine treatment abolished the growth supportive effect of stromal cells.Conclusions Our results show that high VLA-4 expression is a marker of poor prognosis and a potential therapeutic target in children with relapsed acute lymphoblastic leukemia and confirm that cellular interactions and biological effects related to VLA-4 play a decisive role in the survival of leukemia cells and response to therapy.
- Published
- 2011
- Full Text
- View/download PDF
11. Risk factors and outcomes in children with high-risk B-cell precursor and T-cell relapsed acute lymphoblastic leukaemia: combined analysis of ALLR3 and ALL-REZ BFM 2002 clinical trials
- Author
-
Lucie Sramkova, Vaskar Saha, Christiane Chen-Santel, Anthony V. Moorman, Andishe Attarbaschi, Renate Kirschner-Schwabe, Stefanie Groeneveld-Krentz, Shekhar Krishnan, Arend von Stackelberg, Peter M. Hoogerbrugge, Martin Zimmermann, Guenter Henze, Catriona Parker, Jean-Pierre Bourquin, Tamas Revesz, Rosemary Sutton, Jeremy Hancock, Cornelia Eckert, Julie Ae Irving, University of Zurich, Eckert, Cornelia, and Saha, Vaskar
- Subjects
Male ,0301 basic medicine ,Oncology ,Cancer Research ,Neoplasm, Residual ,Time Factors ,medicine.medical_treatment ,High-risk ,Gene Dosage ,Graft vs Host Disease ,Precursor T-Cell Lymphoblastic Leukemia-Lymphoma ,Targeted therapy ,0302 clinical medicine ,Recurrence ,Risk Factors ,Antineoplastic Combined Chemotherapy Protocols ,1306 Cancer Research ,Child ,Clinical Trials as Topic ,Manchester Cancer Research Centre ,Acute lymphoblastic leukaemia ,Hematopoietic Stem Cell Transplantation ,Stem cell transplantation ,Progression-Free Survival ,medicine.anatomical_structure ,Child, Preschool ,030220 oncology & carcinogenesis ,Disease Progression ,2730 Oncology ,Female ,medicine.medical_specialty ,Adolescent ,T cell ,Karyotype ,610 Medicine & health ,Outcomes ,Risk Assessment ,Disease-Free Survival ,03 medical and health sciences ,Precursor B-Cell Lymphoblastic Leukemia-Lymphoma ,Internal medicine ,Biomarkers, Tumor ,medicine ,Humans ,B cell ,Proportional hazards model ,business.industry ,Minimal residual disease ,ResearchInstitutes_Networks_Beacons/mcrc ,Transplantation ,Clinical trial ,030104 developmental biology ,10036 Medical Clinic ,Mutation ,Lymphoblastic leukaemia ,business - Abstract
Aim: Outcomes of children with high-risk (HR) relapsed acute lymphoblastic leukaemia (ALL) (N = 393), recruited to ALLR3 and ALL-REZ BFM 2002 trials, were analysed. Minimal residual disease (MRD) was assessed after induction and at predetermined time points until haematopoietic stem cell transplantation (SCT). Methods: Genetic analyses included karyotype, copy-number alterations and mutation analyses. Ten-year survivals were analysed using Kaplan-Meier and Cox models for multivariable analyses. Results: Outcomes of patients were comparable in ALLR3 and ALL-REZ BFM 2002. The event-free survival of B-cell precursor (BCP) and T-cell ALL (T-ALL) was 22.6% and 26.2% (P = 0.94), respectively, and the overall survival (OS) was 32.6% and 28.2% (P = 0.11), respectively. Induction failures (38%) were associated with deletions of NR3C1 (P = 0.002) and BTG1 (P = 0.03) in BCP-ALL. The disease-free survival (DFS) and OS in patients with good vs poor MRD responses were 57.4% vs 22.6% (P < 0.0001) and 57.8% vs 32.0% (P = 0.0004), respectively. For BCP- and T-ALL, the post-SCT DFS and OS were 42.1% and 56.8% (P = 0.26) and 51.6% and 55.4% (P = 0.67), respectively. The cumulative incidences of post-SCT relapse for BCP- and T-ALL were 36.9% and 17.8% (P = 0.012) and of death were 10.7% and 25.5% (P = 0.013), respectively. Determinants of outcomes after SCT were acute graft versus host disease, pre-SCT MRD (≥10 −3), HR cytogenetics and TP53 alterations in BCP-ALL. Conclusion: Improvements in outcomes for HR ALL relapses require novel compounds in induction therapy to improve remission rates and immune targeted therapy after induction to maintain remission after SCT. Trial registration: ALLR3: NCT00967057; ALL REZ-BFM 2002: NCT00114348
- Published
- 2021
12. Long non-coding RNAs defining major subtypes of B cell precursor acute lymphoblastic leukemia
- Author
-
Hubert Serve, Renate Kirschner-Schwabe, Alva Rani James, Cornelia Eckert, Carsten Müller-Tidow, Jutta Ortiz Tanchez, Claudia D. Baldus, Martin Schrappe, Stefanie Göllner, Konstandina Isaakidis, Arend von Stackelberg, Michael P Schroeder, Michael A. Rieger, Stefan Schwartz, Monika Brüggemann, Cornelia Schlee, Lorenz Bastian, Nicola Gökbuget, Martin Horstman, Altuna Akalin, Thomas Burmeister, and Martin Neumann
