1
|
Wang HC, Chen R, Yang W, Li Y, Muthukumar R, Patel RM, Casey EB, Denby E, Magee JA. Kmt2c restricts G-CSF-driven HSC mobilization and granulocyte production in a methyltransferase-independent manner. Cell Rep 2024; 43:114542. [PMID: 39046877 PMCID: PMC11423277 DOI: 10.1016/j.celrep.2024.114542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 06/10/2024] [Accepted: 07/09/2024] [Indexed: 07/27/2024] Open
Abstract
Granulocyte colony-stimulating factor (G-CSF) is widely used to enhance myeloid recovery after chemotherapy and to mobilize hematopoietic stem cells (HSCs) for transplantation. Unfortunately, through the course of chemotherapy, cancer patients can acquire leukemogenic mutations that cause therapy-related myelodysplastic syndrome (MDS) or acute myeloid leukemia (AML). This raises the question of whether therapeutic G-CSF might potentiate therapy-related MDS/AML by disproportionately stimulating mutant HSCs and other myeloid progenitors. A common mutation in therapy-related MDS/AML involves chromosome 7 deletions that inactivate many tumor suppressor genes, including KMT2C. Here, we show that Kmt2c deletions hypersensitize murine HSCs and myeloid progenitors to G-CSF, as evidenced by increased HSC mobilization and enhanced granulocyte production from granulocyte-monocyte progenitors (GMPs). Furthermore, Kmt2c attenuates the G-CSF response independently from its SET methyltransferase function. Altogether, the data raise concerns that monosomy 7 can hypersensitize progenitors to G-CSF, such that clinical use of G-CSF may amplify the risk of therapy-related MDS/AML.
Collapse
Affiliation(s)
- Helen C Wang
- Department of Pediatrics, Division of Hematology and Oncology, Washington University School of Medicine, 660 S. Euclid Avenue, St. Louis, MO 63110, USA
| | - Ran Chen
- Department of Pediatrics, Division of Hematology and Oncology, Washington University School of Medicine, 660 S. Euclid Avenue, St. Louis, MO 63110, USA
| | - Wei Yang
- Department of Genetics, Washington University School of Medicine, 660 S. Euclid Avenue, St. Louis, MO 63110, USA
| | - Yanan Li
- Department of Pediatrics, Division of Hematology and Oncology, Washington University School of Medicine, 660 S. Euclid Avenue, St. Louis, MO 63110, USA
| | - Rohini Muthukumar
- Department of Pediatrics, Division of Hematology and Oncology, Washington University School of Medicine, 660 S. Euclid Avenue, St. Louis, MO 63110, USA
| | - Riddhi M Patel
- Department of Pediatrics, Division of Hematology and Oncology, Washington University School of Medicine, 660 S. Euclid Avenue, St. Louis, MO 63110, USA
| | - Emily B Casey
- Department of Pediatrics, Division of Hematology and Oncology, Washington University School of Medicine, 660 S. Euclid Avenue, St. Louis, MO 63110, USA
| | - Elisabeth Denby
- Department of Pediatrics, Division of Hematology and Oncology, Washington University School of Medicine, 660 S. Euclid Avenue, St. Louis, MO 63110, USA
| | - Jeffrey A Magee
- Department of Pediatrics, Division of Hematology and Oncology, Washington University School of Medicine, 660 S. Euclid Avenue, St. Louis, MO 63110, USA; Department of Genetics, Washington University School of Medicine, 660 S. Euclid Avenue, St. Louis, MO 63110, USA.
| |
Collapse
|
2
|
Ma S, Li P, Liu H, Xi Y, Xu Q, Qi J, Wang J, Li L, Wang J, Hu J, He H, Han C, Bai L. Genome-wide association analysis of the primary feather growth traits of duck: identification of potential Loci for growth regulation. Poult Sci 2022; 102:102243. [PMID: 36334470 PMCID: PMC9636485 DOI: 10.1016/j.psj.2022.102243] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 10/05/2022] [Accepted: 10/05/2022] [Indexed: 11/28/2022] Open
Abstract
The feather is an important epidermal appendage, plays an important role in the life activities of avian specie, and has important economic value. Revealing the molecular regulation mechanism of feather growth has a significant meaning in studying adaptive evolution, physiology, and mating of avian species and also provides a theoretical reference for poultry breeding. In this study, the genome-wide association analysis (GWAS) of 358 ducks was based on primary feather length phenotypic data (28-60 d), length growth rates (LGRs), and maturity scores (60 d) to explore the genetic basis affecting feather growth and maturation. The results showed that, among the primary feather 1 to 5 in ducks, the mean LGR of primary feather 2 was the fastest, with the longest length. The primary feathers in males grew and matured slightly faster than in females. The mean maturity scores of primary feather 10∼7 were higher than primary feather 1 to 3 in ducks. GWAS further showed 116 SNPs associated with feather length traits. In addition, 2 candidate regions (Chr1: 127,407,230-127,524,879 bp and Chr21: 182,061,707-183,616,298 bp) were associated with LGR, which contain total 13 candidate genes (The extremely significant SNPs were mainly located in 2 genes: Chr1: REPS2 and Chr21: PTPRT). Four candidate regions (Chr1: 29,113,036-28,675,018 bp, Chr2: 18,253,612-149,111,290 bp, Chr15: 6,489,774 to 12,138,221 bp and Chr21: 6,578,021-8,472,904 bp) were associated with feather maturity, which contain total 24 candidate genes (The extremely significant SNPs were mainly located in 4 genes: Chr1: IMMP2L, DOCK4 and DDX10, Chr2: LDLRAD4). In conclusion, sex factors influence feather growth and maturity, and the genetic basis of the growth /maturity trait between different feathers is similar. REPS2, PTPRT genes, and IMMP2L, DOCK4, DDX10, and LDLRAD4 are important candidate genes that influence feather growth and maturity, respectively.
Collapse
Affiliation(s)
- Shengchao Ma
- Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal and Technology (Institute of Animal Genetics and Breeding), Sichuan Agricultural University, P. R. China,Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, P. R. China,College of Animal Science, Xinjiang Agricultural University, P. R. China
| | - Pengcheng Li
- Berry Genomics Corporation, Beijing 100015, P. R. China
| | - Hehe Liu
- Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal and Technology (Institute of Animal Genetics and Breeding), Sichuan Agricultural University, P. R. China,Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, P. R. China,Corresponding author:
| | - Yang Xi
- Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal and Technology (Institute of Animal Genetics and Breeding), Sichuan Agricultural University, P. R. China,Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, P. R. China
| | - Qian Xu
- Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal and Technology (Institute of Animal Genetics and Breeding), Sichuan Agricultural University, P. R. China,Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, P. R. China
| | - Jingjing Qi
- Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal and Technology (Institute of Animal Genetics and Breeding), Sichuan Agricultural University, P. R. China,Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, P. R. China
| | - Jianmei Wang
- Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal and Technology (Institute of Animal Genetics and Breeding), Sichuan Agricultural University, P. R. China,Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, P. R. China
| | - Liang Li
- Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal and Technology (Institute of Animal Genetics and Breeding), Sichuan Agricultural University, P. R. China,Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, P. R. China
| | - Jiwen Wang
- Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal and Technology (Institute of Animal Genetics and Breeding), Sichuan Agricultural University, P. R. China,Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, P. R. China
| | - Jiwei Hu
- Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal and Technology (Institute of Animal Genetics and Breeding), Sichuan Agricultural University, P. R. China,Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, P. R. China
| | - Hua He
- Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal and Technology (Institute of Animal Genetics and Breeding), Sichuan Agricultural University, P. R. China,Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, P. R. China
| | - Chunchun Han
- Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal and Technology (Institute of Animal Genetics and Breeding), Sichuan Agricultural University, P. R. China,Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, P. R. China
| | - Lili Bai
- Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal and Technology (Institute of Animal Genetics and Breeding), Sichuan Agricultural University, P. R. China,Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, P. R. China
| |
Collapse
|
3
|
Abstract
PURPOSE OF REVIEW Loss of chromosome 7 has long been associated with adverse-risk myeloid malignancy. In the last decade, CUX1 has been identified as a critical tumor suppressor gene (TSG) located within a commonly deleted segment of chromosome arm 7q. Additional genes encoded on 7q have also been identified as bona fide myeloid tumor suppressors, further implicating chromosome 7 deletions in disease pathogenesis. This review will discuss the clinical implications of del(7q) and CUX1 mutations, both in disease and clonal hematopoiesis, and synthesize recent literature on CUX1 and other chromosome 7 TSGs. RECENT FINDINGS Two major studies, including a new mouse model, have been published that support a role for CUX1 inactivation in the development of myeloid neoplasms. Additional recent studies describe the cellular and hematopoietic effects from loss of the 7q genes LUC7L2 and KMT2C/MLL3, and the implications of chromosome 7 deletions in clonal hematopoiesis. SUMMARY Mounting evidence supports CUX1 as being a key chromosome 7 TSG. As 7q encodes additional myeloid regulators and tumor suppressors, improved models of chromosome loss are needed to interrogate combinatorial loss of these critical 7q genes.
Collapse
Affiliation(s)
| | - Megan E McNerney
- Department of Pathology
- Department of Pediatrics, Section of Hematology/Oncology
- The University of Chicago Medicine Comprehensive Cancer Center, The University of Chicago, Chicago, Illinois, USA
| |
Collapse
|
4
|
Deng Y, Zhou Y, Shi J, Yang J, Huang H, Zhang M, Wang S, Ma Q, Liu Y, Li B, Yan J, Yang H. Potential genetic biomarkers predict adverse pregnancy outcome during early and mid-pregnancy in women with systemic lupus erythematosus. Front Endocrinol (Lausanne) 2022; 13:957010. [PMID: 36465614 PMCID: PMC9708709 DOI: 10.3389/fendo.2022.957010] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 11/01/2022] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Effectively predicting the risk of adverse pregnancy outcome (APO) in women with systemic lupus erythematosus (SLE) during early and mid-pregnancy is a challenge. This study was aimed to identify potential markers for early prediction of APO risk in women with SLE. METHODS The GSE108497 gene expression dataset containing 120 samples (36 patients, 84 controls) was downloaded from the Gene Expression Omnibus database. Weighted gene co-expression network analysis (WGCNA) was performed, and differentially expressed genes (DEGs) were screened to define candidate APO marker genes. Next, three individual machine learning methods, random forest, support vector machine-recursive feature elimination, and least absolute shrinkage and selection operator, were combined to identify feature genes from the APO candidate set. The predictive performance of feature genes for APO risk was assessed using area under the receiver operating characteristic curve (AUC) and calibration curves. The potential functions of these feature genes were finally analyzed by conventional gene set enrichment analysis and CIBERSORT algorithm analysis. RESULTS We identified 321 significantly up-regulated genes and 307 down-regulated genes between patients and controls, along with 181 potential functionally associated genes in the WGCNA analysis. By integrating these results, we revealed 70 APO candidate genes. Three feature genes, SEZ6, NRAD1, and LPAR4, were identified by machine learning methods. Of these, SEZ6 (AUC = 0.753) showed the highest in-sample predictive performance for APO risk in pregnant women with SLE, followed by NRAD1 (AUC = 0.694) and LPAR4 (AUC = 0.654). After performing leave-one-out cross validation, corresponding AUCs for SEZ6, NRAD1, and LPAR4 were 0.731, 0.668, and 0.626, respectively. Moreover, CIBERSORT analysis showed a positive correlation between regulatory T cell levels and SEZ6 expression (P < 0.01), along with a negative correlation between M2 macrophages levels and LPAR4 expression (P < 0.01). CONCLUSIONS Our preliminary findings suggested that SEZ6, NRAD1, and LPAR4 might represent the useful genetic biomarkers for predicting APO risk during early and mid-pregnancy in women with SLE, and enhanced our understanding of the origins of pregnancy complications in pregnant women with SLE. However, further validation was required.