- Subjects
Male ,0301 basic medicine ,Cancer Research ,Microarray ,DUX4 ,Epigenesis, Genetic ,Transcriptome ,Cohort Studies ,0302 clinical medicine ,Bone Marrow ,Recurrence ,BCP-ALL subtypes ,Child ,Promoter Regions, Genetic ,Epigenetically altered lncRNAs ,Subtype-specific lncRNAs ,Hematology ,Methylation ,Ph-like ,lcsh:Diseases of the blood and blood-forming organs ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Berlin ,medicine.anatomical_structure ,Oncology ,Hippo signaling ,030220 oncology & carcinogenesis ,DNA methylation ,Female ,RNA, Long Noncoding ,Key signaling pathways ,Signal transduction ,Technology Platforms ,Metabolic Networks and Pathways ,Adult ,Computational biology ,Biology ,lcsh:RC254-282 ,03 medical and health sciences ,Precursor B-Cell Lymphoblastic Leukemia-Lymphoma ,medicine ,Biomarkers, Tumor ,Humans ,Epigenetics ,Molecular Biology ,Gene ,PI3K/AKT/mTOR pathway ,B cell ,Integrative bioinformatics ,Base Sequence ,lcsh:RC633-647.5 ,Research ,RNA ,DNA Methylation ,Relapse-specific lncRNAs ,030104 developmental biology ,Cardiovascular and Metabolic Diseases ,NH-HeH - Abstract
Recent studies implicated that long non-coding RNAs (lncRNAs) may play a role in the progression and development of acute lymphoblastic leukemia, however, this role is not yet clear. In order to unravel the role of lncRNAs associated with B-cell precursor Acute Lymphoblastic Leukemia (BCP-ALL) subtypes, we performed transcriptome sequencing and DNA methylation array across 82 BCP-ALL samples from three molecular subtypes (DUX4, Ph-like, and Near Haploid or High Hyperdiploidy). Unsupervised clustering of BCP-ALL samples on the basis of their lncRNAs on transcriptome and DNA methylation profiles revealed robust clusters separating three molecular subtypes. Using extensive computational analysis, we developed a comprehensive catalog of 1235 aberrantly dysregulated BCP-ALL subtype-specific lncRNAs with altered expression and methylation patterns from three subtypes of BCP-ALL. By analyzing the co-expression of subtype-specific lncRNAs and protein-coding genes, we inferred key molecular processes in BCP-ALL subtypes. A strong correlation was identified between the DUX4 specific lncRNAs and activation of TGF-β and Hippo signaling pathways. Similarly, Ph-like specific lncRNAs were correlated with genes involved in activation of PI3K-AKT, mTOR, and JAK-STAT signaling pathways. Interestingly, the relapse-specific differentially expressed lncRNAs correlated with the activation of metabolic and signaling pathways. Finally, we showed a set of epigenetically altered lncRNAs facilitating the expression of tumor genes located at theircislocation. Overall, our study provides a comprehensive set of novel subtype and relapse-specific lncRNAs in BCP-ALL. Our findings suggest a wide range of molecular pathways are associated with lncRNAs in BCP-ALL subtypes and provide a foundation for functional investigations that could lead to new therapeutic approaches.Author SummaryAcute lymphoblastic leukemia is a heterogeneous blood cancer, with multiple molecular subtypes, and with high relapse rate. We are far from the complete understanding of the rationale behind these subtypes and high relapse rate. Long non-coding (lncRNAs) has emerged as a novel class of RNA due to its diverse mechanism in cancer development and progression. LncRNAs does not code for proteins and represent around 70% of human transcripts. Recently, there are a number of studies used lncRNAs expression profile in the classification of various cancers subtypes and displayed their correlation with genomic, epigenetic, pathological and clinical features in diverse cancers. Therefore, lncRNAs can account for heterogeneity and has independent prognostic value in various cancer subtypes. However, lncRNAs defining the molecular subtypes of BCP-ALL are not portrayed yet. Here, we describe a set of relapse and subtype-specific lncRNAs from three major BCP-ALL subtypes and define their potential functions and epigenetic regulation. Our data uncover the diverse mechanism of action of lncRNAs in BCP-ALL subtypes defining how lncRNAs are involved in the pathogenesis of disease and the relevance in the stratification of BCP-ALL subtypes.