Collapse
Affiliation(s)
- Yu Deng
- Department of Obstetrics and Gynecology, Peking University First Hospital, Beijing, China
- Beijing Key Laboratory of Maternal Fetal Medicine of Gestational Diabetes Mellitus, Beijing, China
| | - Yiran Zhou
- Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Jiangcheng Shi
- School of Life Sciences, Tiangong University, Tianjin, China
| | - Junting Yang
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, China
| | - Hong Huang
- Department of Rheumatology and Clinical Immunology, Peking University First Hospital, Beijing, China
| | - Muqiu Zhang
- Department of Obstetrics and Gynecology, Peking University First Hospital, Beijing, China
- Beijing Key Laboratory of Maternal Fetal Medicine of Gestational Diabetes Mellitus, Beijing, China
| | - Shuxian Wang
- Department of Obstetrics and Gynecology, Peking University First Hospital, Beijing, China
- Beijing Key Laboratory of Maternal Fetal Medicine of Gestational Diabetes Mellitus, Beijing, China
| | - Qian Ma
- Department of Clinical Laboratory, Peking University First Hospital, Beijing, China
| | - Yingnan Liu
- Department of Obstetrics and Gynecology, Peking University First Hospital, Beijing, China
- Beijing Key Laboratory of Maternal Fetal Medicine of Gestational Diabetes Mellitus, Beijing, China
| | - Boya Li
- Department of Obstetrics and Gynecology, Peking University First Hospital, Beijing, China
- Beijing Key Laboratory of Maternal Fetal Medicine of Gestational Diabetes Mellitus, Beijing, China
| | - Jie Yan
- Department of Obstetrics and Gynecology, Peking University First Hospital, Beijing, China
- Beijing Key Laboratory of Maternal Fetal Medicine of Gestational Diabetes Mellitus, Beijing, China
| | - Huixia Yang
- Department of Obstetrics and Gynecology, Peking University First Hospital, Beijing, China
- Beijing Key Laboratory of Maternal Fetal Medicine of Gestational Diabetes Mellitus, Beijing, China
- *Correspondence: Huixia Yang,
| |
Collapse
|
5
|
Hecht A, Meyer JA, Jann JC, Sockel K, Giagounidis A, Götze KS, Letsch A, Haase D, Schlenk RF, Haferlach T, Schafhausen P, Bug G, Lübbert M, Thol F, Büsche G, Schuler E, Nowak V, Obländer J, Fey S, Müller N, Metzgeroth G, Hofmann WK, Germing U, Nolte F, Reinwald M, Nowak D. Genome-wide DNA methylation analysis pre- and post-lenalidomide treatment in patients with myelodysplastic syndrome with isolated deletion (5q). Ann Hematol 2021; 100:1463-1471. [PMID: 33903952 PMCID: PMC8116243 DOI: 10.1007/s00277-021-04492-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 03/08/2021] [Indexed: 11/28/2022]
Abstract
Myelodysplastic syndrome (MDS) with isolated deletion of chromosome 5q (MDS del5q) is a distinct subtype of MDS with quite favorable prognosis and excellent response to treatment with lenalidomide. Still, a relevant percentage of patients do not respond to lenalidomide and even experience progression to acute myeloid leukemia (AML). In this study, we aimed to investigate whether global DNA methylation patterns could predict response to lenalidomide. Genome-wide DNA methylation analysis using Illumina 450k methylation arrays was performed on n=51 patients with MDS del5q who were uniformly treated with lenalidomide in a prospective multicenter trial of the German MDS study group. To study potential direct effects of lenalidomide on DNA methylation, 17 paired samples pre- and post-treatment were analyzed. Our results revealed no relevant effect of lenalidomide on methylation status. Furthermore, methylation patterns prior to therapy could not predict lenalidomide response. However, methylation clustering identified a group of patients with a trend towards inferior overall survival. These patients showed hypermethylation of several interesting target genes, including genes of relevant signaling pathways, potentially indicating the evaluation of novel therapeutic targets.
Collapse
Affiliation(s)
- Anna Hecht
- Department of Hematology and Oncology, University Hospital Mannheim, Mannheim, Germany.
| | - Julia A Meyer
- Department of Pediatrics, University of California San Francisco, San Francisco, CA, USA
| | - Johann-Christoph Jann
- Department of Hematology and Oncology, University Hospital Mannheim, Mannheim, Germany
| | - Katja Sockel
- Department of Hematology, University Hospital Dresden, Dresden, Germany
| | - Aristoteles Giagounidis
- Department of Hematology, Oncology and Palliative Care, Marien Hospital Duesseldorf, Duesseldorf, Germany
| | - Katharina S Götze
- Department of Internal Medicine, Technical University of Munich, Munich, Germany
| | - Anne Letsch
- Department of Hematology and Oncology, Charité, Benjamin Franklin University, Berlin, Germany
| | - Detlef Haase
- Clinics of Hematology and Medical Oncology, University Medicine Goettingen, Goettingen, Germany
| | - Richard F Schlenk
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany
| | | | - Philippe Schafhausen
- Department of Oncology, Hematology, BMT with section Pneumology, Hubertus Wald Cancer Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Gesine Bug
- Department of Internal Medicine, University Hospital Frankfurt, Frankfurt, Germany
| | - Michael Lübbert
- Internal Medicine, University Hospital Freiburg, Freiburg, Germany
| | - Felicitas Thol
- Department of Hematology, Hemostasis, Oncology, and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | - Guntram Büsche
- Hannover Medical School, Institute of Pathology, Hannover, Germany
| | - Esther Schuler
- Department for Hematology, Oncology and Clinical Immunology, University Hospital, Heinrich-Heine-University, Duesseldorf, Germany
| | - Verena Nowak
- Department of Hematology and Oncology, University Hospital Mannheim, Mannheim, Germany
| | - Julia Obländer
- Department of Hematology and Oncology, University Hospital Mannheim, Mannheim, Germany
| | - Stephanie Fey
- Department of Hematology and Oncology, University Hospital Mannheim, Mannheim, Germany
| | - Nadine Müller
- Department of Hematology and Oncology, University Hospital Mannheim, Mannheim, Germany
| | - Georgia Metzgeroth
- Department of Hematology and Oncology, University Hospital Mannheim, Mannheim, Germany
| | - Wolf-Karsten Hofmann
- Department of Hematology and Oncology, University Hospital Mannheim, Mannheim, Germany
| | - Ulrich Germing
- Department for Hematology, Oncology and Clinical Immunology, University Hospital, Heinrich-Heine-University, Duesseldorf, Germany
| | - Florian Nolte
- Department of Hematology and Oncology, University Hospital Mannheim, Mannheim, Germany
| | - Mark Reinwald
- Brandenburg Medical School Theodor Fontane, Brandenburg an der Havel, Germany
| | - Daniel Nowak
- Department of Hematology and Oncology, University Hospital Mannheim, Mannheim, Germany
| |
Collapse
|
6
|
Stomper J, Meier R, Ma T, Pfeifer D, Ihorst G, Blagitko-Dorfs N, Greve G, Zimmer D, Platzbecker U, Hagemeijer A, Schmitt-Graeff I, Lübbert M. Integrative study of EZH2 mutational status, copy number, protein expression and H3K27 trimethylation in AML/MDS patients. Clin Epigenetics 2021; 13:77. [PMID: 33845873 PMCID: PMC8043064 DOI: 10.1186/s13148-021-01052-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Accepted: 03/08/2021] [Indexed: 11/21/2022] Open
Abstract
Background Mutations in the EZH2 gene are recurrently found in patients with myeloid neoplasms and are associated with a poor prognosis. We aimed to characterize genetic and epigenetic alterations of EZH2 in 58 patients (51 with acute myeloid leukemia and 7 with myelodysplastic or myeloproliferative neoplasms) by integrating data on EZH2 mutational status, co-occurring mutations, and EZH2 copy number status with EZH2 protein expression, histone H3K27 trimethylation, and EZH2 promoter methylation. Results EZH2 was mutated in 6/51 acute myeloid leukemia patients (12%) and 7/7 patients with other myeloid neoplasms. EZH2 mutations were not overrepresented in patients with chromosome 7q deletions or losses. In acute myeloid leukemia patients, EZH2 mutations frequently co-occurred with CEBPA (67%), ASXL1 (50%), TET2 and RAD21 mutations (33% each). In EZH2-mutated patients with myelodysplastic or myeloproliferative neoplasms, the most common co-mutations were in ASXL1 (100%), NRAS, RUNX1, and STAG2 (29% each). EZH2 mutations were associated with a significant decrease in EZH2 expression (p = 0.0002), which was similar in patients with chromosome 7 aberrations and patients with intact chromosome 7. An association between EZH2 protein expression and H3K27 trimethylation was observed in EZH2-unmutated patients (R2 = 0.2, p = 0.01). The monoallelic state of EZH2 was not associated with EZH2 promoter hypermethylation. In multivariable analyses, EZH2 mutations were associated with a trend towards an increased risk of death (hazard ratio 2.51 [95% confidence interval 0.87–7.25], p = 0.09); similarly, low EZH2 expression was associated with elevated risk (hazard ratio 2.54 [95% confidence interval 1.07–6.04], p = 0.04). Conclusions Perturbations of EZH2 activity in AML/MDS occur on different, genetic and non-genetic levels. Both low EZH2 protein expression and, by trend, EZH2 gene mutations predicted inferior overall survival of AML patients receiving standard chemotherapy. Supplementary information The online version contains supplementary material available at 10.1186/s13148-021-01052-2.
Collapse
Affiliation(s)
- Julia Stomper
- Department of Medicine I (Hematology, Oncology and Stem Cell Transplantation), Medical Center - University of Freiburg, Freiburg, Germany
| | - Ruth Meier
- Department of Medicine I (Hematology, Oncology and Stem Cell Transplantation), Medical Center - University of Freiburg, Freiburg, Germany
| | - Tobias Ma
- Department of Medicine I (Hematology, Oncology and Stem Cell Transplantation), Medical Center - University of Freiburg, Freiburg, Germany
| | - Dietmar Pfeifer
- Department of Medicine I (Hematology, Oncology and Stem Cell Transplantation), Medical Center - University of Freiburg, Freiburg, Germany
| | - Gabriele Ihorst
- Clinical Trials Unit, Faculty of Medicine, Medical Center - University of Freiburg, Freiburg, Germany
| | - Nadja Blagitko-Dorfs
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency (CCI), Medical Center - University of Freiburg, Freiburg, Germany
| | - Gabriele Greve
- Department of Medicine I (Hematology, Oncology and Stem Cell Transplantation), Medical Center - University of Freiburg, Freiburg, Germany
| | - Dennis Zimmer
- Department of Medicine I (Hematology, Oncology and Stem Cell Transplantation), Medical Center - University of Freiburg, Freiburg, Germany
| | - Uwe Platzbecker
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Partner Site Dresden, Dresden, Germany.,Medical Department-Hematology and Cell Therapy, Medical Oncology, Hemostaseology, University of Leipzig Medical Center, Leipzig, Germany
| | | | | | - Michael Lübbert
- Department of Medicine I (Hematology, Oncology and Stem Cell Transplantation), Medical Center - University of Freiburg, Freiburg, Germany. .,German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Partner Site Freiburg, Freiburg, Germany. .,Faculty of Medicine, University of Freiburg, Freiburg, Germany.
| |
Collapse
|
7
|
Kishtagari A, Levine RL. The Role of Somatic Mutations in Acute Myeloid Leukemia Pathogenesis. Cold Spring Harb Perspect Med 2021; 11:cshperspect.a034975. [PMID: 32398288 DOI: 10.1101/cshperspect.a034975] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Acute myeloid leukemia (AML) is characterized by attenuation of lineage differentiation trajectories that results in impaired hematopoiesis and enhanced self-renewal. To date, sequencing studies have provided a rich landscape of information on the somatic mutations that contribute to AML pathogenesis. These studies show that most AML genomes harbor relatively fewer mutations, which are acquired in a stepwise manner. Our understanding of the genetic basis of leukemogenesis informs a broader understanding of what initiates and maintains the AML clone and informs the development of prognostic models and mechanism-based therapeutic strategies. Here, we explore the current knowledge of genetic and epigenetic aberrations in AML pathogenesis and how recent studies are expanding our knowledge of leukemogenesis and using this to accelerate therapeutic development for AML patients.
Collapse
Affiliation(s)
- Ashwin Kishtagari
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, Ohio 44195, USA.,Department of Hematology and Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, Ohio 44195, USA
| | - Ross L Levine
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA.,Department of Medicine, Leukemia Service, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA.,Center for Epigenetics Research, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA.,Center for Hematologic Malignancies, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA.,Molecular Cancer Medicine Service, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
| |
Collapse
|
8
|
Zhu ME, Wang Q, Zhou S, Wang B, Ke L, He P. Recombinant interleukin-2 stimulates lymphocyte recovery in patients with severe COVID-19. Exp Ther Med 2021; 21:227. [PMID: 33603836 PMCID: PMC7851620 DOI: 10.3892/etm.2021.9658] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Accepted: 12/15/2020] [Indexed: 12/15/2022] Open
Abstract
A recently identified type of pneumonia, referred to as coronavirus disease 2019 (COVID-19), which is caused by severe acute respiratory syndrome coronavirus-2, has rapidly spread worldwide. Lymphopenia and a proinflammatory cytokine storm frequently occur in patients with severe COVID-19. However, to the best of our knowledge, no specific immunomodulatory therapy for COVID-19 has been reported to date. In the present retrospective case-control study, the potential therapeutic effect of recombinant human interleukin-2 (rIL-2) in patients with severe COVID-19 was demonstrated. A total of 59 patients with severe COVID-19 were admitted to the Union Hospital of Tongji Medical College (Wuhan, China) between 29th January 2020 and 29th February 2020 and were included in the present study. In total, 20 patients received subcutaneous injection of rIL-2 (1 million IU per day) for 7-10 days in addition to regular treatment and were classified as the rIL-2 group. Furthermore, 20 of the 39 patients receiving regular treatment, without the intervention of rIL-2, were matched as the control group. Patients in these two groups were subjected to propensity score matching in terms of clinical characteristics such as age, sex, symptoms, signs, laboratory data and comorbidities. Changes in the lymphocyte count, as well as IL-6 and C-reactive protein (CRP) levels, were analyzed at the time of admission and discharge and any differences between the rIL-2 and non-rIL-2 groups were determined. The results demonstrated an increase in the lymphocyte count and a decrease in CRP levels in the rIL-2 group compared with that in the non-rIL-2 group. The difference in the change of the lymphocyte count between the rIL-2 group and non-rIL-2 group was statistically significant (P<0.01). Although CRP levels were decreased to a greater extent in the rIL-2 group, the difference between the two groups was not statistically significant (P>0.05). Collectively, the present results suggested that administration of rIL-2 may be a prospective adjuvant therapy for patients with severe COVID-19 and its effects may be mediated by increasing lymphocyte numbers.
Collapse
Affiliation(s)
- Meng-En Zhu
- Department of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Qian Wang
- Department of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Shaoqiong Zhou
- Department of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Bin Wang
- Department of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Li Ke
- Department of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Ping He
- Department of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| |
Collapse
|
9
|
Sundaravel S, Steidl U, Wickrema A. Epigenetic modifiers in normal and aberrent erythropoeisis. Semin Hematol 2021; 58:15-26. [PMID: 33509439 PMCID: PMC7883935 DOI: 10.1053/j.seminhematol.2020.12.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 12/18/2020] [Accepted: 12/22/2020] [Indexed: 12/17/2022]
Abstract
Erythroid differentiation program is comprised of lineage commitment, erythroid progenitor proliferation, and termination differentiation. Each stage of the differentiation program is heavily influenced by epigenetic modifiers that alter the epigenome in a dynamic fashion influenced by cytokines/humeral factors and are amicable to target by drugs. The epigenetic modifiers can be classified as DNA modifiers (DNMT, TET), mRNA modifiers (RNA methylases and demethylases) and histone protein modifiers (methyltransferases, acetyltransferases, demethylases, and deacetylases). Here we describe mechanisms by which these epigenetic modifiers influence and guide erythroid-lineage differentiation during normal and malignant erythropoiesis and also benign diseases that arise from their altered structure or function.
Collapse
Affiliation(s)
- Sriram Sundaravel
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY
| | - Ulrich Steidl
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY; Department of Medicine, Albert Einstein College of Medicine-Montefiore Medical center, Bronx, NY
| | | |
Collapse
|
10
|
Greve G, Schüler J, Grüning BA, Berberich B, Stomper J, Zimmer D, Gutenkunst L, Bönisch U, Meier R, Blagitko-Dorfs N, Grishina O, Pfeifer D, Weichenhan D, Plass C, Lübbert M. Decitabine Induces Gene Derepression on Monosomic Chromosomes: In Vitro and In Vivo Effects in Adverse-Risk Cytogenetics AML. Cancer Res 2020; 81:834-846. [PMID: 33203699 DOI: 10.1158/0008-5472.can-20-1430] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 08/21/2020] [Accepted: 11/12/2020] [Indexed: 11/16/2022]
Abstract
Hypomethylating agents (HMA) have become the backbone of nonintensive acute myeloid leukemia/myelodysplastic syndrome (AML/MDS) treatment, also by virtue of their activity in patients with adverse genetics, for example, monosomal karyotypes, often with losses on chromosome 7, 5, or 17. No comparable activity is observed with cytarabine, a cytidine analogue without DNA-hypomethylating properties. As evidence exists for compounding hypermethylation and gene silencing of hemizygous tumor suppressor genes (TSG), we thus hypothesized that this effect may preferentially be reversed by the HMAs decitabine and azacitidine. An unbiased RNA-sequencing approach was developed to interrogate decitabine-induced transcriptome changes in AML cell lines with or without a deletion of chromosomes 7q, 5q or 17p. HMA treatment preferentially upregulated several hemizygous TSG in this genomic region, significantly derepressing endogenous retrovirus (ERV)3-1, with promoter demethylation, enhanced chromatin accessibility, and increased H3K4me3 levels. Decitabine globally reactivated multiple transposable elements, with activation of the dsRNA sensor RIG-I and interferon regulatory factor (IRF)7. Induction of ERV3-1 and RIG-I mRNA was also observed during decitabine treatment in vivo in serially sorted peripheral blood AML blasts. In patient-derived monosomal karyotype AML murine xenografts, decitabine treatment resulted in superior survival rates compared with cytarabine. Collectively, these data demonstrate preferential gene derepression and ERV reactivation in AML with chromosomal deletions, providing a mechanistic explanation that supports the clinical observation of superiority of HMA over cytarabine in this difficult-to-treat patient group. SIGNIFICANCE: These findings unravel the molecular mechanism underlying the intriguing clinical activity of HMAs in AML/MDS patients with chromosome 7 deletions and other monosomal karyotypes.See related commentary by O'Hagan et al., p. 813.
Collapse
Affiliation(s)
- Gabriele Greve
- Department of Medicine I, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Julia Schüler
- Charles River Discovery Research Services Germany GmbH, Freiburg, Germany
| | - Björn A Grüning
- Bioinformatics Group, Department of Computer Science, University of Freiburg, Freiburg, Germany
| | - Bettina Berberich
- Department of Medicine I, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Julia Stomper
- Department of Medicine I, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Dennis Zimmer
- Department of Medicine I, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Lea Gutenkunst
- Department of Medicine I, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Ulrike Bönisch
- Deep Sequencing Facility, Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
| | - Ruth Meier
- Department of Medicine I, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Clinic for Pediatric and Adolescent Medicine Klinikum Karlsruhe, Karlsruhe, Germany
| | - Nadja Blagitko-Dorfs
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency (CCI), Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Olga Grishina
- Clinical Trials Unit, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Dietmar Pfeifer
- Department of Medicine I, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Dieter Weichenhan
- Division of Cancer Epigenomics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Christoph Plass
- Division of Cancer Epigenomics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Michael Lübbert
- Department of Medicine I, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany. .,DKTK Partner Site Freiburg, German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
11
|
Monosomal karyotype and chromosome 17p loss or TP53 mutations in decitabine-treated patients with acute myeloid leukemia. Ann Hematol 2020; 99:1551-1560. [PMID: 32504186 PMCID: PMC7316846 DOI: 10.1007/s00277-020-04082-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Accepted: 05/12/2020] [Indexed: 11/22/2022]
Abstract
TP53 aberrations reportedly predict favorable responses to decitabine (DAC) in acute myeloid leukemia (AML). We evaluated clinical features and outcomes associated with chromosome 17p loss or TP53 gene mutations in older, unfit DAC-treated AML patients in a phase II trial. Of 178 patients, 25 had loss of 17p in metaphase cytogenetics; 24 of these had a complex (CK+) and 21 a monosomal karyotype (MK+). In analyses in all patients and restricted to CK+ and MK+ patients, 17p loss tended to associate with higher rates of complete remission (CR), partial remission (PR), or antileukemic effect (ALE). Despite favorable response rates, there was no significant OS difference between patients with or without loss of 17p in the entire cohort or in the CK+ and MK+ cohort. TP53 mutations were identified in eight of 45 patients with material available. Five of the eight TP53-mutated patients had 17p loss. TP53-mutated patients had similar rates of CR/PR/ALE but shorter OS than those with TP53 wild type (P = 0.036). Moreover, patients with a subclone based on mutation data had shorter OS than those without (P = 0.05); only one patient with TP53-mutated AML had a subclone. In conclusion, 17p loss conferred a favorable impact on response rates, even among CK+ and MK+ patients that however could not be maintained. The effect of TP53 mutations appeared to be different; however, patient numbers were low. Future research needs to further dissect the impact of the various TP53 aberrations in HMA-based combination therapies. The limited duration of favorable responses to HMA treatment in adverse-risk genetics AML should prompt physicians to advance allografting for eligible patients in a timely fashion.
Collapse
|
12
|
Kreutmair S, Erlacher M, Andrieux G, Istvanffy R, Mueller-Rudorf A, Zwick M, Rückert T, Pantic M, Poggio T, Shoumariyeh K, Mueller TA, Kawaguchi H, Follo M, Klingeberg C, Wlodarski M, Baumann I, Pfeifer D, Kulinski M, Rudelius M, Lemeer S, Kuster B, Dierks C, Peschel C, Cabezas-Wallscheid N, Duque-Afonso J, Zeiser R, Cleary ML, Schindler D, Schmitt-Graeff A, Boerries M, Niemeyer CM, Oostendorp RA, Duyster J, Illert AL. Loss of the Fanconi anemia-associated protein NIPA causes bone marrow failure. J Clin Invest 2020; 130:2827-2844. [PMID: 32338640 PMCID: PMC7260023 DOI: 10.1172/jci126215] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Accepted: 02/13/2020] [Indexed: 12/12/2022] Open
Abstract
Inherited bone marrow failure syndromes (IBMFSs) are a heterogeneous group of disorders characterized by defective hematopoiesis, impaired stem cell function, and cancer susceptibility. Diagnosis of IBMFS presents a major challenge due to the large variety of associated phenotypes, and novel, clinically relevant biomarkers are urgently needed. Our study identified nuclear interaction partner of ALK (NIPA) as an IBMFS gene, as it is significantly downregulated in a distinct subset of myelodysplastic syndrome-type (MDS-type) refractory cytopenia in children. Mechanistically, we showed that NIPA is major player in the Fanconi anemia (FA) pathway, which binds FANCD2 and regulates its nuclear abundance, making it essential for a functional DNA repair/FA/BRCA pathway. In a knockout mouse model, Nipa deficiency led to major cell-intrinsic defects, including a premature aging phenotype, with accumulation of DNA damage in hematopoietic stem cells (HSCs). Induction of replication stress triggered a reduction in and functional decline of murine HSCs, resulting in complete bone marrow failure and death of the knockout mice with 100% penetrance. Taken together, the results of our study add NIPA to the short list of FA-associated proteins, thereby highlighting its potential as a diagnostic marker and/or possible target in diseases characterized by hematopoietic failure.