- Published
- 2019
13. Clinical and genetic characteristics of children with acute lymphoblastic leukemia and Li–Fraumeni syndrome
- Author
-
Stefanie Groeneveld-Krentz, Renate Kirschner-Schwabe, Nienke van Engelen, Marjolijn C.J. Jongmans, Karin Wadt, Simon Bailey, Arend von Stackelberg, Martin Stanulla, Gabriele Escherich, Christian P. Kratz, Cornelia Eckert, Anja Möricke, Tim Ripperger, Lisa Richter, Doris Steinemann, Greta Winter, Olli Lohi, Adela Escudero, and Roula Farah
- Subjects
Male ,Oncology ,Cancer Research ,medicine.medical_specialty ,Letter ,Adolescent ,Lymphoblastic Leukemia ,Medizin ,Li-Fraumeni Syndrome ,Internal medicine ,Genetics research ,Humans ,Medicine ,Genetic Predisposition to Disease ,Child ,Risk factors ,Acute lymphocytic leukaemia ,Germ-Line Mutation ,business.industry ,Hematology ,Precursor Cell Lymphoblastic Leukemia-Lymphoma ,medicine.disease ,Li–Fraumeni syndrome ,Child, Preschool ,Female ,business - Published
- 2021
14. Chromatin accessibility landscape of pediatric T‐lymphoblastic leukemia and human T‐cell precursors
- Author
-
Gabriele Escherich, Beat Bornhauser, Martin Stanulla, Kseniya Bakharevich, Tsvetomir Loukanov, Matthias Gorenflo, Joachim B. Kunz, V. Frismantas, Jean-Pierre Bourquin, Sebastian M. Waszak, Paulina Richter-Pechanska, Cornelia Eckert, Martina U. Muckenthaler, Jan O. Korbel, Martin Schrappe, Büşra Erarslan-Uysal, Tobias Rausch, Andreas E. Kulozik, Malte Paulsen, Vladimir Benes, Ianthe Aem van Belzen, Renate Kirschner-Schwabe, Gunnar Cario, and Diana Ordoñez-Rueada
- Subjects
0301 basic medicine ,Medicine (General) ,T-cell leukemia ,SUMO protein ,ATAC-seq ,Biology ,QH426-470 ,Precursor T-Cell Lymphoblastic Leukemia-Lymphoma ,Chromatin, Epigenetics, Genomics & Functional Genomics ,Article ,03 medical and health sciences ,0302 clinical medicine ,Prophase ,R5-920 ,T‐cell development ,ATAC‐Seq ,medicine ,Genetics ,Humans ,Child ,Gene ,Cancer ,Precursor Cells, T-Lymphoid ,SPI1 ,Articles ,Oncogenes ,medicine.disease ,Chromatin ,Cell biology ,Leukemia ,030104 developmental biology ,chromatin accessibility ,Molecular Medicine ,T‐cell leukemia ,030217 neurology & neurosurgery ,Protein Binding - Abstract
We aimed at identifying the developmental stage at which leukemic cells of pediatric T‐ALLs are arrested and at defining leukemogenic mechanisms based on ATAC‐Seq. Chromatin accessibility maps of seven developmental stages of human healthy T cells revealed progressive chromatin condensation during T‐cell maturation. Developmental stages were distinguished by 2,823 signature chromatin regions with 95% accuracy. Open chromatin surrounding SAE1 was identified to best distinguish thymic developmental stages suggesting a potential role of SUMOylation in T‐cell development. Deconvolution using signature regions revealed that T‐ALLs, including those with mature immunophenotypes, resemble the most immature populations, which was confirmed by TF‐binding motif profiles. We integrated ATAC‐Seq and RNA‐Seq and found DAB1, a gene not related to leukemia previously, to be overexpressed, abnormally spliced and hyper‐accessible in T‐ALLs. DAB1‐negative patients formed a distinct subgroup with particularly immature chromatin profiles and hyper‐accessible binding sites for SPI1 (PU.1), a TF crucial for normal T‐cell maturation. In conclusion, our analyses of chromatin accessibility and TF‐binding motifs showed that pediatric T‐ALL cells are most similar to immature thymic precursors, indicating an early developmental arrest., Analysis of chromatin accessibility of human T‐cell precursors revealed progressive chromatin condensation during maturation. Pediatric T‐ALLs resemble the most immature populations indicating that the epigenetic landscape of this type of leukemia is most similar to the earliest thymic precursors.