Collapse
Affiliation(s)
- Stefanie Kreutmair
- Department of Internal Medicine I, Medical Center — University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Miriam Erlacher
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Pediatric Hematology and Oncology, Department of Pediatrics and Adolescent Medicine, and
| | - Geoffroy Andrieux
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Institute of Medical Bioinformatics and Systems Medicine, University Medical Center — University of Freiburg, Faculty of Medicine, Freiburg, Germany
| | - Rouzanna Istvanffy
- Department of Internal Medicine III, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Alina Mueller-Rudorf
- Department of Internal Medicine I, Medical Center — University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Melissa Zwick
- Department of Internal Medicine I, Medical Center — University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Tamina Rückert
- Department of Internal Medicine I, Medical Center — University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Milena Pantic
- Department of Internal Medicine I, Medical Center — University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Teresa Poggio
- Department of Internal Medicine I, Medical Center — University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Khalid Shoumariyeh
- Department of Internal Medicine I, Medical Center — University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Tony A. Mueller
- Department of Internal Medicine I, Medical Center — University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Hiroyuki Kawaguchi
- Department of Pediatrics, National Defense Medical College, Saitama, Japan
| | - Marie Follo
- Department of Internal Medicine I, Medical Center — University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Cathrin Klingeberg
- Department of Internal Medicine I, Medical Center — University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Marcin Wlodarski
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Pediatric Hematology and Oncology, Department of Pediatrics and Adolescent Medicine, and
| | - Irith Baumann
- Institute of Pathology, Health Center Böblingen, Böblingen, Germany
| | - Dietmar Pfeifer
- Department of Internal Medicine I, Medical Center — University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Michal Kulinski
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Martina Rudelius
- Institute of Pathology, Ludwig Maximilian University Munich, Munich, Germany
| | - Simone Lemeer
- Chair of Proteomics and Bioanalytics, Technical University of Munich, Freising, Germany
| | - Bernhard Kuster
- Chair of Proteomics and Bioanalytics, Technical University of Munich, Freising, Germany
| | - Christine Dierks
- Department of Internal Medicine I, Medical Center — University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Christian Peschel
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Internal Medicine III, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | | | - Jesus Duque-Afonso
- Department of Internal Medicine I, Medical Center — University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Robert Zeiser
- Department of Internal Medicine I, Medical Center — University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Michael L. Cleary
- Department of Pathology, Stanford University School of Medicine, Stanford, California, USA
| | - Detlev Schindler
- Department of Human Genetics, Institute of Human Genetics, Biozentrum, University of Würzburg, Würzburg, Germany
| | | | - Melanie Boerries
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Institute of Medical Bioinformatics and Systems Medicine, University Medical Center — University of Freiburg, Faculty of Medicine, Freiburg, Germany
| | - Charlotte M. Niemeyer
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Pediatric Hematology and Oncology, Department of Pediatrics and Adolescent Medicine, and
| | - Robert A.J. Oostendorp
- Department of Internal Medicine III, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Justus Duyster
- Department of Internal Medicine I, Medical Center — University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Anna Lena Illert
- Department of Internal Medicine I, Medical Center — University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
13
|
Crisà E, Kulasekararaj AG, Adema V, Such E, Schanz J, Haase D, Shirneshan K, Best S, Mian SA, Kizilors A, Cervera J, Lea N, Ferrero D, Germing U, Hildebrandt B, Martínez ABV, Santini V, Sanz GF, Solé F, Mufti GJ. Impact of somatic mutations in myelodysplastic patients with isolated partial or total loss of chromosome 7. Leukemia 2020; 34:2441-2450. [PMID: 32066866 DOI: 10.1038/s41375-020-0728-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Revised: 12/19/2019] [Accepted: 01/28/2020] [Indexed: 11/09/2022]
Abstract
Monosomy 7 [-7] and/or partial loss of chromosome 7 [del(7q)] are associated with poor and intermediate prognosis, respectively, in myelodysplastic syndromes (MDS), but somatic mutations may also play a key complementary role. We analyzed the impact on the outcomes of deep targeted mutational screening in 280 MDS patients with -7/del(7q) as isolated cytogenetic abnormality (86 with del(7q) and 194 with -7). Patients with del(7q) or -7 had similar demographic and disease-related characteristics. Somatic mutations were detected in 79% (93/117) of patients (82% in -7 and 73% in del(7q) group). Median number of mutations per patient was 2 (range 0-8). There was no difference in mutation frequency between the two groups. Patients harbouring ≥2 mutations had a worse outcome than patients with <2 or no mutations (leukaemic transformation at 24 months, 38% and 20%, respectively, p = 0.044). Untreated patients with del(7q) had better overall survival (OS) compared with -7 (median OS, 34 vs 17 months, p = 0.034). In multivariable analysis, blast count, TP53 mutations and number of mutations were independent predictors of OS, whereas the cytogenetic subgroups did not retain prognostic relevance. This study highlights the importance of mutational analysis in terms of prognosis in MDS patients with isolated -7 or del(7q).
Collapse
Affiliation(s)
- Elena Crisà
- Department of Haematological Medicine, King's College Hospital, NHS Foundation Trust, London, UK. .,Division of Hematology, Department of Translational Medicine, University of Eastern Piedmont, Novara, Italy. .,Fondazione Italiana Sindromi Mielodisplastiche (FISiM), Bologna, Italy.
| | - Austin G Kulasekararaj
- Department of Haematological Medicine, King's College Hospital, NHS Foundation Trust, London, UK
| | - Vera Adema
- Institut de Recerca Contra la Leucèmia Josep Carreras, ICO-Hospital Germans Trias i Pujol, Universitat Autònoma de Barcelona, Barcelona, Spain.,Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Esperanza Such
- Department of Hematology, Hospital Universitario La Fe, Valencia, Spain.,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto de Salud Carlos III, Madrid, Spain
| | - Julie Schanz
- Department of Hematology and Medical Oncology, University Medical Center of Göttingen, Göttingen, Germany
| | - Detlef Haase
- Department of Hematology and Medical Oncology, University Medical Center of Göttingen, Göttingen, Germany
| | - Katayoon Shirneshan
- Department of Hematology and Medical Oncology, University Medical Center of Göttingen, Göttingen, Germany
| | - Steven Best
- Laboratory for Molecular Haemato-Oncology, King's College Hospital, NHS Foundation Trust, London, UK
| | - Syed A Mian
- Department of Haematological Medicine, King's College Hospital, NHS Foundation Trust, London, UK.,Haematopoietic Stem Cell Laboratory, The Francis Crick Institute, London, UK
| | - Aytug Kizilors
- Laboratory for Molecular Haemato-Oncology, King's College Hospital, NHS Foundation Trust, London, UK
| | - José Cervera
- Genetics Unit, Hospital Universitario La Fe, Valencia, Spain
| | - Nicholas Lea
- Laboratory for Molecular Haemato-Oncology, King's College Hospital, NHS Foundation Trust, London, UK
| | - Dario Ferrero
- Fondazione Italiana Sindromi Mielodisplastiche (FISiM), Bologna, Italy.,Division of Hematology, University of Torino, AOU Città della Salute e della Scienza, Torino, Italy
| | - Ulrich Germing
- Department of Hematology, Oncology, and Clinical Immunology, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Barbara Hildebrandt
- Institute of Human Genetics, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | | | - Valeria Santini
- Fondazione Italiana Sindromi Mielodisplastiche (FISiM), Bologna, Italy.,MDS UNIT, AOU Careggi, University of Florence, Firenze, Italy
| | - Guillermo F Sanz
- Department of Hematology, Hospital Universitario La Fe, Valencia, Spain.,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto de Salud Carlos III, Madrid, Spain.,Department of Medicine, University of Valencia, Valencia, Spain
| | - Francesc Solé
- Institut de Recerca Contra la Leucèmia Josep Carreras, ICO-Hospital Germans Trias i Pujol, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Ghulam J Mufti
- Department of Haematological Medicine, King's College Hospital, NHS Foundation Trust, London, UK
| |
Collapse
|
14
|
Cunningham I, Hamele‐Bena D, Guo Y, Shiomi T, Papp AC, Chakravarti B, Yang J, Shyr Y, Fisher RA. Extramedullary leukemia behaving as solid cancer: clinical, histologic, and genetic clues to chemoresistance in organ sites. Am J Hematol 2019; 94:1200-1207. [PMID: 31353508 DOI: 10.1002/ajh.25594] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Revised: 07/03/2019] [Accepted: 07/22/2019] [Indexed: 01/14/2023]
Abstract
Recent studies of leukemic tumors in individual extramedullary sites showed they adopt the clinical and metastatic behavior of solid cancers originating in those sites. To elucidate features of leukemic tumors that render them resistant to agents effective against marrow leukemia, we analyzed a series of AML breast tumors by histology, immunohistochemistry, and RNA sequencing. Striking histologic similarities to solid cancers were found: a single-filing architectural pattern virtually identical to that of invasive lobular breast carcinoma and dense desmoplastic keloid-like fibrosis similar to colon, gallbladder, and pancreas carcinomas. Sequencing found 2157 genes significantly downregulated in AML breast tumors compared to normal breast. Comparison to triple-negative breast cancer found 859 genes similarly downregulated. At least 30 of these genes have been associated with poor prognosis in breast cancers. Five were reported in AML marrow studies to correlate with poor prognosis. The findings of this pilot study suggest the seed-and-soil interaction recognized in solid cancer growth may help explain how leukemic cells, in some patients, adopt solid tumor behavior in non-marrow sites. Transformed cells that metastasize from tumor to marrow can impart chemoresistance and be an unrecognized cause of treatment failure and death. Further studies comparing leukemic tumor to simultaneous marrow could potentially identify biomarkers that predict extramedullary resistance and lead to new therapeutic targets. Recognizing the potential for leukemia to adopt solid tumor phenotype, and implementation of body scanning and ablative tumor treatment, could decrease the persistently high rates of marrow resistance and treatment failure.
Collapse
Affiliation(s)
- Isabel Cunningham
- Division of Hematology OncologyColumbia University Vagelos College of Physicians and Surgeons New York New York
| | - Diane Hamele‐Bena
- Department of Pathology and Cell BiologyColumbia University Vagelos College of Physicians and Surgeons New York New York
| | - Yan Guo
- BioinformaticsUniversity of New Mexico Albuquerque New Mexico
| | - Takayuki Shiomi
- Department of PathologyInternational University of Health and Welfare Chiba Japan
| | - Audrey C. Papp
- Center for PharmacogenomicsOhio State University Columbus Ohio
| | | | - Jianqi Yang
- Department of PharmacologyUniversity of Iowa Iowa City Iowa
| | - Yu Shyr
- Department of BiostatisticsVanderbilt University Nashville Tennessee
| | - Rory A. Fisher
- Department of PharmacologyUniversity of Iowa Iowa City Iowa
| |
Collapse
|
15
|
Sundaravel S, Kuo WL, Jeong JJ, Choudhary GS, Gordon-Mitchell S, Liu H, Bhagat TD, McGraw KL, Gurbuxani S, List AF, Verma A, Wickrema A. Loss of Function of DOCK4 in Myelodysplastic Syndromes Stem Cells is Restored by Inhibitors of DOCK4 Signaling Networks. Clin Cancer Res 2019; 25:5638-5649. [PMID: 31308061 DOI: 10.1158/1078-0432.ccr-19-0924] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 06/13/2019] [Accepted: 07/10/2019] [Indexed: 12/17/2022]
Abstract
PURPOSE Myelodysplastic syndromes (MDS) with deletion of chromosome 7q/7 [-7/(del)7q MDS] is associated with worse outcomes and needs novel insights into pathogenesis. Reduced expression of signaling protein dedicator of cytokinesis 4 (DOCK4) in patients with -7/(del)7q MDS leads to a block in hematopoietic stem cell (HSC) differentiation. Identification of targetable signaling networks downstream of DOCK4 will provide means to restore hematopoietic differentiation in MDS.Experimental Design: We utilized phosphoproteomics approaches to identify signaling proteins perturbed as a result of reduced expression of DOCK4 in human HSCs and tested their functional significance in primary model systems. RESULTS We demonstrate that reduced levels of DOCK4 lead to increased global tyrosine phosphorylation of proteins in primary human HSCs. LYN kinase and phosphatases INPP5D (SHIP1) and PTPN6 (SHP1) displayed greatest levels of tyrosine phosphorylation when DOCK4 expression levels were reduced using DOCK4-specific siRNA. Our data also found that increased phosphorylation of SHIP1 and SHP1 phosphatases were due to LYN kinase targeting these phosphatases as substrates. Increased migration and impediment of HSC differentiation were consequences of these signaling alterations. Pharmacologic inhibition of SHP1 reversed these functional aberrations in HSCs expressing low DOCK4 levels. In addition, differentiation block seen in DOCK4 haplo-insufficient [-7/(del)7q] MDS was rescued by inhibition of SHP1 phosphatase. CONCLUSIONS LYN kinase and phosphatases SHP1 and SHIP1 are perturbed when DOCK4 expression levels are low. Inhibition of SHP1 promotes erythroid differentiation in healthy HSCs and in -7/(del)7q MDS samples with low DOCK4 expression. Inhibitors of LYN, SHP1 and SHIP1 also abrogated increased migratory properties in HSCs expressing reduced levels of DOCK4.
Collapse
Affiliation(s)
- Sriram Sundaravel
- Section of Hematology/Oncology, Department of Medicine, The University of Chicago, Chicago, Illinois
| | - Wen-Liang Kuo
- Section of Hematology/Oncology, Department of Medicine, The University of Chicago, Chicago, Illinois
| | - Jong Jin Jeong
- Section of Hematology/Oncology, Department of Medicine, The University of Chicago, Chicago, Illinois
| | - Gaurav S Choudhary
- Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, New York
| | | | - Hui Liu
- Section of Hematology/Oncology, Department of Medicine, The University of Chicago, Chicago, Illinois
| | - Tushar D Bhagat
- Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, New York
| | | | - Sandeep Gurbuxani
- Department of Pathology, The University of Chicago, Chicago, Illinois
| | | | - Amit Verma
- Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, New York
| | - Amittha Wickrema
- Section of Hematology/Oncology, Department of Medicine, The University of Chicago, Chicago, Illinois.
| |
Collapse
|
16
|
El Fakih R, Komrokji R, Shaheen M, Almohareb F, Rasheed W, Hassanein M. Azacitidine Use for Myeloid Neoplasms. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2018; 18:e147-e155. [DOI: 10.1016/j.clml.2018.02.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Revised: 01/23/2018] [Accepted: 02/02/2018] [Indexed: 01/08/2023]
|
17
|
Abstract
PURPOSE OF REVIEW Adverse karyotype acute myeloid leukemia is a disease particularly of older patients, but also observed in younger patients. Despite all efforts, standard chemotherapy is still generally applied in fit patients, as already for decades, and for nearly all different subtypes of acute myeloid leukemia. Lack of more specifically targeted therapy and the often older age of the patients are complicating treatment, and in the subgroup of patients achieving a complete remission, the strikingly high frequency of relapse is a characteristic of this disease. This review aims to give an overview of current treatment approaches as well as emerging therapies. RECENT FINDINGS Currently, the approach of a targeted therapy specific to the genetic and/or epigenetic aberrations detected in the individual patient is still not possible, and a 'one treatment fits all' course of action is still used, with allografting as curative consolidation. However, first immunotherapeutic approaches are emerging as treatment options and first phase 1 and 2 studies are described. SUMMARY Treatment of acute myeloid leukemia with adverse karyotype is still not individualized, most treatment options currently not being curative. This can change in the near future, but recent findings will have to be implemented into larger phase 3 studies before being standard of care.
Collapse
|
18
|
Single-cell RNA-seq reveals a distinct transcriptome signature of aneuploid hematopoietic cells. Blood 2017; 130:2762-2773. [PMID: 29030335 DOI: 10.1182/blood-2017-08-803353] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Accepted: 10/03/2017] [Indexed: 12/30/2022] Open
Abstract
Cancer cells frequently exhibit chromosomal abnormalities. Specific cytogenetic aberrations often are predictors of outcome, especially in hematologic neoplasms, such as monosomy 7 in myeloid malignancies. The functional consequences of aneuploidy at the cellular level are difficult to assess because of a lack of convenient markers to distinguish abnormal from diploid cells. We performed single-cell RNA sequencing (scRNA-seq) to study hematopoietic stem and progenitor cells from the bone marrow of 4 healthy donors and 5 patients with bone marrow failure and chromosome gain or loss. In total, transcriptome sequences were obtained from 391 control cells and 588 cells from patients. We characterized normal hematopoiesis as binary differentiation from stem cells to erythroid and myeloid-lymphoid pathways. Aneuploid cells were distinguished from diploid cells in patient samples by computational analyses of read fractions and gene expression of individual chromosomes. We confirmed assignment of aneuploidy to individual cells quantitatively, by copy-number variation, and qualitatively, by loss of heterozygosity. When we projected patients' single cells onto the map of normal hematopoiesis, diverse patterns were observed, broadly reflecting clinical phenotypes. Patients' monosomy 7 cells showed downregulation of genes involved in immune response and DNA damage checkpoint and apoptosis pathways, which may contribute to the clonal expansion of monosomy 7 cells with accumulated gene mutations. scRNA-seq is a powerful technique through which to infer the functional consequences of chromosome gain and loss and explore gene targets for directed therapy.
Collapse
|
19
|
Bhagat TD, Chen S, Bartenstein M, Barlowe AT, Von Ahrens D, Choudhary GS, Tivnan P, Amin E, Marcondes AM, Sanders MA, Hoogenboezem RM, Kambhampati S, Ramachandra N, Mantzaris I, Sukrithan V, Laurence R, Lopez R, Bhagat P, Giricz O, Sohal D, Wickrema A, Yeung C, Gritsman K, Aplan P, Hochedlinger K, Yu Y, Pradhan K, Zhang J, Greally JM, Mukherjee S, Pellagatti A, Boultwood J, Will B, Steidl U, Raaijmakers MHGP, Deeg HJ, Kharas MG, Verma A. Epigenetically Aberrant Stroma in MDS Propagates Disease via Wnt/β-Catenin Activation. Cancer Res 2017; 77:4846-4857. [PMID: 28684528 DOI: 10.1158/0008-5472.can-17-0282] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Revised: 06/06/2017] [Accepted: 06/30/2017] [Indexed: 11/16/2022]
Abstract
The bone marrow microenvironment influences malignant hematopoiesis, but how it promotes leukemogenesis has not been elucidated. In addition, the role of the bone marrow stroma in regulating clinical responses to DNA methyltransferase inhibitors (DNMTi) is also poorly understood. In this study, we conducted a DNA methylome analysis of bone marrow-derived stromal cells from myelodysplastic syndrome (MDS) patients and observed widespread aberrant cytosine hypermethylation occurring preferentially outside CpG islands. Stroma derived from 5-azacytidine-treated patients lacked aberrant methylation and DNMTi treatment of primary MDS stroma enhanced its ability to support erythroid differentiation. An integrative expression analysis revealed that the WNT pathway antagonist FRZB was aberrantly hypermethylated and underexpressed in MDS stroma. This result was confirmed in an independent set of sorted, primary MDS-derived mesenchymal cells. We documented a WNT/β-catenin activation signature in CD34+ cells from advanced cases of MDS, where it associated with adverse prognosis. Constitutive activation of β-catenin in hematopoietic cells yielded lethal myeloid disease in a NUP98-HOXD13 mouse model of MDS, confirming its role in disease progression. Our results define novel epigenetic changes in the bone marrow microenvironment, which lead to β-catenin activation and disease progression of MDS. Cancer Res; 77(18); 4846-57. ©2017 AACR.
Collapse
Affiliation(s)
- Tushar D Bhagat
- Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, New York
| | - Si Chen
- Erasmus Medical Center Cancer Institute, Department of Hematology and Erasmus Stem Cell Institute, Rotterdam, the Netherlands
| | | | | | - Dagny Von Ahrens
- Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, New York
| | - Gaurav S Choudhary
- Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, New York
| | - Patrick Tivnan
- Memorial Sloan Kettering Cancer Center, New York, New York
| | - Elianna Amin
- Memorial Sloan Kettering Cancer Center, New York, New York
| | - A Mario Marcondes
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington.,Division of Oncology, University of Washington, Seattle, Washington
| | - Mathijs A Sanders
- Erasmus Medical Center Cancer Institute, Department of Hematology and Erasmus Stem Cell Institute, Rotterdam, the Netherlands
| | - Remco M Hoogenboezem
- Erasmus Medical Center Cancer Institute, Department of Hematology and Erasmus Stem Cell Institute, Rotterdam, the Netherlands
| | | | - Nandini Ramachandra
- Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, New York
| | - Iaonnis Mantzaris
- Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, New York
| | - Vineeth Sukrithan
- Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, New York
| | - Remi Laurence
- Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, New York
| | - Robert Lopez
- Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, New York
| | - Prafullla Bhagat
- Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, New York
| | - Orsi Giricz
- Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, New York
| | - Davendra Sohal
- Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, New York
| | | | - Cecilia Yeung
- Memorial Sloan Kettering Cancer Center, New York, New York
| | - Kira Gritsman
- Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, New York
| | - Peter Aplan
- National Institutes of Health, Bethesda, Maryland
| | | | - Yiting Yu
- Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, New York
| | - Kith Pradhan
- Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, New York
| | - Jinghang Zhang
- Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, New York
| | - John M Greally
- Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, New York
| | | | - Andrea Pellagatti
- Bloodwise Molecular Haematology Unit, NDCLS, NIHR Biomedical Research Centre, Oxford University Hospitals, Oxford, United Kingdom
| | - Jacqueline Boultwood
- Bloodwise Molecular Haematology Unit, NDCLS, NIHR Biomedical Research Centre, Oxford University Hospitals, Oxford, United Kingdom
| | - Britta Will
- Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, New York
| | - Ulrich Steidl
- Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, New York
| | - Marc H G P Raaijmakers
- Erasmus Medical Center Cancer Institute, Department of Hematology and Erasmus Stem Cell Institute, Rotterdam, the Netherlands.
| | - H Joachim Deeg
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington. .,Division of Oncology, University of Washington, Seattle, Washington
| | | | - Amit Verma
- Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, New York.
| |
Collapse
|
20
|
Ross PJ, Canovas S. Mechanisms of epigenetic remodelling during preimplantation development. Reprod Fertil Dev 2017; 28:25-40. [PMID: 27062872 DOI: 10.1071/rd15365] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Epigenetics involves mechanisms independent of modifications in the DNA sequence that result in changes in gene expression and are maintained through cell divisions. Because all cells in the organism contain the same genetic blueprint, epigenetics allows for cells to assume different phenotypes and maintain them upon cell replication. As such, during the life cycle, there are moments in which the epigenetic information needs to be reset for the initiation of a new organism. In mammals, the resetting of epigenetic marks occurs at two different moments, which both happen to be during gestation, and include primordial germ cells (PGCs) and early preimplantation embryos. Because epigenetic information is reversible and sensitive to environmental changes, it is probably no coincidence that both these extensive periods of epigenetic remodelling happen in the female reproductive tract, under a finely controlled maternal environment. It is becoming evident that perturbations during the extensive epigenetic remodelling in PGCs and embryos can lead to permanent and inheritable changes to the epigenome that can result in long-term changes to the offspring derived from them, as indicated by the Developmental Origins of Health and Disease (DOHaD) hypothesis and recent demonstration of inter- and trans-generational epigenetic alterations. In this context, an understanding of the mechanisms of epigenetic remodelling during early embryo development is important to assess the potential for gametic epigenetic mutations to contribute to the offspring and for new epimutations to be established during embryo manipulations that could affect a large number of cells in the offspring. It is of particular interest to understand whether and how epigenetic information can be passed on from the gametes to the embryo or offspring, and whether abnormalities in this process could lead to transgenerationally inheritable phenotypes. The aim of this review is to highlight recent progress made in understanding the nature and mechanisms of epigenetic remodelling that ensue after fertilisation.