- Published
- 2020
15. Clonal evolution mechanisms in NT5C2 mutant relapsed acute lymphoblastic leukemia
- Author
-
Alberto Ambesi-Impiombato, Hossein Khiabanian, Mignon L. Loh, Gannie Tzoneva, Koichi Oshima, Chelsea L. Dieck, Giuseppe Basso, Maria Luisa Sulis, Julie M. Gastier-Foster, Maddalena Paganin, Ilaria Iacobucci, Chioma J. Madubata, Adolfo A. Ferrando, Esmé Waanders, Jiangyan Yu, Raul Rabadan, Charles G. Mullighan, Marta Sanchez-Martin, Motohiro Kato, Katsuyoshi Koh, and Renate Kirschner-Schwabe
- Subjects
0301 basic medicine ,Male ,medicine.medical_treatment ,Mutant ,Drug Resistance ,medicine.disease_cause ,Somatic evolution in cancer ,Mice ,0302 clinical medicine ,IMP Dehydrogenase ,Recurrence ,hemic and lymphatic diseases ,Tumours of the digestive tract Radboud Institute for Molecular Life Sciences [Radboudumc 14] ,Receptor, Notch1 ,Cytotoxicity ,5-Nucleotidase ,5'-Nucleotidase ,Cancer ,Pediatric ,Mutation ,Multidisciplinary ,Guanosine ,Mercaptopurine ,Lymphoblast ,Hematology ,Precursor Cell Lymphoblastic Leukemia-Lymphoma ,Phenotype ,3. Good health ,5.1 Pharmaceuticals ,030220 oncology & carcinogenesis ,Gain of Function Mutation ,Female ,Development of treatments and therapeutic interventions ,Receptor ,Pediatric Cancer ,Childhood Leukemia ,General Science & Technology ,Biology ,Article ,Clonal Evolution ,03 medical and health sciences ,Rare Diseases ,All institutes and research themes of the Radboud University Medical Center ,Nucleotidase ,medicine ,Genetics ,Animals ,Humans ,Cell Proliferation ,Chemotherapy ,Notch1 ,Animal ,Xenograft Model Antitumor Assays ,Disease Models, Animal ,030104 developmental biology ,HEK293 Cells ,Drug Resistance, Neoplasm ,Purines ,Disease Models ,Cancer research ,Neoplasm - Abstract
Relapsed acute lymphoblastic leukaemia (ALL) is associated with resistance to chemotherapy and poor prognosis. Gain-of-function mutations in the 5'-nucleotidase, cytosolic II (NT5C2) gene induce resistance to 6-mercaptopurine and are selectively present in relapsed ALL. Yet, the mechanisms involved in NT5C2 mutation-driven clonal evolution during the initiation of leukaemia, disease progression and relapse remain unknown. Here we use a conditional-and-inducible leukaemia model to demonstrate that expression of NT5C2(R367Q), a highly prevalent relapsed-ALL NT5C2 mutation, induces resistance to chemotherapy with 6-mercaptopurine at the cost of impaired leukaemia cell growth and leukaemia-initiating cell activity. The loss-of-fitness phenotype of NT5C2+/R367Q mutant cells is associated with excess export of purines to the extracellular space and depletion of the intracellular purine-nucleotide pool. Consequently, blocking guanosine synthesis by inhibition of inosine-5'-monophosphate dehydrogenase (IMPDH) induced increased cytotoxicity against NT5C2-mutant leukaemia lymphoblasts. These results identify the fitness cost of NT5C2 mutation and resistance to chemotherapy as key evolutionary drivers that shape clonal evolution in relapsed ALL and support a role for IMPDH inhibition in the treatment of ALL.