Collapse
Affiliation(s)
- Pablo Juan Ross
- Department of Animal Science, University of California, Davis, CA 95616 USA
| | - Sebastian Canovas
- LARCEL (Laboratorio Andaluz de Reprogramación Celular), BIONAND, Centro Andaluz de Nanomedicina y Biotecnología Campanillas, Malaga 29590, Spain
| |
Collapse
|
21
|
Eisfeld AK, Kohlschmidt J, Mrózek K, Volinia S, Blachly JS, Nicolet D, Oakes C, Kroll K, Orwick S, Carroll AJ, Stone RM, Byrd JC, de la Chapelle A, Bloomfield CD. Mutational Landscape and Gene Expression Patterns in Adult Acute Myeloid Leukemias with Monosomy 7 as a Sole Abnormality. Cancer Res 2016; 77:207-218. [PMID: 27784745 DOI: 10.1158/0008-5472.can-16-1386] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Revised: 09/16/2016] [Accepted: 09/21/2016] [Indexed: 12/31/2022]
Abstract
Monosomy of chromosome 7 is the most frequent autosomal monosomy in acute myeloid leukemia (AML), where it associates with poor clinical outcomes. However, molecular features associated with this sole monosomy subtype (-7 AML), which may give insights into the basis for its poor prognosis, have not been characterized. In this study, we analyzed 36 cases of -7 AML for mutations in 81 leukemia/cancer-associated genes using a customized targeted next-generation sequencing panel (Miseq). Global gene and miRNA expression profiles were also determined using paired RNA and small RNA sequencing data. Notably, gene mutations were detected in all the major AML-associated functional groups, which include activated signaling, chromatin remodeling, cohesin complex, methylation, NPM1, spliceosome, transcription factors, and tumor suppressors. Gene mutations in the chromatin remodeling groups were relatively more frequent in patients <60 years of age, who also had less mutations in the methylation and spliceosome groups compared with patients ≥60 years of age. Novel recurrent mutational events in AML were identified in the SMARCA2 gene. In patients ≥60 years of age, the presence of spliceosome mutations associated with a lower complete remission rate (P = 0.03). RNA sequencing revealed distinct gene and miRNA expression patterns between the sole -7 and non -7 AML cases, with reduced expression, as expected, of many genes and miRNAs mapped to chromosome 7, and overexpression of ID1, MECOM, and PTPRM, among others. Overall, our findings illuminate a number of molecular features of the underlying aggressive pathobiology in -7 AML patients. Cancer Res; 77(1); 207-18. ©2016 AACR.
Collapse
Affiliation(s)
| | - Jessica Kohlschmidt
- The Ohio State University Comprehensive Cancer Center, Columbus, Ohio.,Alliance Statistics and Data Center, Mayo Clinic, Rochester, Minnesota
| | - Krzysztof Mrózek
- The Ohio State University Comprehensive Cancer Center, Columbus, Ohio
| | - Stefano Volinia
- Department of Morphology, Surgery, and Experimental Medicine, University of Ferrara, Ferrara, Italy
| | - James S Blachly
- Division of Hematology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, Ohio
| | - Deedra Nicolet
- The Ohio State University Comprehensive Cancer Center, Columbus, Ohio.,Alliance Statistics and Data Center, Mayo Clinic, Rochester, Minnesota
| | - Christopher Oakes
- Division of Hematology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, Ohio
| | - Karl Kroll
- Division of Hematology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, Ohio
| | - Shelley Orwick
- Division of Hematology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, Ohio
| | | | | | - John C Byrd
- Division of Hematology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, Ohio
| | | | | |
Collapse
|
22
|
Patel SJ, Dao S, Darie CC, Clarkson BD. Defective quorum sensing of acute lymphoblastic leukemic cells: evidence of collective behavior of leukemic populations as semi-autonomous aberrant ecosystems. Am J Cancer Res 2016; 6:1177-230. [PMID: 27429840 PMCID: PMC4937729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Accepted: 04/04/2016] [Indexed: 06/06/2023] Open
Abstract
Quorum sensing (QS) is a generic term used to describe cell-cell communication and collective decision making by bacterial and social insects to regulate the expression of specific genes in controlling cell density and other properties of the populations in response to nutrient supply or changes in the environment. QS mechanisms also have a role in higher organisms in maintaining homeostasis, regulation of the immune system and collective behavior of cancer cell populations. In the present study, we used a p190(BCR-ABL) driven pre-B acute lymphoblastic leukemia (ALL3) cell line derived from the pleural fluid of a terminally ill patient with ALL to test the QS hypothesis in leukemia. ALL3 cells don't grow at low density (LD) in liquid media but grow progressively faster at increasingly high cell densities (HD) in contrast to other established leukemic cell lines that grow well at very low starting cell densities. The ALL3 cells at LD are poised to grow but shortly die without additional stimulation. Supernates of ALL3 cells (HDSN) and some other primary cells grown at HD stimulate the growth of the LD ALL3 cells without which they won't survive. To get further insight into the activation processes we performed microarray analysis of the LD ALL3 cells after stimulation with ALL3 HDSN at days 1, 3, and 6. This screen identified several candidate genes, and we linked them to signaling networks and their functions. We observed that genes involved in lipid, cholesterol, fatty acid metabolism, and B cell activation are most up- or down-regulated upon stimulation of the LD ALL3 cells using HDSN. We also discuss other pathways that are differentially expressed upon stimulation of the LD ALL3 cells. Our findings suggest that the Ph+ ALL population achieves dominance by functioning as a collective aberrant ecosystem subject to defective quorum-sensing regulatory mechanisms.
Collapse
Affiliation(s)
- Sapan J Patel
- Memorial Sloan Kettering Cancer Center, Molecular Pharmacology Program1275 York Avenue, Box #96, New York, NY 10065, USA
- Department of Chemistry and Biomolecular Science, Biochemistry and Proteomics Group, Clarkson University8 Clarkson Avenue, Potsdam, NY 13699-5810, USA
| | - Su Dao
- Memorial Sloan Kettering Cancer Center, Molecular Pharmacology Program1275 York Avenue, Box #96, New York, NY 10065, USA
| | - Costel C Darie
- Department of Chemistry and Biomolecular Science, Biochemistry and Proteomics Group, Clarkson University8 Clarkson Avenue, Potsdam, NY 13699-5810, USA
| | - Bayard D Clarkson
- Memorial Sloan Kettering Cancer Center, Molecular Pharmacology Program1275 York Avenue, Box #96, New York, NY 10065, USA
| |
Collapse
|
23
|
Li MY, Xu YY, Kang HY, Wang XR, Gao L, Cen J, Wang W, Wang N, Li YH, Wang LL, Yu L. Quantitative Detection of ID4 Gene Aberrant Methylation in the Differentiation of Myelodysplastic Syndrome from Aplastic Anemia. Chin Med J (Engl) 2016; 128:2019-25. [PMID: 26228212 PMCID: PMC4717959 DOI: 10.4103/0366-6999.161351] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
BACKGROUND The diagnosis of myelodysplastic syndrome (MDS), especially hypoplastic MDS, and MDS with low blast counts or normal karyotype may be problematic. This study characterized ID4 gene methylation in patients with MDS and aplastic anemia (AA). METHODS The methylation status of ID4 was analyzed by bisulfite sequencing polymerase chain reaction (PCR) and quantitative real-time methylation-specific PCR (MethyLight PCR) in 100 patients with MDS and 31 patients with AA. RESULTS The MDS group had a higher ID4 gene methylation positivity rate (22.22%) and higher methylation levels (0.21 [0-3.79]) than the AA group (P < 0.05). Furthermore, there were significant differences between the hypoplastic MDS and AA groups, the MDS with low blast count and the AA groups, and the MDS with normal karyotype and the AA groups. The combination of genetic and epigenetic markers was used in much more patients with MDS (62.5% [35/56]) than the use of genetic markers only (51.79% [29/56]). CONCLUSIONS These results showed that the detection of ID4 methylation positivity rates and levels could be a useful biomarker for MDS diagnosis.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Li Yu
- Department of Hematology, Chinese PLA General Hospital, Beijing 100853, China
| |
Collapse
|
24
|
H2AX deficiency is associated with erythroid dysplasia and compromised haematopoietic stem cell function. Sci Rep 2016; 6:19589. [PMID: 26791933 PMCID: PMC4726203 DOI: 10.1038/srep19589] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Accepted: 11/10/2015] [Indexed: 12/22/2022] Open
Abstract
Myelodysplastic syndromes (MDS) are clonal disorders of haematopoiesis characterised by dysplastic changes of major myeloid cell lines. However, the mechanisms underlying these dysplastic changes are poorly understood. Here, we used a genetically modified mouse model and human patient data to examine the physiological roles of H2AX in haematopoiesis and how the loss of H2AX contributes to dyserythropoiesis in MDS. H2AX knockout mice showed cell-autonomous anaemia and erythroid dysplasia, mimicking dyserythropoiesis in MDS. Also, dyserythropoiesis was increased in MDS patients with the deletion of chromosome 11q23, where H2AX is located. Although loss of H2AX did not affect the early stage of terminal erythropoiesis, enucleation was decreased. H2AX deficiency also led to the loss of quiescence of hematopoietic stem and progenitor cells, which dramatically compromised their bone marrow engraftment. These results reveal important roles of H2AX in late-stage terminal erythropoiesis and hematopoietic stem cell function.
Collapse
|
25
|
Reduced DOCK4 expression leads to erythroid dysplasia in myelodysplastic syndromes. Proc Natl Acad Sci U S A 2015; 112:E6359-68. [PMID: 26578796 DOI: 10.1073/pnas.1516394112] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Anemia is the predominant clinical manifestation of myelodysplastic syndromes (MDS). Loss or deletion of chromosome 7 is commonly seen in MDS and leads to a poor prognosis. However, the identity of functionally relevant, dysplasia-causing, genes on 7q remains unclear. Dedicator of cytokinesis 4 (DOCK4) is a GTPase exchange factor, and its gene maps to the commonly deleted 7q region. We demonstrate that DOCK4 is underexpressed in MDS bone marrow samples and that the reduced expression is associated with decreased overall survival in patients. We show that depletion of DOCK4 levels leads to erythroid cells with dysplastic morphology both in vivo and in vitro. We established a novel single-cell assay to quantify disrupted F-actin filament network in erythroblasts and demonstrate that reduced expression of DOCK4 leads to disruption of the actin filaments, resulting in erythroid dysplasia that phenocopies the red blood cell (RBC) defects seen in samples from MDS patients. Reexpression of DOCK4 in -7q MDS patient erythroblasts resulted in significant erythropoietic improvements. Mechanisms underlying F-actin disruption revealed that DOCK4 knockdown reduces ras-related C3 botulinum toxin substrate 1 (RAC1) GTPase activation, leading to increased phosphorylation of the actin-stabilizing protein ADDUCIN in MDS samples. These data identify DOCK4 as a putative 7q gene whose reduced expression can lead to erythroid dysplasia.