- Published
- 2018
16. Structure and mechanisms of NT5C2 mutations driving thiopurine resistance in relapsed lymphoblastic leukemia
- Author
-
Scott Lew, Farhad Forouhar, Renate Kirschner-Schwabe, Adolfo A. Ferrando, Jayaraman Seetharaman, Chelsea L. Dieck, Gannie Tzoneva, Liang Tong, Zachary Carpenter, Marta Sanchez-Martin, and Alberto Ambesi-Impiombato
- Subjects
0301 basic medicine ,Models, Molecular ,Protein Conformation, alpha-Helical ,Cancer Research ,Antimetabolites, Antineoplastic ,medicine.medical_treatment ,Allosteric regulation ,medicine.disease_cause ,Article ,03 medical and health sciences ,Jurkat Cells ,Structure-Activity Relationship ,0302 clinical medicine ,Allosteric Regulation ,Recurrence ,Catalytic Domain ,Hydrolase ,medicine ,Animals ,Humans ,Gene ,5'-Nucleotidase ,Mutation ,Chemotherapy ,Thiopurine methyltransferase ,biology ,Mechanism (biology) ,Chemistry ,Gene Expression Regulation, Leukemic ,Mercaptopurine ,Cell Biology ,Precursor Cell Lymphoblastic Leukemia-Lymphoma ,Mice, Inbred C57BL ,Cytosol ,030104 developmental biology ,HEK293 Cells ,Oncology ,Drug Resistance, Neoplasm ,030220 oncology & carcinogenesis ,Cancer research ,biology.protein - Abstract
Summary Activating mutations in the cytosolic 5′-nucleotidase II gene NT5C2 drive resistance to 6-mercaptopurine in acute lymphoblastic leukemia. Here we demonstrate that constitutively active NT5C2 mutations K359Q and L375F reconfigure the catalytic center for substrate access and catalysis in the absence of allosteric activator. In contrast, most relapse-associated mutations, which involve the arm segment and residues along the surface of the inter-monomeric cavity, disrupt a built-in switch-off mechanism responsible for turning off NT5C2. In addition, we show that the C-terminal acidic tail lost in the Q523X mutation functions to restrain NT5C2 activation. These results uncover dynamic mechanisms of enzyme regulation targeted by chemotherapy resistance-driving NT5C2 mutations, with important implications for the development of NT5C2 inhibitor therapies.
- Published
- 2018
17. Author Correction : The landscape of genomic alterations across childhood cancers
- Author
-
Christian P. Kratz, Benedikt Brors, Manfred Gessler, Jan J. Molenaar, Sebastian M. Waszak, Dietmar R. Lohmann, Vasilisa A. Rudneva, Kristian W. Pajtler, Gideon Zipprich, Daniel Baumhoer, Roland Kappler, Michael Heinold, Matthias Schlesner, Birgit Burkhardt, Stefan Rutkowski, Ewa Koscielniak, Sander Lambo, Sebastian Bender, Stephan Wolf, Michaela Nathrath, Angela J. Waanders, Jürgen Eils, Barbara C. Worst, Angelika Eggert, Michael C. Frühwald, Susanne Gröbner, Cornelia Eckert, Barbara Hutter, Hendrik Witt, Yanling Liu, Paul A. Northcott, Maia Segura-Wang, Pablo Landgraf, Sebastian Brabetz, Danny A. Zwijnenburg, Jenny Wegert, Arndt Borkhardt, Marcel Kool, Gudrun Fleischhack, Renate Kirschner-Schwabe, Kortine Kleinheinz, Christof M. Kramm, Daniel Hübschmann, Pascal Johann, Simone Fulda, Dominik Sturm, Gunther Richter, Peter Lichter, Katja von Hoff, Michaela Kuhlen, Gilles Vassal, Jan O. Korbel, Johannes H. Schulte, Rosario M. Piro, Joachim Weischenfeldt, Reiner Siebert, Udo Kontny, Christian Lawerenz, Gnana Prakash Balasubramanian, David T.W. Jones, Charlotte M. Niemeyer, Uta Dirksen, Lukas Chavez, Serap Erkek, Adam C. Resnick, Frank Westermann, Stefan S. Bielack, Stefan M. Pfister, Ursula D. Weber, Xin Zhou, Marc Zapatka, Cornelis M. van Tilburg, Roland Eils, Jan Koster, Stefan Burdach, Simone Hettmer, Thomas Klingebiel, Andreas E. Kulozik, Olaf Witt, Ivo Buchhalter, Pichai Raman, Claudia Blattmann, Jinghui Zhang, and Elke Pfaff
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Multidisciplinary ,biology ,Published Erratum ,MEDLINE ,Medizin ,Translational research ,biology.organism_classification ,language.human_language ,German ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Paediatric cancer ,030220 oncology & carcinogenesis ,Family medicine ,medicine ,language ,book.journal ,Center (algebra and category theory) ,Memphis ,Psychology ,Developmental neurobiology ,book - Abstract
In this Article, author Benedikt Brors was erroneously associated with affiliation number '8' (Department of Developmental Neurobiology, St Jude Children's Research Hospital, Memphis, Tennessee, USA); the author's two other affiliations (affiliations '3' and '7', both at the German Cancer Research Center (DKFZ)) were correct. This has been corrected online.
- Published
- 2018
18. Intragenic amplification of PAX5: a novel subgroup in B-cell precursor acute lymphoblastic leukemia?
- Author
-
Yuka Yamashita, Grazia Fazio, Hélène Cavé, Sabine Strehl, Kentaro Ohki, Judith M. Boer, Renate Kirschner-Schwabe, Agata Pastorczak, Eva Froňková, Joelle Tchinda, Markéta Žaliová, Marleen Bakkus, Monique L. den Boer, Cristina Leschi, Christine J. Harrison, Lucy Chilton, Stefanie Groeneveld-Krentz, Renate Panzer-Grümayer, Anthony V. Moorman, Wojciech Młynarski, Martin Stanulla, Giovanni Cazzaniga, Andishe Attarbaschi, Maria S. Pombo-de-Oliveira, Thayana Conceição Barbosa, Rosemary Sutton, Jan Trka, Gisela Barbany, Karin Nebral, Claire Schwab, Toshihiko Imamura, Roland P. Kuiper, Esmé Waanders, Ingegerd Ivanov Öfverholm, University of Zurich, Clinical Biology, Hematology, Pediatrics, Schwab, C, Nebral, K, Chilton, L, Leschi, C, Waanders, E, Boer, J, Žaliová, M, Sutton, R, Öfverholm, I, Ohki, K, Yamashita, Y, Groeneveld-Krentz, S, Froňková, E, Bakkus, M, Tchinda, J, Barbosa, T, Fazio, G, Mlynarski, W, Pastorczak, A, Cazzaniga, G, Pombo-de-Oliveira, M, Trka, J, Kirschner-Schwabe, R, Imamura, T, Barbany, G, Stanulla, M, Attarbaschi, A, Panzer-Grümayer, R, Kuiper, R, den Boer, M, Cavé, H, Moorman, A, Harrison, C, and Strehl, S
- Subjects
0301 basic medicine ,MED/03 - GENETICA MEDICA ,business.industry ,Lymphoblastic Leukemia ,2720 Hematology ,hemic and immune systems ,610 Medicine & health ,Hematology ,3. Good health ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Text mining ,medicine.anatomical_structure ,immune system diseases ,10036 Medical Clinic ,030220 oncology & carcinogenesis ,hemic and lymphatic diseases ,Cancer research ,Medicine ,PAX5 ,business ,PAX5, B-cell precursor acute lymphoblastic leukemia, poor outcome ,B cell - Abstract
Key Points Intragenic PAX5 amplification defines a novel, relapse-prone subtype of B-cell precursor acute lymphoblastic leukemia with a poor outcome.