Collapse
|
26
|
Wong JC, Weinfurtner KM, Alzamora MDP, Kogan SC, Burgess MR, Zhang Y, Nakitandwe J, Ma J, Cheng J, Chen SC, Ho TT, Flach J, Reynaud D, Passegué E, Downing JR, Shannon K. Functional evidence implicating chromosome 7q22 haploinsufficiency in myelodysplastic syndrome pathogenesis. eLife 2015; 4. [PMID: 26193121 PMCID: PMC4569895 DOI: 10.7554/elife.07839] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Accepted: 07/17/2015] [Indexed: 11/13/2022] Open
Abstract
Chromosome 7 deletions are highly prevalent in myelodysplastic syndrome (MDS) and likely contribute to aberrant growth through haploinsufficiency. We generated mice with a heterozygous germ line deletion of a 2-Mb interval of chromosome band 5A3 syntenic to a commonly deleted segment of human 7q22 and show that mutant hematopoietic cells exhibit cardinal features of MDS. Specifically, the long-term hematopoietic stem cell (HSC) compartment is expanded in 5A3(+/del) mice, and the distribution of myeloid progenitors is altered. 5A3(+/del) HSCs are defective for lymphoid repopulating potential and show a myeloid lineage output bias. These cell autonomous abnormalities are exacerbated by physiologic aging and upon serial transplantation. The 5A3 deletion partially rescues defective repopulation in Gata2 mutant mice. 5A3(+/del) hematopoietic cells exhibit decreased expression of oxidative phosphorylation genes, increased levels of reactive oxygen species, and perturbed oxygen consumption. These studies provide the first functional data linking 7q22 deletions to MDS pathogenesis.
Collapse
Affiliation(s)
- Jasmine C Wong
- Department of Pediatrics, University of California, San Francisco, San Francisco, United States
| | - Kelley M Weinfurtner
- Department of Pediatrics, University of California, San Francisco, San Francisco, United States
| | | | - Scott C Kogan
- Department of Laboratory Medicine, University of California, San Francisco, San Francisco, United States
| | - Michael R Burgess
- Division of Hematology/Oncology, University of California, San Francisco, San Francisco, United States
| | - Yan Zhang
- Unit of Hematopoietic Stem Cell and Transgenic Animal Models, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China
| | - Joy Nakitandwe
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, United States
| | - Jing Ma
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, United States
| | - Jinjun Cheng
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, United States
| | - Shann-Ching Chen
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, United States
| | - Theodore T Ho
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, Department of Medicine, University of California, San Francisco, San Francisco, United States
| | - Johanna Flach
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, Department of Medicine, University of California, San Francisco, San Francisco, United States
| | - Damien Reynaud
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, Department of Medicine, University of California, San Francisco, San Francisco, United States
| | - Emmanuelle Passegué
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, Department of Medicine, University of California, San Francisco, San Francisco, United States
| | - James R Downing
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, United States
| | - Kevin Shannon
- Department of Pediatrics, University of California, San Francisco, San Francisco, United States
| |
Collapse
|
27
|
McGraw KL, Basiorka AA, Johnson JO, Clark J, Caceres G, Padron E, Heaton R, Ozawa Y, Wei S, Sokol L, List AF. Lenalidomide induces lipid raft assembly to enhance erythropoietin receptor signaling in myelodysplastic syndrome progenitors. PLoS One 2014; 9:e114249. [PMID: 25469886 PMCID: PMC4254997 DOI: 10.1371/journal.pone.0114249] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Accepted: 11/05/2014] [Indexed: 02/06/2023] Open
Abstract
Anemia remains the principal management challenge for patients with lower risk Myelodysplastic Syndromes (MDS). Despite appropriate cytokine production and cellular receptor display, erythropoietin receptor (EpoR) signaling is impaired. We reported that EpoR signaling is dependent upon receptor localization within lipid raft microdomains, and that disruption of raft integrity abolishes signaling capacity. Here, we show that MDS erythroid progenitors display markedly diminished raft assembly and smaller raft aggregates compared to normal controls (p = 0.005, raft number; p = 0.023, raft size). Because lenalidomide triggers raft coalescence in T-lymphocytes promoting immune synapse formation, we assessed effects of lenalidomide on raft assembly in MDS erythroid precursors and UT7 cells. Lenalidomide treatment rapidly induced lipid raft formation accompanied by EpoR recruitment into raft fractions together with STAT5, JAK2, and Lyn kinase. The JAK2 phosphatase, CD45, a key negative regulator of EpoR signaling, was displaced from raft fractions. Lenalidomide treatment prior to Epo stimulation enhanced both JAK2 and STAT5 phosphorylation in UT7 and primary MDS erythroid progenitors, accompanied by increased STAT5 DNA binding in UT7 cells, and increased erythroid colony forming capacity in both UT7 and primary cells. Raft induction was associated with F-actin polymerization, which was blocked by Rho kinase inhibition. These data indicate that deficient raft integrity impairs EpoR signaling, and provides a novel strategy to enhance EpoR signal fidelity in non-del(5q) MDS.
Collapse
Affiliation(s)
- Kathy L. McGraw
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center, 12902 Magnolia Drive, Tampa, FL 33612, United States of America
| | - Ashley A. Basiorka
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center-Cancer Biology Ph.D. Program, University of South Florida, 12902 Magnolia Drive, Tampa, FL, 33612, United States of America
| | - Joseph O. Johnson
- Analytic Microscopy Core Facility, H. Lee Moffitt Cancer Center, 12902 Magnolia Drive, Tampa, FL, 33612, United States of America
| | - Justine Clark
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center, 12902 Magnolia Drive, Tampa, FL 33612, United States of America
| | - Gisela Caceres
- Morsani Molecular Diagnostic Laboratory, H. Lee Moffitt Cancer Center, 10902 N. McKinley Drive, Tampa, FL, 33612, United States of America
| | - Eric Padron
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center, 12902 Magnolia Drive, Tampa, FL 33612, United States of America
| | - Ruth Heaton
- Department of Pathology, University of Arizona, 1501 N Campbell Ave, Tucson, AZ, 85724, United States of America
| | - Yukiyasu Ozawa
- Department of Hematology, Japanese Red Cross Nagoya First Hospital, 3-35 Michishita-cho, Nakamura-ku, Aichi, 453-8511, Japan
| | - Sheng Wei
- Department of Immunology, H. Lee Moffitt Cancer Center, 12902 Magnolia Drive Tampa, FL, 33612, United States of America
| | - Lubomir Sokol
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center, 12902 Magnolia Drive, Tampa, FL 33612, United States of America
| | - Alan F. List
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center, 12902 Magnolia Drive, Tampa, FL 33612, United States of America
- * E-mail:
| |
Collapse
|
28
|
Ko YA, Mohtat D, Suzuki M, Park ASD, Izquierdo MC, Han SY, Kang HM, Si H, Hostetter T, Pullman JM, Fazzari M, Verma A, Zheng D, Greally JM, Susztak K. Cytosine methylation changes in enhancer regions of core pro-fibrotic genes characterize kidney fibrosis development. Genome Biol 2014; 14:R108. [PMID: 24098934 PMCID: PMC4053753 DOI: 10.1186/gb-2013-14-10-r108] [Citation(s) in RCA: 182] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2013] [Accepted: 09/24/2013] [Indexed: 01/15/2023] Open
Abstract
Background One in eleven people is affected by chronic kidney disease, a condition characterized by kidney fibrosis and progressive loss of kidney function. Epidemiological studies indicate that adverse intrauterine and postnatal environments have a long-lasting role in chronic kidney disease development. Epigenetic information represents a plausible carrier for mediating this programming effect. Here we demonstrate that genome-wide cytosine methylation patterns of healthy and chronic kidney disease tubule samples obtained from patients show significant differences. Results We identify differentially methylated regions and validate these in a large replication dataset. The differentially methylated regions are rarely observed on promoters, but mostly overlap with putative enhancer regions, and they are enriched in consensus binding sequences for important renal transcription factors. This indicates their importance in gene expression regulation. A core set of genes that are known to be related to kidney fibrosis, including genes encoding collagens, show cytosine methylation changes correlating with downstream transcript levels. Conclusions Our report raises the possibility that epigenetic dysregulation plays a role in chronic kidney disease development via influencing core pro-fibrotic pathways and can aid the development of novel biomarkers and future therapeutics.
Collapse
|
29
|
Namekata K, Kimura A, Kawamura K, Harada C, Harada T. Dock GEFs and their therapeutic potential: neuroprotection and axon regeneration. Prog Retin Eye Res 2014; 43:1-16. [PMID: 25016980 DOI: 10.1016/j.preteyeres.2014.06.005] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2014] [Revised: 06/26/2014] [Accepted: 06/30/2014] [Indexed: 12/17/2022]
Abstract
The dedicator of cytokinesis (Dock) family is composed of atypical guanine exchange factors (GEFs) that activate the Rho GTPases Rac1 and Cdc42. Rho GTPases are best documented for their roles in actin polymerization and they regulate important cellular functions, including morphogenesis, migration, neuronal development, and cell division and adhesion. To date, 11 Dock family members have been identified and their roles have been reported in diverse contexts. There has been increasing interest in elucidating the roles of Dock proteins in recent years and studies have revealed that they are potential therapeutic targets for various diseases, including glaucoma, Alzheimer's disease, cancer, attention deficit hyperactivity disorder and combined immunodeficiency. Among the Dock proteins, Dock3 is predominantly expressed in the central nervous system and recent studies have revealed that Dock3 plays a role in protecting retinal ganglion cells from neurotoxicity and oxidative stress as well as in promoting optic nerve regeneration. In this review, we discuss the current understanding of the 11 Dock GEFs and their therapeutic potential, with a particular focus on Dock3 as a novel target for the treatment of glaucoma and other neurodegenerative diseases.
Collapse
Affiliation(s)
- Kazuhiko Namekata
- Visual Research Project, Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya-ku, Tokyo 156-8506, Japan
| | - Atsuko Kimura
- Visual Research Project, Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya-ku, Tokyo 156-8506, Japan
| | - Kazuto Kawamura
- Visual Research Project, Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya-ku, Tokyo 156-8506, Japan
| | - Chikako Harada
- Visual Research Project, Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya-ku, Tokyo 156-8506, Japan
| | - Takayuki Harada
- Visual Research Project, Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya-ku, Tokyo 156-8506, Japan.
| |
Collapse
|
30
|
Honda H, Nagamachi A, Inaba T. -7/7q- syndrome in myeloid-lineage hematopoietic malignancies: attempts to understand this complex disease entity. Oncogene 2014; 34:2413-25. [PMID: 24998854 DOI: 10.1038/onc.2014.196] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2014] [Revised: 05/27/2014] [Accepted: 06/03/2014] [Indexed: 01/19/2023]
Abstract
The recurrence of chromosomal abnormalities in a specific subtype of cancer strongly suggests that dysregulated gene expression in the corresponding region has a critical role in disease pathogenesis. -7/7q-, defined as the entire loss of chromosome 7 and partial deletion of its long arm, is among the most frequently observed chromosomal aberrations in myeloid-lineage hematopoietic malignancies such as myelodysplastic syndrome and acute myeloid leukemia, particularly in patients treated with cytotoxic agents and/or irradiation. Tremendous efforts have been made to clarify the molecular mechanisms underlying the disease development, and several possible candidate genes have been cloned. However, the study is still underway, and the entire nature of this syndrome is not completely understood. In this review, we focus on the attempts to identify commonly deleted regions in patients with -7/7q-; isolate the candidate genes responsible for disease development, cooperative genes and the factors affecting disease prognosis; and determine effective and potent therapeutic approaches. We also refer to the possibility that the accumulation of multiple gene haploinsufficiency, rather than the loss of a single tumor suppressor gene, may contribute to the development of diseases with large chromosomal deletions such as -7/7q-.