- Published
- 2017
19. NOTCH1 mutation, TP53 alteration and myeloid antigen expression predict outcome heterogeneity in children with first relapse of T-cell acute lymphoblastic leukemia
- Author
-
Stefanie Groeneveld-Krentz, Obul Reddy Bandapalli, Günter Henze, Cornelia Eckert, Martina U. Muckenthaler, Leonid Karawajew, Richard Ratei, Corinne Kox, Katharina Schedel, Renate Kirschner-Schwabe, Jana Hof, Wolf-Dieter Ludwig, Joachim B. Kunz, Andreas E. Kulozik, Peter Rhein, and Arend von Stackelberg
- Subjects
Oncology ,medicine.medical_specialty ,business.industry ,Proportional hazards model ,Lymphoblastic Leukemia ,T cell ,Hematopoietic stem cell ,Hematology ,Myeloid antigen ,03 medical and health sciences ,First relapse ,0302 clinical medicine ,medicine.anatomical_structure ,Immunophenotyping ,hemic and lymphatic diseases ,030220 oncology & carcinogenesis ,Internal medicine ,Immunology ,Medicine ,business ,Online Only Articles ,030215 immunology - Abstract
Relapse of T-cell acute lymphoblastic leukemia (T-ALL) has a dismal prognosis, with only 20% of afflicted children surviving.[1][1] Children with relapsed T-ALL are commonly treated within high-risk arms of second-line treatment protocols that include mandatory hematopoietic stem cell
- Published
- 2017
20. Activating mutations in the NT5C2 nucleotidase gene drive chemotherapy resistance in relapsed ALL
- Author
-
Martin S. Tallman, Elisabeth Paietta, Gannie Tzoneva, Renate Kirschner-Schwabe, Adolfo A. Ferrando, Jacob M. Rowe, Arianne Perez-Garcia, Valeria Tosello, Maddalena Paganin, Giuseppe Basso, Hossein Khiabanian, Zachary Carpenter, Teresa Palomero, Maddalena Allegretta, Jana Hof, Raul Rabadan, and Janis Racevskis
- Subjects
Nucleotidase activity ,medicine.medical_treatment ,Antineoplastic Agents ,Drug resistance ,Biology ,General Biochemistry, Genetics and Molecular Biology ,Article ,Malignant transformation ,5'-nucleotidase ,Cell Line ,03 medical and health sciences ,0302 clinical medicine ,Recurrence ,Nucleotidase ,medicine ,Humans ,Thioguanine ,5'-Nucleotidase ,030304 developmental biology ,0303 health sciences ,Chemotherapy ,Base Sequence ,Mercaptopurine ,leukemia ,Cancer ,General Medicine ,Sequence Analysis, DNA ,Precursor Cell Lymphoblastic Leukemia-Lymphoma ,medicine.disease ,3. Good health ,Leukemia ,HEK293 Cells ,Drug Resistance, Neoplasm ,030220 oncology & carcinogenesis ,Immunology ,Mutation ,Cancer research ,Arabinonucleosides - Abstract
Acute lymphoblastic leukemia (ALL) is an aggressive hematological tumor resulting from the malignant transformation of lymphoid progenitors. Despite intensive chemotherapy, 20% of pediatric patients and over 50% of adult patients with ALL do not achieve a complete remission or relapse after intensified chemotherapy, making disease relapse and resistance to therapy the most substantial challenge in the treatment of this disease. Using whole-exome sequencing, we identify mutations in the cytosolic 5'-nucleotidase II gene (NT5C2), which encodes a 5'-nucleotidase enzyme that is responsible for the inactivation of nucleoside-analog chemotherapy drugs, in 20/103 (19%) relapse T cell ALLs and 1/35 (3%) relapse B-precursor ALLs. NT5C2 mutant proteins show increased nucleotidase activity in vitro and conferred resistance to chemotherapy with 6-mercaptopurine and 6-thioguanine when expressed in ALL lymphoblasts. These results support a prominent role for activating mutations in NT5C2 and increased nucleoside-analog metabolism in disease progression and chemotherapy resistance in ALL.