Collapse
Affiliation(s)
- H Honda
- Department of Disease Model, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
| | - A Nagamachi
- Department of Molecular Oncology and Leukemia Program Project, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
| | - T Inaba
- Department of Molecular Oncology and Leukemia Program Project, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
| |
Collapse
|
31
|
Gadea G, Blangy A. Dock-family exchange factors in cell migration and disease. Eur J Cell Biol 2014; 93:466-77. [PMID: 25022758 DOI: 10.1016/j.ejcb.2014.06.003] [Citation(s) in RCA: 164] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2014] [Revised: 06/10/2014] [Accepted: 06/17/2014] [Indexed: 02/06/2023] Open
Abstract
Dock family proteins are evolutionary conserved exchange factors for the Rho GTPases Rac and Cdc42. There are 11 Dock proteins in mammals, named Dock1 (or Dock180) to Dock11 that play different cellular functions. In particular, Dock proteins regulate actin cytoskeleton, cell adhesion and migration. Not surprisingly, members of the Dock family have been involved in various pathologies, including cancer and defects in the central nervous and immune systems. This review proposes an update of the recent findings regarding the function of Dock proteins, focusing on their role in the control of cell migration and invasion and the consequences in human diseases.
Collapse
Affiliation(s)
- Gilles Gadea
- CNRS UMR 5237, Centre de Recherche de Biochimie Macromoléculaire, France; Montpellier University, France
| | - Anne Blangy
- CNRS UMR 5237, Centre de Recherche de Biochimie Macromoléculaire, France; Montpellier University, France.
| |
Collapse
|
32
|
Bartholdy B, Christopeit M, Will B, Mo Y, Barreyro L, Yu Y, Bhagat TD, Okoye-Okafor UC, Todorova TI, Greally JM, Levine RL, Melnick A, Verma A, Steidl U. HSC commitment-associated epigenetic signature is prognostic in acute myeloid leukemia. J Clin Invest 2014; 124:1158-67. [PMID: 24487588 DOI: 10.1172/jci71264] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2013] [Accepted: 11/14/2013] [Indexed: 12/15/2022] Open
Abstract
Acute myeloid leukemia (AML) is characterized by disruption of HSC and progenitor cell differentiation. Frequently, AML is associated with mutations in genes encoding epigenetic modifiers. We hypothesized that analysis of alterations in DNA methylation patterns during healthy HSC commitment and differentiation would yield epigenetic signatures that could be used to identify stage-specific prognostic subgroups of AML. We performed a nano HpaII-tiny-fragment-enrichment-by-ligation-mediated-PCR (nanoHELP) assay to compare genome-wide cytosine methylation profiles between highly purified human long-term HSC, short-term HSC, common myeloid progenitors, and megakaryocyte-erythrocyte progenitors. We observed that the most striking epigenetic changes occurred during the commitment of short-term HSC to common myeloid progenitors and these alterations were predominantly characterized by loss of methylation. We developed a metric of the HSC commitment–associated methylation pattern that proved to be highly prognostic of overall survival in 3 independent large AML patient cohorts, regardless of patient treatment and epigenetic mutations. Application of the epigenetic signature metric for AML prognosis was superior to evaluation of commitment-based gene expression signatures. Together, our data define a stem cell commitment–associated methylome that is independently prognostic of poorer overall survival in AML.
Collapse
|
33
|
Santini V, Melnick A, Maciejewski JP, Duprez E, Nervi C, Cocco L, Ford KG, Mufti G. Epigenetics in focus: Pathogenesis of myelodysplastic syndromes and the role of hypomethylating agents. Crit Rev Oncol Hematol 2013; 88:231-45. [DOI: 10.1016/j.critrevonc.2013.06.004] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2013] [Revised: 05/14/2013] [Accepted: 06/12/2013] [Indexed: 12/22/2022] Open
|
34
|
Suzuki M, Greally JM. Genome-wide DNA methylation analysis using massively parallel sequencing technologies. Semin Hematol 2013; 50:70-7. [PMID: 23507485 DOI: 10.1053/j.seminhematol.2013.01.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
"Epigenetics" refers to a heritable change in transcriptional status without alteration in the primary nucleotide sequence. Epigenetics provides an extra layer of transcriptional control and plays a crucial role in normal development, as well as in pathological conditions. DNA methylation is one of the best known and well-studied epigenetic modifications. Genome-wide DNA methylation profiling has become recognized as a biologically and clinically important epigenomic assay. In this review, we discuss the strengths and weaknesses of the protocols for genome-wide DNA methylation profiling using massively parallel sequencing (MPS) techniques. We will also describe recently discovered DNA modifications, and the protocols to detect these modifications.
Collapse
Affiliation(s)
- Masako Suzuki
- Center for Epigenomics, Department of Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| | | |
Collapse
|
35
|
DOCK4 deletion at 7q31.1 in a de novo acute myeloid leukemia with a normal karyotype. Cell Oncol (Dordr) 2013; 36:395-403. [DOI: 10.1007/s13402-013-0145-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/06/2013] [Indexed: 10/26/2022] Open
|
36
|
Vasilatou D, Papageorgiou SG, Dimitriadis G, Pappa V. Epigenetic alterations and microRNAs: new players in the pathogenesis of myelodysplastic syndromes. Epigenetics 2013; 8:561-70. [PMID: 23760524 DOI: 10.4161/epi.24897] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The term epigenetics refers to the heritable changes in gene expression that do not represent changes in DNA sequence. DNA methylation and histone modification are the best studied epigenetic mechanisms. However, microRNAs, which affect gene expression at the posttranscriptional level, should be considered as members of the epigenetic machinery too. Myelodysplastic syndromes (MDS) are clone disorders of the hematopoietic stem cell with increased risk of leukemic transformation. Over the years, increased number of studies indicates the role of epigenetic mechanisms, including microRNAs, in MDS pathogenesis and prognosis. Indeed, epigenetic therapy with demethylating agents has already been applied to MDS. In this review we summarize current knowledge on the role of epigenetic alterations in MDS pathogenesis and treatment.
Collapse
Affiliation(s)
- Diamantina Vasilatou
- Second Department of Internal Medicine and Research Institute; Hematology Unit; Athens University Medical School; "Attikon" University General Hospital; Athens, Greece
| | | | | | | |
Collapse
|
37
|
Heuck CJ, Mehta J, Bhagat T, Gundabolu K, Yu Y, Khan S, Chrysofakis G, Schinke C, Tariman J, Vickrey E, Pulliam N, Nischal S, Zhou L, Bhattacharyya S, Meagher R, Hu C, Maqbool S, Suzuki M, Parekh S, Reu F, Steidl U, Greally J, Verma A, Singhal SB. Myeloma is characterized by stage-specific alterations in DNA methylation that occur early during myelomagenesis. THE JOURNAL OF IMMUNOLOGY 2013; 190:2966-75. [PMID: 23408834 DOI: 10.4049/jimmunol.1202493] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Epigenetic changes play important roles in carcinogenesis and influence initial steps in neoplastic transformation by altering genome stability and regulating gene expression. To characterize epigenomic changes during the transformation of normal plasma cells to myeloma, we modified the HpaII tiny fragment enrichment by ligation-mediated PCR assay to work with small numbers of purified primary marrow plasma cells. The nano-HpaII tiny fragment enrichment by ligation-mediated PCR assay was used to analyze the methylome of CD138(+) cells from 56 subjects representing premalignant (monoclonal gammopathy of uncertain significance), early, and advanced stages of myeloma, as well as healthy controls. Plasma cells from premalignant and early stages of myeloma were characterized by striking, widespread hypomethylation. Gene-specific hypermethylation was seen to occur in the advanced stages, and cell lines representative of relapsed cases were found to be sensitive to decitabine. Aberrant demethylation in monoclonal gammopathy of uncertain significance occurred primarily in CpG islands, whereas differentially methylated loci in cases of myeloma occurred predominantly outside of CpG islands and affected distinct sets of gene pathways, demonstrating qualitative epigenetic differences between premalignant and malignant stages. Examination of the methylation machinery revealed that the methyltransferase, DNMT3A, was aberrantly hypermethylated and underexpressed, but not mutated in myeloma. DNMT3A underexpression was also associated with adverse overall survival in a large cohort of patients, providing insights into genesis of hypomethylation in myeloma. These results demonstrate widespread, stage-specific epigenetic changes during myelomagenesis and suggest that early demethylation can be a potential contributor to genome instability seen in myeloma. We also identify DNMT3A expression as a novel prognostic biomarker and suggest that relapsed cases can be therapeutically targeted by hypomethylating agents.
Collapse
|
38
|
Pandolfi A, Barreyro L, Steidl U. Concise review: preleukemic stem cells: molecular biology and clinical implications of the precursors to leukemia stem cells. Stem Cells Transl Med 2013; 2:143-50. [PMID: 23349328 DOI: 10.5966/sctm.2012-0109] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Recent experimental evidence has shown that acute myeloid leukemia (AML) and myelodysplastic syndromes (MDS) arise from transformed immature hematopoietic cells following the accumulation of multiple stepwise genetic and epigenetic changes in hematopoietic stem cells and committed progenitors. The series of transforming events initially gives rise to preleukemic stem cells (pre-LSC), preceding the formation of fully transformed leukemia stem cells (LSC). Despite the established use of poly-chemotherapy, relapse continues to be the most common cause of death in AML and MDS. The therapeutic elimination of all LSC, as well as pre-LSC, which provide a silent reservoir for the re-formation of LSC, will be essential for achieving lasting cures. Conventional sequencing and next-generation genome sequencing have allowed us to describe many of the recurrent mutations in the bulk cell populations in AML and MDS, and recent work has also focused on identifying the initial molecular changes contributing to leukemogenesis. Here we review recent and ongoing advances in understanding the roles of pre-LSC, and the aberrations that lead to pre-LSC formation and subsequent LSC transformation.
Collapse
Affiliation(s)
- Ashley Pandolfi
- Department of Cell Biology, Albert Einstein College of Medicine, New York, New York, USA
| | | | | |
Collapse
|
39
|
Pakes NK, Veltman DM, Williams RSB. Zizimin and Dock guanine nucleotide exchange factors in cell function and disease. Small GTPases 2012; 4:22-7. [PMID: 23247359 PMCID: PMC3620097 DOI: 10.4161/sgtp.22087] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Zizimin proteins belong to the Dock (Dedicator of Cytokinesis) superfamily of Guanine nucleotide Exchange Factor (GEF) proteins. This family of proteins plays a role in the regulation of Rho family small GTPases. Together the Rho family of small GTPases and the Dock/Zizimin proteins play a vital role in a number of cell processes including cell migration, apoptosis, cell division and cell adhesion. Our recent studies of Zizimin proteins, using a simple biomedical model, the eukaryotic social amoeba Dictyostelium discoideum, have helped to elucidate the cellular role of these proteins. In this article, we discuss the domain structure of Zizimin proteins from an evolutionary viewpoint. We also compare what is currently known about the mammalian Zizimin proteins to that of related Dock proteins. Understanding the cellular functions of these proteins will provide a better insight into their role in cell signaling, and may help in treating disease pathology associated with mutations in Dock/Zizimin proteins.
Collapse
Affiliation(s)
- Nicholl K Pakes
- Centre for Biomedical Sciences, Royal Holloway University of London, Surrey, UK
| | | | | |
Collapse
|
40
|
Flohr H, Breull W. Effect of etafenone on total and regional myocardial blood flow. Semin Hematol 1976; 50:16-37. [PMID: 23 DOI: 10.1053/j.seminhematol.2013.01.001] [Citation(s) in RCA: 40] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The distribution of blood flow to the subendocardial, medium and subepicardial layers of the left ventricular free wall was studied in anaesthetized dogs under normoxic (A), hypoxic (B) conditions and under pharmacologically induced (etafenone) coronary vasodilation (C). Regional myocardial blood flow was determined by means of the particle distribution method. In normoxia a transmural gradient of flow was observed, with the subendocardial layers receiving a significantly higher flow rate compared with the subepicardial layers. In hypoxia induced vasodilation this transmural gradient of flow was persistent. In contrast a marked redistribution of regional flow was observed under pharmacologically induced vasodilation. The transmural gradient decreased. In contrast to some findings these experiments demonstrate that a considerable vasodilatory capacity exists in all layers of the myocardium and can be utilized by drugs. The differences observed for the intramural distribution pattern of flow under hypoxia and drug induced vasodilation support the hypothesis that this pattern reflects corresponding gradients of regional myocardial metabolism.
Collapse
|