- Published
- 2013
21. Frequent and sex-biased deletion of SLX4IP by illegitimate V(D)J-mediated recombination in childhood acute lymphoblastic leukemia
- Author
-
Markus Metzler, Beat Bornhauser, Renate Kirschner-Schwabe, Eva Ellinghaus, Martin Stanulla, Marketa Zaliova, Claus R. Bartram, T Bartram, Gunnar Cario, Jean-Pierre Bourquin, Geertruy te Kronnie, Petra Dörge, Cornelia Eckert, Andrea Teigler-Schlegel, Anja Möricke, Rolf Koehler, Arend von Stackelberg, Smadar Avigad, André Schrauder, Jan Trka, Magdalena Sokalska-Duhme, Giuseppe Basso, Andre Franke, Giovanni Cazzaniga, Ivana Hermanova, B Meissner, Martin Schrappe, Martin Zimmermann, Andishe Attarbaschi, Renate Panzer-Grümayer, Julia Hauer, Shai Izraeli, Meissner, B, Bartram, T, Eckert, C, Trka, J, Panzer-Grümayer, R, Hermanova, I, Ellinghaus, E, Franke, A, Möricke, A, Schrauder, A, Teigler-Schlegel, A, Dörge, P, von Stackelberg, A, Basso, G, Bartram, C, Kirschner-Schwabe, R, Bornhäuser, B, Bourquin, J, Cazzaniga, G, Hauer, J, Attarbaschi, A, Izraeli, S, Zaliova, M, Cario, G, Zimmermann, M, Avigad, S, Sokalska-Duhme, M, Metzler, M, Schrappe, M, Koehler, R, Te Kronnie, G, and Stanulla, M
- Subjects
Male ,Basic Helix-Loop-Helix Transcription Factor ,Cohort Studies ,0302 clinical medicine ,Medizinische Fakultät ,Basic Helix-Loop-Helix Transcription Factors ,Child ,Genetics (clinical) ,T-Cell Acute Lymphocytic Leukemia Protein 1 ,Genetics ,0303 health sciences ,Proto-Oncogene Protein ,Proto-Oncogene Proteins c-et ,V(D)J recombination ,General Medicine ,Precursor Cell Lymphoblastic Leukemia-Lymphoma ,030220 oncology & carcinogenesis ,Child, Preschool ,Core Binding Factor Alpha 2 Subunit ,Recombinase ,Female ,Human ,Adolescent ,Locus (genetics) ,Biology ,Recombinases ,03 medical and health sciences ,Proto-Oncogene Proteins ,Acute lymphocytic leukemia ,medicine ,Humans ,ddc:610 ,Molecular Biology ,Childhood Acute Lymphoblastic Leukemia ,030304 developmental biology ,Proto-Oncogene Proteins c-ets ,Breakpoint ,Infant ,Repressor Protein ,medicine.disease ,V(D)J Recombination ,Repressor Proteins ,ETV6 ,Cohort Studie ,Carrier Proteins ,Carrier Protein ,Gene Deletion ,TAL1 - Abstract
Acute lymphoblastic leukemia (ALL) accounts for ∼25% of pediatric malignancies. Of interest, the incidence of ALL is observed ∼20% higher in males relative to females. The mechanism behind the phenomenon of sex-specific differences is presently not understood. Employing genome-wide genetic aberration screening in 19 ALL samples, one of the most recurrent lesions identified was monoallelic deletion of the 5' region of SLX4IP. We characterized this deletion by conventional molecular genetic techniques and analyzed its interrelationships with biological and clinical characteristics using specimens and data from 993 pediatric patients enrolled into trial AIEOP-BFM ALL 2000. Deletion of SLX4IP was detected in ∼30% of patients. Breakpoints within SLX4IP were defined to recurrent positions and revealed junctions with typical characteristics of illegitimate V(D)J-mediated recombination. In initial and validation analyses, SLX4IP deletions were significantly associated with male gender and ETV6/RUNX1-rearranged ALL (both overall P < 0.0001). For mechanistic validation, a second recurrent deletion affecting TAL1 and caused by the same molecular mechanism was analyzed in 1149 T-cell ALL patients. Validating a differential role by sex of illegitimate V(D)J-mediated recombination at the TAL1 locus, 128 out of 1149 T-cell ALL samples bore a deletion and males were significantly more often affected (P = 0.002). The repeatedly detected association of SLX4IP deletion with male sex and the extension of the sex bias to deletion of the TAL1 locus suggest that differential illegitimate V(D)J-mediated recombination events at specific loci may contribute to the consistent observation of higher incidence rates of childhood ALL in boys compared with girls.
- Published
- 2014
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.